Category: 8. Health

  • Graphene quantum dot-integrated nanocomposites could help fight brain tumors

    Scientists from India have studied the use of graphene quantum dot-integrated nanocomposites as a novel therapeutic strategy against glioblastoma, an aggressive and treatment-resistant type of brain tumor. 

    This innovative approach leverages the unique physicochemical properties of graphene quantum dots (GQDs) to enhance delivery, targeting, and efficacy of anti-cancer agents within the brain’s complex environment.

     

    Glioblastoma multiforme (GBM) remains one of the deadliest forms of brain cancer, characterized by rapid growth, diffuse infiltration into surrounding brain tissue, and resistance to conventional therapies such as surgery, radiotherapy, and chemotherapy. The integration of graphene quantum dots within nanocomposites, capitalizing on the exceptional attributes of graphene-based nanomaterials, could have potential for overcoming existing limitations in glioblastoma treatment.

    Graphene quantum dots are ultrafine, nanoscale fragments of graphene sheets exhibiting unique quantum confinement and edge effects. These properties endow GQDs with superior biocompatibility, tunable photoluminescence, remarkable surface area, and facile functionalization capabilities. When embedded into nanocomposites, these quantum dots enhance the platform’s capacity for drug loading, controlled release, and deep tissue penetration—critical parameters for effectively targeting GBM cells dispersed within the brain’s intricate architecture.

    The research explores the synthesis, characterization, and biological performance of these GQD-integrated nanocomposites. By engineering the nanocomposites to possess optimized size, surface chemistry, and charge, the team achieved improved blood-brain barrier (BBB) permeability—an obstacle that has historically hindered efficient drug delivery to brain tumors. Such advancements directly address a central challenge in neuro-oncology, whereby therapeutic agents often fail to reach adequate concentrations at the tumor site.

    Beyond enhanced delivery, graphene quantum dots impart additional therapeutic functionalities. Their intrinsic photoluminescence permits real-time imaging and tracking of the nanocomposites within biological systems, enabling precision in monitoring distribution and accumulation within glioblastoma tissues. Furthermore, GQDs exhibit photothermal properties, whereby exposure to near-infrared light can induce localized heating, triggering tumor cell apoptosis while sparing healthy brain cells—this multi-modal approach synergistically combines chemotherapy with photothermal therapy for potentiated anti-tumor activity.

    Critically, the cytotoxicity assays presented confirm that GQD-based nanocomposites maintain high biocompatibility with normal brain cells while exerting targeted cytotoxic effects against glioblastoma cell lines. This selectivity minimizes off-target damage, a major concern in brain cancer treatments, thus promising improved patient safety profiles. The ability to achieve such selective toxicity underscores the transformative potential of nanomanipulation strategies in precision oncology.

    The study elucidates cellular uptake pathways of these nanocomposites, demonstrating that their physicochemical modifications enable efficient endocytosis by GBM cells. Intracellular trafficking studies reveal that once internalized, the nanocomposites localize predominantly within lysosomes and the cytoplasm, facilitating the release of encapsulated anti-cancer drugs in a spatially controlled manner. This precise intracellular delivery enhances cytotoxic efficacy while mitigating systemic side effects.

    In vivo experimentation conducted on glioblastoma-bearing animal models corroborates the translational promise of this technology. Treated subjects exhibited significant tumor regression, prolonged survival time, and reduced neurologic deficits compared to control groups receiving standard chemotherapy alone. Imaging data further validated the ability of GQD-nanocomposites to accumulate selectively in tumor tissue, highlighting their targeting efficiency and real-time imaging capability.

    The modular nature of graphene quantum dot integration allows for facile customization of the nanocomposite surface with targeting ligands such as peptides, antibodies, or aptamers that recognize glioblastoma-specific biomarkers. Such functionalization not only improves selectivity but also addresses the heterogeneity inherent in GBM tumors, potentially mitigating resistance mechanisms that frequently lead to therapeutic failure.

    Intriguingly, the photostability and chemical robustness of graphene quantum dots impart durability to these nanoconstructs, ensuring sustained therapeutic effect and reproducibility across repeated treatment cycles. This contrasts with some organic nanoparticles susceptible to rapid degradation or aggregation, which impair clinical applicability. Consequently, GQD-integrated platforms may offer superior consistency in treatment outcomes.

    Although promising, several translational hurdles remain to be addressed before clinical application. Scalability of high-quality graphene quantum dots, long-term toxicity profiles, and comprehensive pharmacokinetics require extensive investigation. Moreover, the complex immunological landscape of the brain mandates rigorous assessment to preclude unintended inflammatory or immunosuppressive effects induced by the nanocomposites.

    Nonetheless, the multidisciplinary collaboration embodied in this research—from material science to oncology to neurobiology—exemplifies the innovative spirit necessary to tackle formidable challenges like glioblastoma. The convergence of nanotechnology and cancer therapy continues to pave a new paradigm that could fundamentally shift current clinical approaches and improve patient prognoses in one of the most challenging diseases.

    In conclusion, the development of graphene quantum dot-integrated nanocomposites offers a highly promising avenue toward more effective, precise, and multimodal glioblastoma treatment. By dramatically enhancing drug delivery across the blood-brain barrier, enabling real-time imaging, and synergistically combining chemotherapeutic and photothermal modalities, this technology stands poised to redefine the therapeutic landscape. 

    Continue Reading

  • Exploring how women with HIV develop hazardous drinking patterns: a qualitative assessment of drinking histories | BMC Public Health

    Exploring how women with HIV develop hazardous drinking patterns: a qualitative assessment of drinking histories | BMC Public Health

    We interviewed 20 WWH with a history of hazardous drinking patterns who completed a randomized clinical trial to reduce or quit their drinking. Among participants, the mean age was 49.3 (Standard Deviation [SD] = 6.9), 85% were Black women, 60% had less than a high school education, 60% were single, and 95% were unemployed. At the 7-month follow-up of the parent study, most women reported either a reduction in drinking (30%) or complete cessation of drinking (55%). Table 2 provides a detailed summary of participant demographics.

    Table 2 Sociodemographic profile and drinking patterns of the participants (N = 20)

    The qualitative analysis revealed three overarching domains characterizing the lifetime drinking trajectories of WWH. These domains include: (1) Onset of Alcohol Use, encompassing early alcohol exposure, the influence of parental drinking during adolescence, and the role of peer dynamics in shaping risk behaviors; (2) Escalation to Hazardous Drinking, which captures patterns of increasing alcohol consumption, shifts in drinking behavior, the use of alcohol as a coping mechanism, and the co-use of alcohol and other substances; and (3) Post-Study Drinking Continuation, which reflects ongoing alcohol use following the conclusion of the parent study, including alcohol’s perceived impact on mental health, its social functions, perceived health benefits, and factors contributing to successful reductions in drinking. Table 3 presents the thematic codes and their definitions. A detailed discussion of each domain and its prominent codes is provided below.

    Table 3 Themes and codes identified in qualitative analysis, with definitions

    Drinking onset

    When WWH talked about the circumstances surrounding the onset of their drinking, several themes emerged:

    Early alcohol exposure

    Many participants began the story of their drinking onset by recounting early exposures to alcohol during childhood or adolescence. These early encounters, occurring in both supervised and unsupervised settings, appeared to normalize alcohol consumption and contributed to earlier patterns of escalation. Most participants reported exposure to alcohol before the age of 18, with some recalling initial experiences as young as 10 years old. Notably, some participants differentiated between their first taste of alcohol and the onset of regular drinking, highlighting a developmental progression from experimentation to habitual use.

    “My first beer I had when I was 11 years old. I remember it was a Miller Lite. It was the little bottle, it was this big. They used to sell them here [name of state]. That was my first beer. But then I started drinking when I was 16.” (Age 45, Hispanic/Latino, Quit drinking).

    “Fourteen… Oh no, about 10 to 14–14 is when I really started escalating. From 10 to 14 I was just stealing drinks here and there. When my mom would leave an empty drink and I would go drink it, stuff like that.” (Age 50, non-Hispanic White, Quit drinking).

    The effect of parental drinking on adolescents

    Many participants highlighted the influential role of family in shaping their early alcohol use. Several described familial behaviors that normalized or facilitated drinking, such as collective family drinking or parents consuming alcohol openly in front of their children. In numerous accounts, participants recalled their parents not only modeling alcohol use but also actively encouraging them to drink, sometimes beginning in childhood or early adolescence. These experiences contributed to the perception that alcohol consumption was acceptable or even expected within the family environment.

    “My dad taught us that stuff. It was like an Italian family where you drink wine at the dinner table. That’s what we did. We drank wine. Sometimes my dad would give me more than one cup. So that’s how I started. And then we would always go to the club and other things where people were heavily drinking. Anywhere we went with my parents, it was like a cocktail party or something. That’s the way it was.” (Age 52, non-Hispanic White, Did not reduce or quit drinking).

    “My mother was a bad alcoholic. She passed away in 2006. She had quit drinking towards the end of her life and all that, but when us kids were growing up she’d even take us kids to the [name of club] and get us Shirley Temples when she drank.” (Age 50, non-Hispanic White, Quit drinking).

    Other participants described acquiring knowledge and behaviors related to alcohol use, including preferences for specific types of alcohol, strategies for obtaining it, and information regarding its perceived effects, such as health benefits.

    “When I was coming up, I would drink alcohol. My mama would give me alcohol, not thinking that I would become addicted to it. When I was a kid, it was good for worms. She’d send me to the store when she wasn’t able to go to the store. She’d tell me to go to the man on the corner and tell him to get her two beers. I learned how to lie, because when I got older, I went to the man and asked the man to give me four beers for me and my friends. When I got a certain age, I used my mama’s name, saying that she said to get her four beers, and since I did it for such a period of time, it was easy for the same guy to give me the alcohol. That’s when it took off.” (Age 58, non-Hispanic Black/African American, Quit drinking).

    The role of peer influences on development and risk

    In addition to familial influences, participants described a range of external social factors that contributed to the initiation of their alcohol use. Relationships with friends and romantic partners were frequently cited as key influences, with several participants specifically noting the role of peer pressure in prompting them to begin drinking. Others reported that the desire to fit in socially or to participate in group activities involving alcohol served as a primary motivation for their initial alcohol consumption.

    “My peers, you could say peer pressure. Everybody was doing it, so I’m going to do it, too, that kind of thing.” (Age 52, non-Hispanic Black/African American, Quit drinking).

    The escalation to hazardous drinking

    The transition from casual to hazardous drinking was often marked by either a gradual or abrupt escalation of alcohol use, polysubstance use, and the adoption of drinking as a coping mechanism.

    Escalating drinking patterns

    Many participants described a gradual or sudden escalation in their drinking. Some participants described beginning with occasional drinking that gradually escalated over time, while others reported initiating alcohol use with heavy consumption and maintaining high levels of frequency and volume throughout their lives. One participant noted a specific event, retirement, that changed their life and caused them to escalate their drinking, while others just described incremental increases over time.

    “Oh no, my experience is I used to not drink that much, and of course, I worked at the hospital, so I only drank on the weekends or I’d have only one beer. I drink beer mostly; once in a while, wine… Then when I retired, I got really bored and I guess depressed…So I started drinking every day, and then sometimes I would drink too much the night before, like six beers or something, and then in the morning, I’d need a beer badly, you know so that’s how that started.” (Age 52, non-Hispanic White, Did not reduce or quit drinking).

    Changing drinking patterns

    Some participants described having on-and-off patterns of drinking that changed during their lifetime, with specific or non-specific triggers. The on-and-off patterns described by participants varied in nature. For some, these shifts reflected transitions between periods of hazardous drinking and abstinence, often influenced by life events or changes in responsibilities. For others, the pattern involved moving between hazardous use and more moderate or controlled drinking.

    “So, I stopped drinking for 13 [years], and then something hit me, and I started to have a beer. Somebody before introduced me to wine and that struck it.” (Age 39, non-Hispanic Black/African American, Reduced drinking).

    Coping and mental health

    Many participants reported that their alcohol use intensified as a coping mechanism for managing negative emotions. This pattern of use was frequently associated with diagnosed mental health conditions or significant adverse life experiences, suggesting a link between psychological distress and the escalation of drinking behavior.

    “It made me forget the hurt. That’s where the numbness came in. I didn’t focus just on the relationship going sour. It’s like okay, it’s gone, but this drink is making me not even think about it. It’s a sense of being numb, but then covering up my true feelings of how I was feeling at that present time.” (Age 49, non-Hispanic Black/African American, Quit drinking).

    “My manic depression just got a hold of me and it wouldn’t let go. It was a terrible feeling. Now they’ve got my meds regulated and everything. That’s good.” (Age 50, non-Hispanic White, Quit drinking).

    In addition to using alcohol to numb negative emotions, some participants described experiencing positive emotional effects that contributed to their coping strategies. These included feelings of increased confidence, emotional release, and a temporary sense of relief from psychological distress. One participant described how the sense of empowerment they felt while drinking helped them cope with feelings of powerlessness and provided an escape from their circumstances:

    “The feeling, the false image of being something that I’m not. It allowed me to speak whatever came to my mind, no filter, say how I feel. If I didn’t drink most of the things that I said I probably wouldn’t have said it sober. [Alcohol] just made me feel like superhero. I know it’s false. I know it’s due to the alcohol because I’m not the same person that I am when I’m sober. I know it’s the alcohol.” (Age 36, non-Hispanic Black/African American, Reduced drinking).

    The combination of drinking and drug use

    A few participants described that, as their alcohol use increased, they began using other drugs. However, some participants said that they started using alcohol as a substitute for drugs or to aid them in overcoming their drug use disorders.

    “I got older, the drugs came because of the relationships I was in, trying to fit in and trying to be loved when I knew it was wrong. As I got in my twenties, I graduated to alcohol and liquor, and then I started doing drugs.” (Age 51, non-Hispanic Black/African American, Reduced drinking).

    “I stopped [drinking], hooked up with this guy for a long time, he broke my heart. I didn’t want to go back in the streets to do drugs like crack and pot and that, so I went to beer, drank 15, then to 20 cans.” (Age 52, non-Hispanic Black/African American, Quit drinking).

    “You know, I’ve dealt with crack, too, so you know the drinking was just like– you know, being an addict I have to have some way of relief. You know, I have to have it.” (Age 39, non-Hispanic Black/African American, Reduced drinking).

    Moreover, participants generally attributed their alcohol use to its greater accessibility compared to other substances, rather than to a belief that alcohol was inherently healthier or carried fewer negative consequences. This distinction was explicitly noted by one participant, who emphasized convenience over perceived safety:

    “Just because I know I can. It’s not like going to buy dope, like I’m going to get busted or anything, so it’s not scary. It’s not as scary to me going in a store and purchasing alcohol…but see, alcohol is not expensive. If you get $5, you can drink all day, compared to those shoes that you want for $50, and that cable that you got to keep on, it’s not a hard decision to make because it’s legal. It’s so easy to get that I think it’s more of how easy it is to get that makes people just drink it just to be drinking it.” (Age 36, non-Hispanic Black/African American, Reduced drinking).

    Drinking continuation

    Nine participants reported that they either did not reduce their alcohol consumption or continued to drink despite some reduction by the end of the study period. The reasons for continued alcohol use among WWH included emotional responses associated with drinking, its perceived impact on mental health, the social nature of alcohol consumption, and beliefs about its potential health benefits.

    Alcohol’s impact on mental health

    The most frequently cited reason for continued alcohol use following study participation was the emotional response associated with drinking. Many WWH talked about feeling positive, using alcohol to cope with daily stress or traumatic events, or using alcohol to relax. Some also spoke about enjoying alcohol or describing drinking and situations in which they would drink as being fun.

    “I guess it’s just the atmosphere you know it loosens you up it helps you relax sometimes, and relieves a lot of tension and stress from the workday.” (Age 57, non-Hispanic Black/African American, Quit drinking).

    The social aspect of drinking

    Some participants reported that they continued drinking due to the social benefits it provided. Alcohol consumption was described as a means of fostering connection with friends, family members, or romantic partners. For several individuals, drinking occurred exclusively in social contexts, highlighting the role of alcohol in facilitating interpersonal relationships and social engagement.

    “It’s like when you’re dealing with someone who drinks, and you want to be a part of them. You start socializing, and you find yourself drinking more often than the norm, than you would normally do. I found myself drinking.” (Age 49, non-Hispanic Black/African American, Quit drinking).

    Perceived health benefits associated with drinking alcohol

    Some participants said they continued to drink because they perceived health benefits from drinking. Some said they generally felt better or that drinking was part of their overall healthy routine, while others described specific situations or conditions for which they perceived benefits from drinking. However, these participants frequently describe other motivations overlapping with perceived health benefits. For instance, some participants reported not only deriving enjoyment from alcohol consumption but also perceiving functional benefits, such as improved sleep, enhanced relaxation, or relief from physical pain. This suggests that perceived health benefits may reinforce continued alcohol use and complicate efforts to reduce alcohol use, particularly when such beliefs are intertwined with emotional relief.

    “Usually, I have pains when I sleep; my body hurts. I don’t sleep very well. Uh, probably before the alcohol. I heard it was caused by the alcohol, so I don’t know, but I don’t like taking pills…I’m tired of pills. So it’s like that I could take a sleeping pill instead, but I don’t. I just like drinking. [Laughs] I’m going to be honest. I just like it.” (Age 52, non-Hispanic White, Did not reduce or quit drinking).

    Facilitators of successful drinking reduction

    Finally, some WWH described not why they continued drinking but why they reduced or stopped. Although all participants reported hazardous drinking at the time of enrollment, several indicated that they had reduced their alcohol consumption either by the time of the interview or at some point in the past. Participants attributed their behavior change to internal motivation and personal experiences. One of these facilitators of drinking reduction was only drinking occasionally or only in specific scenarios, such as in social situations.

    “It’s just I was an occasional drinker, like when I go to places, ah, when I’m hangin’ with certain people.” (Age 44, non-Hispanic Black/African American, Did not reduce or quit drinking).

    Others indicated that they had a negative emotional or physical experience that prompted them to reduce or stop drinking, or that they were not getting the positive feelings and outcomes that they expected from alcohol, which made them stop drinking.

    “Well, as far as experience, I don’t like the way it makes me feel anymore. I didn’t do things that I wasn’t supposed to while under the influence. I actually don’t drink as much as I used to for my own reasons. It wasn’t a good experience. It felt good when I did it, but the aftermath didn’t feel good at all because I had consequences behind it. That’s my experience.” (Age 36, non-Hispanic Black/African American, Reduced drinking).

    These accounts suggest that for some WWH, drinking reduction was facilitated not by external intervention or formal support but by a personal reassessment of alcohol’s role in their lives. This reliance on internal resources highlights the importance of self-directed pathways to change.

    Continue Reading

  • Zhang L, Wu Y, Nie Y, Lv W, Li Y, Zhu B, Jin S, Shen Z, Li F, Liu H, et al. The serum free triiodothyronine to free thyroxine ratio as a potential prognostic biomarker of chronic kidney disease in patients with glomerular crescents: a retrospective study. Front Endocrinol (Lausanne). 2022;13:977355.

    PubMed 

    Google Scholar 

  • Teumer A, Chaker L, Groeneweg S, Li Y, Di Munno C, Barbieri C, Schultheiss UT, Traglia M, Ahluwalia TS, Akiyama M, et al. Genome-wide analyses identify a role for SLC17A4 and AADAT in thyroid hormone regulation. Nat Commun. 2018;9(1):4455.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Alkemade A. Central and peripheral effects of thyroid hormone signalling in the control of energy metabolism. J Neuroendocrinol. 2010;22(1):56–63.

    PubMed 

    Google Scholar 

  • Bianco AC, Dumitrescu A, Gereben B, Ribeiro MO, Fonseca TL, Fernandes GW, Bocco B. Paradigms of dynamic control of thyroid hormone signaling. Endocr Rev. 2019;40(4):1000–47.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Laclaustra M, Moreno-Franco B, Lou-Bonafonte JM, Mateo-Gallego R, Casasnovas JA, Guallar-Castillon P, Cenarro A, Civeira F. Impaired sensitivity to thyroid hormones is associated with diabetes and metabolic syndrome. Diabetes Care. 2019;42(2):303–10.

    PubMed 

    Google Scholar 

  • Bian N, Sun X, Zhou B, Zhang L, Wang Q, An Y, Li X, Li Y, Liu J, Meng H, et al. Obese patients with higher TSH levels had an obvious metabolic improvement after bariatric surgery. Endocr Connect. 2021;10(10):1326–36.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Liu ZM, Li G, Wu Y, Zhang D, Zhang S, Hao YT, Chen W, Huang Q, Li S, Xie Y, et al. Increased central and peripheral thyroid resistance indices during the first half of gestation were associated with lowered risk of gestational diabetes-analyses based on huizhou birth cohort in south China. Front Endocrinol (Lausanne). 2022;13:806256.

    PubMed 

    Google Scholar 

  • Štěpánek L, Horáková D, Štěpánek L, Janout V, Janoutová J, Bouchalová K, Martiník K. Free triiodothyronine/free thyroxine (FT3/FT4) ratio is strongly associated with insulin resistance in euthyroid and hypothyroid adults: a cross-sectional study. Endokrynol Pol. 2021;72(1):8–13.

    PubMed 

    Google Scholar 

  • KDIGO. Clinical practice guideline for the management of blood pressure in chronic kidney disease. Kidney Int. 2021;99(3s):S1-s87.

    Google Scholar 

  • Kang HT, Linton JA, Kwon SK, Park BJ, Lee JH. Ferritin level is positively associated with chronic kidney disease in Korean men, based on the 2010–2012 Korean National Health and Nutrition Examination Survey. Int J Environ Res Public Health. 2016. https://doi.org/10.3390/ijerph13111058.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Rhee CM, Brent GA, Kovesdy CP, Soldin OP, Nguyen D, Budoff MJ, Brunelli SM, Kalantar-Zadeh K. Thyroid functional disease: an under-recognized cardiovascular risk factor in kidney disease patients. Nephrol Dial Transplant. 2015;30(5):724–37.

    PubMed 

    Google Scholar 

  • Lin CC, Chen TW, Ng YY, Chou YH, Yang WC. Thyroid dysfunction and nodular goiter in hemodialysis and peritoneal dialysis patients. Perit Dial Int. 1998;18(5):516–21.

    PubMed 

    Google Scholar 

  • Ellervik C, Mora S, Ridker PM, Chasman DI. Hypothyroidism and kidney function: a Mendelian randomization study. Thyroid. 2020;30(3):365–79.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Zhang Y, Chang Y, Ryu S, Cho J, Lee WY, Rhee EJ, Kwon MJ, Pastor-Barriuso R, Rampal S, Han WK, et al. Thyroid hormone levels and incident chronic kidney disease in euthyroid individuals: the Kangbuk Samsung health study. Int J Epidemiol. 2014;43(5):1624–32.

    PubMed 

    Google Scholar 

  • Gopinath B, Harris DC, Wall JR, Kifley A, Mitchell P. Relationship between thyroid dysfunction and chronic kidney disease in community-dwelling older adults. Maturitas. 2013;75(2):159–64.

    PubMed 

    Google Scholar 

  • Mehran L, Delbari N, Amouzegar A, Hasheminia M, Tohidi M, Azizi F. Reduced sensitivity to thyroid hormone is associated with diabetes and hypertension. J Clin Endocrinol Metab. 2022;107(1):167–76.

    PubMed 

    Google Scholar 

  • Sun H, Zhu W, Liu J, An Y, Wang Y, Wang G. Reduced sensitivity to thyroid hormones is associated with high remnant cholesterol levels in Chinese euthyroid adults. J Clin Endocrinol Metab. 2022;108(1):166–74.

    PubMed 

    Google Scholar 

  • Ellervik C, Roselli C, Christophersen IE, Alonso A, Pietzner M, Sitlani CM, Trompet S, Arking DE, Geelhoed B, Guo X, et al. Assessment of the relationship between genetic determinants of thyroid function and atrial fibrillation: a Mendelian randomization study. JAMA Cardiol. 2019;4(2):144–52.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Jostel A, Ryder WD, Shalet SM. The use of thyroid function tests in the diagnosis of hypopituitarism: definition and evaluation of the TSH index. Clin Endocrinol (Oxf). 2009;71(4):529–34.

    PubMed 

    Google Scholar 

  • Yagi H, Pohlenz J, Hayashi Y, Sakurai A, Refetoff S. Resistance to thyroid hormone caused by two mutant thyroid hormone receptors beta, R243Q and R243W, with marked impairment of function that cannot be explained by altered in vitro 3,5,3’-triiodothyroinine binding affinity. J Clin Endocrinol Metab. 1997;82(5):1608–14.

    PubMed 

    Google Scholar 

  • Liu C, Hua L, Liu K, Xin Z. Impaired sensitivity to thyroid hormone correlates to osteoporosis and fractures in euthyroid individuals. J Endocrinol Invest. 2023. https://doi.org/10.1007/s40618-023-02035-1.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Zipf G, Chiappa M, Porter KS, Ostchega Y, Lewis BG, Dostal J. National health and nutrition examination survey: plan and operations, 1999-2010. Vital Health Stat 1. 2013;56:1–37.

    Google Scholar 

  • Levey AS, Stevens LA, Schmid CH, Zhang YL, Castro AF 3rd, Feldman HI, Kusek JW, Eggers P, Van Lente F, Greene T, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604–12.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Qin Z, Li H, Wang L, Geng J, Yang Q, Su B, Liao R. Systemic immune-inflammation index is associated with increased urinary albumin excretion: a population-based study. Front Immunol. 2022;13:863640.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Guo X, Wang H, Song Q, Li N, Liang Q, Su W, Liang M, Ding X, Sun C, Lowe S, et al. Association between exposure to organophosphorus pesticides and the risk of diabetes among US adults: cross-sectional findings from the national health and nutrition examination survey. Chemosphere. 2022;301:134471.

    PubMed 

    Google Scholar 

  • Bernhardt PW. Model validation and influence diagnostics for regression models with missing covariates. Stat Med. 2018;37(8):1325–42.

    PubMed 

    Google Scholar 

  • Iglesias P, Díez JJ. Thyroid dysfunction and kidney disease. Eur J Endocrinol. 2009;160(4):503–15.

    PubMed 

    Google Scholar 

  • Yang S, Lai S, Wang Z, Liu A, Wang W, Guan H. Thyroid feedback quantile-based index correlates strongly to renal function in euthyroid individuals. Ann Med. 2021;53(1):1945–55.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Anderson JLC, Gruppen EG, van Tienhoven-Wind L, Eisenga MF, de Vries H, Gansevoort RT, Bakker SJL, Dullaart RPF. Glomerular filtration rate is associated with free triiodothyronine in euthyroid subjects: comparison between various equations to estimate renal function and creatinine clearance. Eur J Intern Med. 2018;48:94–9.

    PubMed 

    Google Scholar 

  • Shin DH, Lee MJ, Kim SJ, Oh HJ, Kim HR, Han JH, Koo HM, Doh FM, Park JT, Han SH, et al. Preservation of renal function by thyroid hormone replacement therapy in chronic kidney disease patients with subclinical hypothyroidism. J Clin Endocrinol Metab. 2012;97(8):2732–40.

    PubMed 

    Google Scholar 

  • Katz AI, Lindheimer MD. Renal sodium- and potassium-activated adenosine triphosphatase and sodium reabsorption in the hypothyroid rat. J Clin Invest. 1973;52(4):796–804.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Kim SH, Min HK, Lee SW. Relationship between thyroid and kidney function: analysis from the Korea national health and nutrition examination survey between 2013 and 2015. Kidney Blood Press Res. 2020;45(3):442–54.

    PubMed 

    Google Scholar 

  • Wang K, Xie K, Gu L, Xu B, Chen J, Lou Q, Yu Y, Shan S, Wu D, Dai L, et al. Association of thyroid function with the estimated glomerular filtration rate in a large Chinese euthyroid population. Kidney Blood Press Res. 2018;43(4):1075–83.

    PubMed 

    Google Scholar 

  • Peixoto de Miranda É JF, Bittencourt MS, Goulart AC, Santos IS, de Oliveira Titan SM, Ladeira RM, Barreto SM, Lotufo PA, Benseñor IJM: Thyrotropin levels are associated with chronic kidney disease among healthy subjects in cross-sectional analysis of the Brazilian Longitudinal Study of Adult Health (ELSA-Brasil). Clin Exp Nephrol 2017, 21(6):1035–1043.

  • Das G, Taylor PN, Abusahmin H, Ali A, Tennant BP, Geen J, Okosieme O. Relationship between serum thyrotropin and urine albumin excretion in euthyroid subjects with diabetes. Ann Clin Biochem. 2019;56(1):155–62.

    PubMed 

    Google Scholar 

  • Schairer B, Jungreithmayr V, Schuster M, Reiter T, Herkner H, Gessl A, Sengölge G, Winnicki W. Effect of thyroid hormones on kidney function in patients after kidney transplantation. Sci Rep. 2020;10(1):2156.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Asvold BO, Bjøro T, Vatten LJ. Association of thyroid function with estimated glomerular filtration rate in a population-based study: the HUNT study. Eur J Endocrinol. 2011;164(1):101–5.

    PubMed 

    Google Scholar 

  • Sun MT, Hsiao FC, Su SC, Pei D, Hung YJ. Thyrotropin as an independent factor of renal function and chronic kidney disease in normoglycemic euthyroid adults. Endocr Res. 2012;37(3):110–6.

    PubMed 

    Google Scholar 

  • Kar PM, Hirani A, Allen MJ. Acute renal failure in a hypothyroid patient with rhabdomyolysis. Clin Nephrol. 2003;60(6):428–9.

    PubMed 

    Google Scholar 

  • Sekine N, Yamamoto M, Michikawa M, Enomoto T, Hayashi M, Ozawa E, Kobayashi T. Rhabdomyolysis and acute renal failure in a patient with hypothyroidism. Intern Med. 1993;32(3):269–71.

    PubMed 

    Google Scholar 

  • Kaptein EM. Thyroid hormone metabolism and thyroid diseases in chronic renal failure. Endocr Rev. 1996;17(1):45–63.

    PubMed 

    Google Scholar 

  • Singh PA, Bobby Z, Selvaraj N, Vinayagamoorthi R. An evaluation of thyroid hormone status and oxidative stress in undialyzed chronic renal failure patients. Indian J Physiol Pharmacol. 2006;50(3):279–84.

    PubMed 

    Google Scholar 

  • Pasqualetti G, Calsolaro V, Bernardini S, Linsalata G, Bigazzi R, Caraccio N, Monzani F. Degree of peripheral thyroxin deiodination, frailty, and long-term survival in hospitalized older patients. J Clin Endocrinol Metab. 2018;103(5):1867–76.

    PubMed 

    Google Scholar 

  • Yuan D, Zhang C, Jia S, Liu Y, Jiang L, Xu L, Zhang Y, Xu J, Xu B, Hui R, et al. Predictive value of free triiodothyronine (FT3) to free thyroxine (FT4) ratio in long-term outcomes of euthyroid patients with three-vessel coronary artery disease. Nutr Metab Cardiovasc Dis. 2021;31(2):579–86.

    PubMed 

    Google Scholar 

  • Yu T, Tian C, Song J, He D, Wu J, Wen Z, Sun Z, Sun Z. Value of the fT3/fT4 ratio and its combination with the GRACE risk score in predicting the prognosis in euthyroid patients with acute myocardial infarction undergoing percutaneous coronary intervention: a prospective cohort study. BMC Cardiovasc Disord. 2018;18(1):181.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Brozaitiene J, Mickuviene N, Podlipskyte A, Burkauskas J, Bunevicius R. Relationship and prognostic importance of thyroid hormone and N-terminal pro-B-Type natriuretic peptide for patients after acute coronary syndromes: a longitudinal observational study. BMC Cardiovasc Disord. 2016;16:45.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Li S, Zeng M. J-shaped association of free triiodothyronine to free thyroxine ratio with 5-year mortality among patients with chronic kidney disease: a prospective cohort study. Int Urol Nephrol. 2023;55(10):2567–78.

    PubMed 

    Google Scholar 

  • Kong SH, Kim JH, Park YJ, Lee JH, Hong AR, Shin CS, Cho NH. Low free T3 to free T4 ratio was associated with low muscle mass and impaired physical performance in community-dwelling aged population. Osteoporos Int. 2020;31(3):525–31.

    PubMed 

    Google Scholar 

  • Chen Y, Zhang W, Wang N, Wang Y, Wang C, Wan H, Lu Y. Thyroid parameters and kidney disorder in type 2 diabetes: results from the METAL study. J Diabetes Res. 2020;2020:4798947.

    PubMed 
    PubMed Central 

    Google Scholar 

Continue Reading

  • Advances in cGAS-STING Signaling in Fibrosis Diseases: Therapeutic Tar

    Advances in cGAS-STING Signaling in Fibrosis Diseases: Therapeutic Tar

    Introduction

    Fibrosis, or tissue scarring, refers to the irreversible progression of fibrogenesis, which occurs in multiple organs—such as the liver, lungs, kidneys, heart, skin, or central nervous system—and leads to their dysfunction.1,2 The dominant pathology of organ fibrosis is featured by increased fibrous connective tissue and decreased parenchymal cells in organs.3 Organ fibrosis is a major contributor to global morbidity and mortality, accounting for approximately 45% or more of deaths in developed societies.4 However, due to the unclear cellular and molecular mechanisms that regulate fibrosis, effective antifibrotic targets and therapeutic interventions remain scarce.

    Insights into the fibrotic tissue environment have deepened our understanding of the progression and resolution of fibrogenesis, in which immune cells, inflammasomes, inflammatory cytokines, and intracellular signaling pathways play pivotal roles.5–7 The innate immune system provides the first line of defense against tissue injury and plays a pivotal role in the initiation of fibrosis, while the adaptive immune system contributes to the chronicity and specificity of the fibrotic response—particularly in autoimmune-associated fibrosis. In essence, innate immunity initiates and orchestrates the fibrotic response, whereas adaptive immunity sustains and refines its progression.8–10 The interplay between the innate and adaptive immune systems is crucial in determining whether tissue injury resolves or progresses to chronic fibrosis. Therefore, understanding this immunological balance opens new avenues for immunomodulatory therapies that target fibrosis at various stages.

    The stimulator of interferon genes (STING), discovered by Glen N. Barber’s team in 2008, is a crucial mediator that engages in innate immunity, inflammation and infectious diseases.11,12 Cyclic GMP-AMP synthase (cGAS), positioned upstream of STING, detects pathogens and aberrant DNA, producing cyclic GMP-AMP (cGAMP) as a secondary messenger that activates STING. This activation leads to the phosphorylation of downstream molecules such as interferon regulatory factor 3 (IRF3), TANK-binding kinase 1 (TBK1), and nuclear factor kappa B (NF-κB).13 Activation of the cGAS–STING pathway triggers the production of type I interferons (IFN-I) and proinflammatory cytokines, such as interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α), thereby enhancing the immune response.13,14

    Beyond infectious diseases, an increasing number of studies have shown that cGAS–STING signaling is also involved in autoimmune, cancerous, fibrotic, and aging-related neurodegenerative diseases.15 For example, the excessive inflammation response induced by aberrant activation of cGAS-STING resulted in fibrotic pulmonary diseases.16 Moreover, a variety of natural extractives, such as licorice extract and naringenin, have demonstrated potential immunoregulatory effects by inhibiting the cGAS–STING pathway, thereby preventing liver inflammation and fibrogenesis.17,18 In addition, a non-canonical cGAS-STING signaling, cGAS-STING-PERK-eukaryotic initiation factor 2α (eIF2α) pathway, was shown to participate in lung and kidney fibrosis.19 Notably, cGAS–STING-mediated inflammation plays a critical role in organ fibrosis. Therefore, with the discovery of inhibitors targeting the cGAS–STING pathway, inflammation and fibrosis driven by this signaling cascade could be significantly ameliorated.

    This review is devoted to detailing the molecular and cellular mechanisms of cGAS–STING signaling across various organ fibrotic diseases. Furthermore, a comprehensive compilation of cGAS–STING-related antagonists applied in fibrotic diseases is presented. We also discuss relevant challenges and offer our perspectives on directions for future research.

    Overview of cGAS-STING Signaling

    The cGAS–STING pathway is a crucial mediator that regulates inflammation in response to infections, cellular stress, and tissue injury. Insights into the structure, molecular components, activation mechanisms, and crosstalk of cGAS–STING signaling are highly significant for the development of targeted therapeutics for human inflammatory diseases.

    Structural and Molecular Biology of the cGAS-STING Signaling

    The cGAS–STING signaling pathway is primarily composed of cGAS, cGAMP, and STING. Discovered by Zhijian J. Chen’s team in 2013, cGAS functions as both a sensor and receptor of DNA. It was initially thought to be a cytosolic protein that remains inactive under physiological conditions.20 In 2019, Jonathan C. Kagan et al first demonstrated that inactive cGAS is localized at the plasma membrane through the function of an N-terminal phosphoinositide-binding domain, which prevents unwanted activation by avoiding recognition of small amounts of DNA scattered in the cytoplasm.21

    Upon invasion by external pathogens or internal cellular damage, microbial and host-derived DNAs are often exposed. When extraneous or autologous DNAs appear outside the nucleus or specific organelles like mitochondria, cGAS catalytic activity is triggered through interaction with short double-stranded DNA (dsDNA).15 Once activated, cGAS catalyzes the synthesis of 2′3′-cGAMP from GTP and ATP.22 Subsequently, cGAMP, acting as a second messenger, activates STING by binding to its cyclic dinucleotide (CDN)-binding pocket, initiating a signaling cascade that leads to the production of type I interferons (IFN-I) and other immune mediators.23,24

    STING is a transmembrane protein localized to the endoplasmic reticulum (ER) that functions as a pattern recognition receptor (PRR) capable of detecting CDN.11,25,26 It consists of a cytosolic N-terminal domain, a four-segment transmembrane region, and a cytosolic C-terminal domain (CTD).27 STING typically exists as a dimer, with its CTD forming a V-shaped structure that binds cGAMP at the dimer interface through both direct and solvent-bridged hydrogen bonds.28,29 Upon cGAMP binding, STING undergoes a conformational change that triggers its translocation from the endoplasmic reticulum (ER) to the Golgi apparatus and leads to its accumulation in perinuclear vesicles.30–32 When STING translocates to the ER–Golgi intermediate compartment (ERGIC), it recruits TBK1, which phosphorylates both IRF3 and itself.33,34 Phosphorylated IRF3 then dimerizes and translocates to the nucleus, where it promotes the expression of IFN-I and interferon-stimulated genes (ISGs), orchestrating a robust antiviral response34 (Figure 1).

    Figure 1 Molecular mechanism of the cGAS-STING signaling. Double-stranded DNA (dsDNA)—from microbes, damaged tissues or dead cells, and injured mitochondria—present in the cytoplasm will be sensed by cyclic GMP-AMP synthase (cGAS). Then cGAS will catalyze ATP and GTP into the second messenger 2′,3′-cyclic GMP-AMP (cGAMP), which will further activate the stimulator of interferon genes (STING) that located at the endoplasmic reticulum in a conformational change of oligomerization. The activated STING will move to Golgi. At Golgi, STING will recruit and phosphorylate the TANK-binding kinase 1 (TBK1) and/or inhibitor of kappa B kinase (IKK), thereafter phosphorylating interferon regulatory factor 3 (IRF3) into dimerization and inhibitor of nuclear factor-kappa B (IκB) into degradation, resulting in IRF3 and nuclear factor-kappa B (NF-κB) activation and translocation into the nucleus to induce transcription of type I interferon (IFN-I) and interferon-stimulated genes (ISGs) as well as proinflammatory cytokines, such as IL-1β, IL-6, TNF-α.

    The above process is referred to as the canonical cGAS–STING signaling pathway. In other words, non-canonical cGAS–STING signaling pathways also exist. To date, several non-canonical pathways have been reported, including cGAS-STING/NF-κB, cGAS-STING/p38-MAPK, cGAS-STING-NLRP3, cGAS-STING-PERK-eIF2α, and cGAS-STING-induced LC3 lipidation-mediated autophagy.35–38 The upstream activation of NF-κB in the cGAS–STING/NF-κB pathway is similar to that of IRF3, but the downstream effects primarily lead to enhanced inflammation through the production of proinflammatory mediators such as IL-1β, IL-6, and TNF-α39 (Figure 1).

    Effects of cGAS-STING Signaling Activation

    The initiation of cGAS–STING signaling in mammalian cells is closely associated with antiviral activity, primarily through the downstream induction of IFN-I and ISGs.40 As our understanding of the molecular mechanisms and cellular roles of cGAS–STING signaling deepens, it has become clear that activation of this pathway leads to multiple effects. Beyond antiviral defense, the cGAS–STING pathway is also closely linked to cancer immunology and tumorigenesis.41 Pharmacological targeting of STING has shown promise in promoting antitumor immunity.42 Moreover, prior research has indicated that overactivation of the cGAS–STING pathway is associated with several autoimmune diseases, including rheumatoid arthritis, Aicardi-Goutières syndrome, and systemic lupus erythematosus.43 Although the cGAS–STING signaling pathway typically regulates immune surveillance by detecting viral, bacterial, and damaged self-DNAs, its overactivation can be detrimental.44 In the context of neuroinflammation, sustained activation of the cGAS–STING pathway contributes to neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis.45–48

    In terms of cellular functions, the cGAS–STING signaling pathway is involved in processes such as DNA repair, metabolism, cellular aging, cell death, and autophagy.15 The translocation of cGAS into the nucleus, which depends on importin-α and phosphorylation of cGAS at tyrosine 215, occurs in response to DNA damage.49 Thereafter, cGAS associates with double-stranded DNA breaks and interacts with poly (ADP-ribose) polymerase 1 (PARP1), impeding the formation of the PARP1–Timeless complex and thereby suppressing DNA repair.49 Additionally, cGAMP can inhibit homologous recombination (HR) by reducing cellular NAD+ levels and suppressing poly ADP ribosylation, a crucial post-translational modification required for assembling DNA repair proteins.50 The cGAS–STING signaling pathway also plays a role in lipid and glucose metabolism, potentially contributing to obesity, diabetes, nonalcoholic fatty liver disease (NAFLD), and cardiovascular diseases (CVDs).51–53 Cellular senescence is a biological process that facilitates generational change but has also emerged as a significant contributor to aging, age-related pathologies, and tumorigenesis.54,55 Recent studies have found that cGAS–STING signaling contributes to pro-tumorigenic effects and aging-related diseases by triggering the senescence-associated secretory phenotype (SASP) in senescent cells.56,57 Cell death encompasses a variety of types, including apoptosis, necroptosis, pyroptosis, ferroptosis, and autophagic/lysosomal cell death.58,59 More importantly, cGAS-STING signaling is involved in all of them.60 Particularly, cGAS-STING signaling-mediated autophagy is highly exploited.38 However, there remains a significant gap between our understanding of cGAS–STING signaling and the pathological mechanisms and cellular functions it mediates.

    Crosstalk of cGAS-STING Signaling with Other Signaling Transductions and Cellular Processes

    Upstream Regulators

    Caspase family members are protease enzymes essential for initiating cell death through their unique cysteine-dependent protease activity. In this process, a cysteine residue in the active site performs a nucleophilic attack, cleaving target proteins immediately after aspartic acid residues.61 The majority of caspases act as negative regulators of the cGAS–STING pathway. For example, Caspase-1 reduces IFN-β production by binding to and cleaving cGAS at residues D140/157, subsequently dampening TBK1 phosphorylation and IRF3 nuclear translocation in Mycobacterium bovis–infected cells.62,63 Conversely, the absence of Caspase-1 leads to increased IFN-I production during DNA virus infections. Similarly, Caspase-3 cleaves cGAS at D319, MAVS at D429/490, and IRF3 at D121/125 in cells infected with DNA or RNA viruses, resulting in decreased IFN-I production.64 Additionally, in humans, caspase-4 and caspase-5, along with caspase-11 in mice, act as upstream activators of caspase-1, facilitating the cleavage of IL-1β and IL-18.65,66 However, the specific cleavage sites on cGAS require further investigation. Moreover, Caspases-3, −7, and −9 promote IFN-β secretion from apoptotic cells while inhibiting STING recruitment, as well as the phosphorylation and dimerization of TBK1 and IRF3.67 This suggests that they function as inhibitors of signaling pathways activated by mitochondrial DNA (mtDNA)-mediated damage-associated molecular patterns (DAMPs), thereby reducing IFN-α/β production via the cGAS–STING–TBK1–IRF3 axis.67 Subsequently, the cGAS–STING signaling pathway initiates caspase-3, −7, and −9 dependent apoptosis by promoting the degradation of X-linked inhibitor of apoptosis (XIAP), following TBK1/IKK-mediated phosphorylation of XIAP at serine 430.68

    Downstream Signaling

    Pyroptosis is a highly inflammatory form of programmed cell death characterized by cell rupture, typically occurring during infections with intracellular pathogens and playing a crucial role in the antimicrobial response.69 When pathogens infiltrate cells or cellular stress disrupts mitochondrial homeostasis, double-stranded DNA can accumulate in the cytoplasm, triggering inflammatory responses via activation of the cGAS–STING pathway. This, in turn, initiates pyroptosis through the formation of a large supramolecular complex—known as the inflammasome (or pyroptosome)—in response to intracellular danger signals.70 Additionally, inflammasome-associated proteins—such as caspase-1, gasdermin D, the CARD domain of ASC, and potassium channels—play regulatory roles in the cGAS–STING pathway.70 This intricate crosstalk leads to a cascade amplification effect, intensifying the immune response and potentially exacerbating the pathological processes underlying inflammatory and autoimmune diseases.

    Autophagy is an essential and evolutionarily conserved cellular process that degrades unnecessary or dysfunctional components through a lysosome-dependent mechanism. In 2019, research teams led by Zhijian J. Chen and Quan Chen identified a novel link between autophagy and the cGAS–STING signaling pathway. They discovered that STING can activate autophagy as a primary function of the pathway—independently of TBK1 activation and interferon induction. Instead, this process relies on WD repeat domain phosphoinositide-interacting protein 2 (WIPI2) and autophagy-related protein 5 (ATG5) to facilitate LC3 lipidation, thereby promoting the formation of autophagosomes.38,71 Autophagy plays a dual role in cGAS–STING signaling by both promoting inflammatory responses and facilitating the degradation of STING.72,73 Since then, an increasing number of studies have confirmed that several substances—such as Meteorin-like hormone (Metrnl), the deubiquitinating enzyme TRABID, Activin A, Unc-93 homolog B1 (UNC93B1), and metformin—can inactivate cGAS–STING signaling through autophagy-dependent mechanisms, thereby alleviating disease or suppressing pathological cellular processes.74–78

    Functional Interaction

    NF-κB is a critical inflammatory pathway that regulates the immune response to infection and can be activated downstream of cGAS–STING signaling as a transcriptional regulator. Moreover, NF-κB activation—mediated by signaling pathways such as Toll-like receptors (TLRs), interleukin-1 receptor (IL-1R), tumor necrosis factor receptors (TNFRs), growth factor receptors (GFRs), and protein kinase C (PKC)—can induce microtubule depolymerization, which inhibits STING’s trafficking and degradation in lysosomes via the microtubule network.79 Taken together, these findings indicate that cGAS–STING signaling can initiate NF-κB activation, while NF-κB activation, in turn, enhances cGAS–STING signaling by regulating microtubule-mediated STING trafficking.

    Yes-associated protein (YAP) functions as a transcriptional coregulator, promoting the expression of genes that drive cell proliferation while simultaneously suppressing those that induce apoptosis.80 Studies have shown that cGAMP directly promotes YAP phosphorylation at serine residues S127 and S397, thereby regulating cell proliferation through the YAP signaling pathway.81,82 Additionally, activation of the cGAS–STING pathway can trigger the Hippo signaling pathway, resulting in the inactivation of YAP1 and its paralog TAZ (also known as WWTR1), which contributes to the suppression of tumorigenesis.83,84 Interestingly, previous research has found that activation of YAP/TAZ can inhibit cGAS–STING signaling, thereby impairing immune surveillance in non-small cell lung cancer (NSCLC).84

    cGAS-STING Signaling in Fibrosis Diseases

    cGAS-STING Signaling in Pulmonary Fibrosis

    Pulmonary fibrosis (PF) refers to a group of lung diseases characterized by progressive scarring and stiffening of lung tissue, leading to an irreversible decline in the lungs’ oxygen diffusion capacity.85 PF is classified as one of the interstitial lung diseases (ILDs), which can result from identifiable causes such as environmental pollutants, certain medications, and infections. Similar to other ILDs, idiopathic pulmonary fibrosis (IPF)—a form with no known cause—is defined as a chronic, progressive fibrotic lung disease.86,87 The current understanding of PF pathogenesis is that it results from an abnormal wound-healing response in lung tissue, triggered by various disease-specific factors. These factors activate key effector cells—primarily pulmonary fibroblasts—leading to excessive inflammation and fibrosis.88,89 Acute lung inflammation is considered a precursor to PF, while chronic inflammation is thought to promote its progression.90,91 Recent studies have highlighted that the aberrant activation of the cGAS-STING pathway contributes to pulmonary inflammation and fibrosis.

    A previous study showed that polystyrene microplastics—emerging environmental pollutants—induced ferroptosis, leading to varying degrees of lung damage and fibrosis in a mouse model by activating the cGAS–STING signaling pathway, and inhibition of both ferroptosis and cGAS–STING signaling reduced lung injury and pulmonary fibrosis following exposure to polystyrene microplastics.92 Similarly, Ficolin B—a recognition molecule secreted via alveolar macrophage-derived exosomes—was found to exacerbate bleomycin-induced lung injury through cGAS–STING-mediated ferroptosis.93 Additionally, PF was induced in mice by pharyngeal aspiration of a novel nanomaterial, graphitized multi-walled carbon nanotubes (GMWCNTs), which triggered high expression of the cGAS–STING signaling pathway at the gene level, but co-administration of a STING inhibitor effectively reduced GMWCNT-induced pulmonary inflammation and fibrosis.94 Furthermore, honokiol—a natural extract from Magnolia bark with known antioxidant and anti-inflammatory properties—was shown to attenuate silica-induced pyroptosis and protect against PF by modulating the cGAS–STING signaling pathway, offering potential therapeutic applications for silicosis and related inflammatory responses.95 Moreover, fluvoxamine, a selective serotonin reuptake inhibitor, has demonstrated therapeutic effects in IPF by reducing fibroblast activation and migration in response to transforming growth factor-beta 1 (TGF-β1) stimulation, through inhibition of the cGAS–STING signaling pathway and its downstream targets96 (Figure 2).

    Figure 2 cGAS-STING signaling in pulmonary, hepatic, renal, cardiac, and cutaneous inflammation and fibrosis. In pulmonary fibrosis, polystyrene microplastics (PS-MPs), graphitized multi-walled carbon nanotubes (GMWCNTs), silica and bleomycin could activate cGAS-STING signaling and result in pulmonary inflammation and fibrosis. Previous research also illustrated that STING plays a protective role in pulmonary fibrosis associated with prolonged neutrophilic inflammation. In hepatic fibrosis, TAR DNA-binding protein 43 (TDP-43), transforming growth factor-beta (TGF-β), polystyrene microplastics (PS-MPs), hexafluoropropylene oxide trimer acid (HFPO-TA) and X-box binding protein 1 (XBP1) could upregulate cGAS-STING activation and lead to hepatic stellate cells (HSCs) activation, resulting in hepatic fibrosis. Whereas Oroxylin A could antagonize hepatic fibrosis by cGAS-STING/IRF3-induced HSCs senescence via IRF3 and retinoblastoma (RB) interaction. In renal fibrosis, hypoxia, chronic heat, N6-adenosine-methyltransferase 70 kDa subunit (METTL3), mitochondrial transcription factor A (TFAM) deficiency and protein kinase C-delta (PKC-δ) could activate cGAS-STING signaling and result in renal inflammation and fibrosis. In cardiac fibrosis, cardiomyocytes-derived small extracellular vesicles (sEVs) and hypertension could induce myocardial infarction (MI) and cardiac fibrosis by activating cGAS-STING signaling. In cutaneous fibrosis, the autoimmune disorder is the cause of Systemic Sclerosis and leads to cGAS-STING activation, resulting in fibrosis in the skin. However, burn injury, wound infection or surgery-induced pathological scarring, characterized by excessive accumulation of extracellular matrix (ECM), could be driven through cGAS-STING activation or not is unknown.

    Paradoxically, some research illustrated that STING plays a protective role in PF that is independent of IFN-I signaling but associated with prolonged neutrophilic inflammation, showing that STING deficiency leads to aggravated PF, characterized by increased collagen deposition in the lungs and excessive expression of remodeling factors97 (Figure 2).

    Anyhow, the paradoxical effects of cGAS-STING signaling in pulmonary fibrosis may stem from differences in the microenvironments studied or from crosstalk with other signaling pathways. Therefore, further investigation into the precise role of cGAS-STING signaling in PF is essential to develop effective therapies.

    cGAS-STING Signaling in Renal Fibrosis

    Renal fibrosis (RF) represents the final common pathway of progressive kidney diseases, culminating in irreversible renal failure. It is characterized by excessive extracellular matrix (ECM) deposition that leads to parenchymal scarring, making it the most prevalent pathological feature of chronic kidney diseases (CKDs).98 Due to the severity and irreversibility of kidney failure, treatment options such as hemodialysis, peritoneal dialysis, or kidney transplantation often impose significant inconvenience and financial burdens on patients.99,100 Currently, it is widely accepted that RF represents a failed wound-healing process in kidney tissue. This leads to tubular atrophy, persistent interstitial inflammation, tissue fibrosis, glomerular scarring, and reduced vascular density following sustained injury. The process involves activation of mesangial cells and fibroblasts, as well as tubular epithelial-mesenchymal transition, which serve as the primary sources of matrix-producing cells in kidney disease.101,102 Inflammation triggered by kidney injury initially acts as a protective response; however, when prolonged and excessive, it contributes to the progression of renal diseases, ultimately leading to end-stage RF.103 Therefore, as one of the key inflammatory pathways, cGAS-STING signaling presents significant regulations in renal inflammation and fibrosis.

    Studies have demonstrated that the cGAS-STING signaling pathway promotes RF under hypoxic conditions, a process linked to glycolysis mediated by 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3). Inhibition of STING or IRF3 effectively reduced the elevated expression of PFKFB3 and alleviated hypoxia-induced RF. These findings suggest that the cGAS-STING/IRF3/PFKFB3 signaling axis may serve as a promising therapeutic target for renal inflammation and early-stage RF.104 In addition, previous research demonstrated that m6A RNA modification, facilitated by the 70 kDa subunit of N6-adenosine-methyltransferase (METTL3), may promote renal fibrosis by specifically enriching and stabilizing the cGAS-STING signaling pathway.105 Interestingly, Ki Wung Chung et al observed significant mitochondrial defects in both human and animal models of renal fibrosis, characterized by a deficiency of mitochondrial transcription factor A (TFAM) in renal tubular cells.106 Furthermore, they discovered that improper packaging of mtDNA led to its leakage into the cytosol, triggering cGAS-STING activation in renal cells of TFAM knockout mice. Additionally, deletion of STING ameliorated renal fibrosis in these mouse models, highlighting a critical regulatory role of cGAS-STING signaling in renal inflammation and fibrosis.106 Moreover, chronic heat exposure was found to induce renal fibrosis and mitochondrial damage in laying hens by activating the mtDNA-cGAS-STING signaling pathway, which subsequently triggered inflammation.107 Thereafter, protein kinase C-delta (PKC-δ) was found to be significantly upregulated in renal fibrosis biopsy samples from both humans and mice. Furthermore, the PKC-δ inhibitor rottlerin attenuated renal fibrosis induced by unilateral ureteral ligation and suppressed activation of the cGAS-STING signaling pathway, highlighting PKC-δ as a key regulator of renal fibrosis via this pathway108 (Figure 2).

    Based on the above research findings, the cGAS-STING signaling pathway plays a critical role in the pathogenesis and progression of renal inflammation and renal fibrosis. Therefore, targeting molecules or employing techniques that modulate the cGAS-STING pathway may offer promising new therapeutic strategies for kidney inflammation and RF.

    cGAS-STING Signaling in Hepatic Fibrosis

    Hepatic fibrosis (HF) is characterized by excessive accumulation of connective tissue in the liver due to an overly exuberant wound-healing response triggered by chronic or repeated injury. This condition occurs in most types of chronic liver diseases (CLDs), especially those with inflammatory components, and can ultimately lead to portal hypertension—where scarring impedes blood flow through the liver—or cirrhosis, which distorts normal liver structure and function.109,110 However, unlike pulmonary or renal fibrosis, some forms of HF are potentially reversible. Based on current understanding, HF develops when hepatic stellate cells (HSCs) become activated and transdifferentiate into hepatic myofibroblasts in response to injury or inflammation.109,111 Hepatitis refers to liver inflammation caused by viral infections or liver injury and can present as either an acute condition (lasting less than six months) or a chronic condition (persisting for six months or more).112,113 The key difference is that acute (short-term) hepatitis often resolves on its own, whereas chronic hepatitis can lead to severe and potentially fatal complications, including cirrhosis, liver cancer, and liver failure.114 With increasing research into the pathogenesis of liver diseases, the cGAS–STING signaling pathway has been shown to promote liver inflammation and fibrosis.115

    Evidence has shown that activation of the cGAS–STING signaling pathway, triggered by TAR DNA-binding protein 43 (TDP-43)-induced mtDNA release, is involved in HF. Moreover, the activation of cGAS–STING signaling and the colocalization of mitochondria with TDP-43 were found to correlate with the severity of HF.116 Subsequently, another study reported similar findings: activation of the cGAS–STING signaling pathway, along with mtDNA release induced by TGF-β, is essential for the transcriptional regulation and transdifferentiation of HSCs, a key event in the initiation of HF. They also found that inhibition of STING effectively blocked TGF-β-induced HSCs transdifferentiation and reduced HF both prophylactically and therapeutically.117 In addition, recent studies have demonstrated that polystyrene microplastics not only contribute to lung injury and PF, but their prolonged accumulation also induces HF. Rong Shen et al revealed that polystyrene microplastics smaller than 1μm can enter cells and accumulate in the liver via the bloodstream, and even at low concentrations, these particles may cause hepatic injury and dysfunction by activating the cGAS–STING–NF-κB signaling pathway.118 Similarly, hexafluoropropylene oxide trimer acid (HFPO-TA), a contemporary substitute for perfluorooctanoic acid with emerging environmental toxicity, has been shown to induce a cascade of pathological events in mice.119 Specifically, HFPO-TA exposure increased mitochondrial reactive oxygen species (mtROS) levels, activated the cGAS–STING signaling pathway, and triggered NLRP3-mediated pyroptosis, ultimately leading to HF.119 These findings suggest that HFPO-TA–induced HF involves a mtROS-driven cGAS–STING–NLRP3 pyroptotic signaling axis.119 Furthermore, X-box binding protein 1 (XBP1), a transcription factor essential for regulating immune-related gene expression, has been shown to activate STING signaling in macrophages by promoting the cytosolic leakage of self-derived mtDNA, thereby contributing to the progression of HF.120 Accordingly, Li Chen et al demonstrated that naringenin, a natural flavonoid with anti-inflammatory properties, functions as a specific antagonist of cGAS, which effectively prevents HSCs activation and disrupts the secretion of proinflammatory factors mediated by the cGAS-STING signaling pathway, thereby impeding the progression of HF in murine models18 (Figure 2).

    Interestingly, a recent study demonstrated that treatment with Oroxylin A—a naturally occurring O-methylated flavone derived from Scutellaria baicalensis Georgi—can activate ferritinophagy in HSCs via the cGAS-STING signaling pathway, thereby inducing HSCs senescence and subsequently attenuating HF.121 Additionally, Qirou Wu et al unexpectedly discovered that IRF3, activated through the cGAS-STING signaling pathway, forms significant endogenous nuclear complexes with retinoblastoma protein (RB), a key regulator of the cell cycle. This interaction attenuates RB hyperphosphorylation, thereby promoting HSCs senescence. These findings reveal an unforeseen yet pivotal role of STING–IRF3–RB signaling in inducing HSCs senescence and limiting the progression of HF122 (Figure 2).

    Similar to its role in PF, the cGAS–STING signaling pathway exhibits both protective and pathogenic effects in HF. Therefore, further investigation is essential to elucidate the precise pathophysiological mechanisms by which cGAS–STING signaling contributes to liver inflammation and fibrosis.

    cGAS-STING Signaling in Cardiac Fibrosis

    Cardiac fibrosis (CF) is a pathological condition marked by excessive ECM deposition in the myocardium, or abnormal thickening of the pericardium and endocardium—particularly around heart valves such as the tricuspid and pulmonary valves—resulting from aberrant proliferation and activation of cardiac fibroblasts (CFs). This process increases the risk of arrhythmias, impairs cardiac function, and ultimately contributes to the development of heart failure.123,124 Similar to HF, changes associated with CF may be reversible in certain contexts, as the condition can be self-limiting and may resolve completely with appropriate intervention or removal of the underlying cause.

    The most extensive fibrotic remodeling of the heart typically occurs following acute cardiomyocyte death, which results in the abrupt loss of a substantial number of cardiomyocytes. This event triggers a robust inflammatory response and subsequently leads to the replacement of necrotic myocardium with collagen-rich scar tissue.124 Additionally, pressure overload conditions—such as hypertension and aortic stenosis—as well as volume overloads caused by valvular regurgitant lesions, along with cardiomyopathies like hypertrophic or post-viral dilated cardiomyopathy, can also lead to extensive CF. This fibrotic remodeling increases myocardial stiffness and impairs diastolic function, ultimately contributing to ventricular dilation and the progression to heart failure.123,124

    Heart inflammation is the body’s innate immune response to cardiac infection or injury and is generally classified into three main types based on the affected tissue: endocarditis (inflammation of the inner lining of the heart chambers and valves), myocarditis (inflammation of the heart muscle), and pericarditis (inflammation of the protective sac surrounding the heart).125 To date, activation of the cGAS–STING signaling pathway has been identified as a key promoter of the early inflammatory response, as well as of pathological cardiac remodeling and dysfunction.126 Recent research has shown that small extracellular vesicles (sEVs) derived from cardiomyocytes, containing mitochondrial components, can enter CFs and initiate CF by promoting CFs activation and proliferation through the cGAS–STING signaling pathway.127 Thus, inhibitors of the cGAS-STING signaling pathway have demonstrated protective effects against CF. Lavinia Rech et al confirmed that pharmacological inhibition of STING using C178 or H-151 reduced infarct expansion, myocardial scarring, and hypertrophy three weeks after reperfused myocardial infarction in a preclinical murine model.128 At the same time, another study confirmed that treatment with H-151 significantly preserved myocardial function and attenuated cardiac fibrosis129 (Figure 2).

    Consequently, inhibitors targeting the cGAS-STING signaling pathway may represent promising therapeutics to improve wound healing and pathological remodeling, thereby alleviating CF.

    cGAS-STING Signaling in Cutaneous Fibrosis

    The hallmarks of skin injury—pathogen invasion and localized tissue damage—result in the release of DNA. When this free DNA accumulates in the cytoplasm, it activates the cGAS-STING signaling pathway, triggering the production of IFN-I, inflammatory cytokines, and chemokines, thereby connecting wound healing and scar formation with immune system activation.130 In the environment of tissue injury, resident cells and immune cells release an excess of growth factors—such as fibroblast growth factor (FGF), TGF-β1, IL-1, and IL-6—which activate fibroblasts and promote their differentiation into secretory myofibroblasts through the fibroblast-to-myofibroblast transition process.131 The high quantity of myofibroblasts leads to the excessive production of ECM proteins.132

    Systemic sclerosis (SSc), also known as scleroderma, is a complex autoimmune disease targeting connective tissues and often resulting in progressive skin fibrosis.133 The activation of fibroblasts plays a pivotal role in the progression of fibrosis in SSc, characterized by connective tissue thickening and excessive accumulation of ECM, especially collagen types I and III.131 Previous studies demonstrated that centromere dysfunction leads to chromosomal instability—manifested as aneuploidy and micronuclei formation—in SSc, and they found that the formation of micronuclei in SSc is closely linked to cGAS-STING pathway activation and correlates with the clinical severity of skin fibrosis134 (Figure 2).

    Pathological scarring, including hypertrophic scars and keloids, is a common form of cutaneous fibrosis characterized by thick, raised, red scars that extend above the normal skin surface due to excessive collagen deposition during wound healing.135 Pathological scarring is considered a result of dysregulated inflammation during wound healing, along with persistent chronic inflammation in the reticular dermis.136,137 Additionally, our previous study summarized the immunoregulatory roles of immune cells in wound healing and skin scarring, highlighting the potential involvement of cGAS-STING signaling modulation in scar formation.138 Therefore, regulating the cGAS-STING–mediated inflammatory response during wound healing may offer a promising strategy to prevent or mitigate pathological scar formation (Figure 2).

    Therapeutic Potential Targeting cGAS-STING Signaling in Fibrosis Diseases

    Considering that immunomodulation of the cGAS-STING signaling pathway represents a promising target for first-in-class immunotherapies, inhibitors—particularly those targeting the key protein STING—have shown encouraging therapeutic potential in preclinical models of fibrotic diseases.16 Each of these inhibitors is specifically discussed in the following paragraphs (Table 1).

    Table 1 Application of cGAS–STING Pathway-Related Inhibitors in Fibrosis Diseases

    RU.521

    RU.521 is a potent and selective inhibitor of cGAS that effectively blocks cGAS-mediated upregulation of IFN-I. Studies have shown that intracisternal administration of RU.521 reduces microglial activation and neuroinflammation, as well as restores the balance between sympathetic and parasympathetic nervous system activities, which collectively contribute to lowering blood pressure.139 This reduction in hypertension subsequently alleviates myocardial interstitial fibrosis, cardiomyocyte hypertrophy, and impaired cardiac function in Angiotensin II (Ang II)-induced hypertensive mice139 (Figure 3).

    Figure 3 Application of inhibitors and natural products targeting cGAS and STING in fibrosis diseases.

    H-151

    H-151 is a potent and selective covalent antagonist of STING that effectively suppresses STING palmitoylation and reduces TBK1 phosphorylation, demonstrating strong inhibitory effects both in vitro and in vivo. Shiyu Hu et al showed that H-151 treatment markedly decreases the IFN-I response in bone marrow-derived macrophages (BMDMs) stimulated by cardiac dsDNA, which the suppression leads to reduced apoptosis of adult cardiomyocytes and decreased scar formation in cardiac fibroblasts cultured with conditioned medium from BMDMs.129 These findings indicate that H-151-mediated inhibition of STING can preserve myocardial function and attenuate cardiac fibrosis following myocardial infarction (Figure 3).

    C-176 and C-178

    C-176 and C-178 are nitrofuran derivatives that selectively bind covalently to the transmembrane cysteine residue 91 of STING, effectively blocking activation-induced STING palmitoylation. These compounds also possess the ability to cross the blood-brain barrier. A recent study demonstrated that treatment with C-176 significantly reduced histopathological features in a mouse model of PF induced by GMWCNTs.94 Specifically, it decreased alveolar wall thickening, alveolar collapse, and inflammatory cell infiltration. Also, mRNA and protein expression levels of cGAS, STING, NF-κB, IL-1β, and TGF-β1 were markedly reduced after C-176 administration, suggesting that STING inhibition by C-176 may effectively mitigate pulmonary inflammation and fibrosis.94 However, it was identified that the species-specific activity of C-178 and C-176 compounds directly targets mouse STING but not human STING145 (Figure 3).

    IFM-0044907

    IFM-0044907 is a small-molecule inhibitor of STING. Suyavaran Arumugam et al demonstrated that IFM-0044907 effectively blocked the transactivation of HSCs and reversed carbon tetrachloride (CCl₄)-induced HF in mice.117 These findings underscore the pivotal role of STING in HF and highlight IFM-0044907 as a promising therapeutic candidate for treating liver fibrosis (Figure 3).

    Other Natural Products

    Several natural products targeting the cGAS-STING signaling pathway have been identified to attenuate PF. For instance, 20(S)-Protopanaxadiol, derived from ginseng, and Heterophyllin B, extracted from Radix Pseudostellariae, demonstrated promising pharmacological effects on bleomycin-induced PF by reducing STING expression through the phosphorylation and activation of AMPK.140,141 Tanreqing injection (TRQ), a well-established traditional Chinese medicine, effectively alleviates bleomycin-induced pulmonary fibrosis by inhibiting STING signaling through modulation of endoplasmic reticulum stress pathways.142 Juglanin, a compound derived from the green husks of walnuts (Juglans mandshurica) or the Chinese herb He Shou Wu, can reduce inflammation and attenuate pulmonary fibrosis by inhibiting STING in human lung fibroblasts and mouse epithelial cells, as well as downregulating fibrotic markers such as TGF-β1, α-SMA, fibronectin, matrix metalloproteinase-9 (MMP-9), and collagen I.143 Qingfei Xieding prescription, a traditional Chinese medicine effective as an adjuvant treatment for pulmonary diseases, alleviated bleomycin-induced PF by activating autophagy and inhibiting inflammation mediated through the mtDNA-cGAS-STING pathway144 (Figure 3).

    However, despite the therapeutic potential of existing cGAS-STING signaling inhibitors, challenges persist in developing novel agonists and optimizing agonist delivery systems to enhance biotherapeutic outcomes.

    Future Perspective

    Organ fibrosis accounts for up to 45% of all deaths in the developed world due to its relentlessly progressive and irreversible nature.4 Despite significant progress in understanding the pathobiology of fibrosis, effective therapies for clinical patients remain limited. Given that inflammatory regulation plays a key role in the initiation and progression of fibrosis, targeting inflammatory pathways and molecules offers promising therapeutic potential.146

    The cGAS-STING signaling pathway has emerged as a critical mediator of inflammation in response to infection, cellular stress, and tissue damage.147 It has also shown significant roles in various pathological conditions, including inflammatory diseases, cancer, autoimmune disorders, fibrotic diseases, and neurodegeneration.41,148–150 With growing research into the cellular and molecular mechanisms of cGAS-STING signaling in fibrotic diseases—including pulmonary, renal, hepatic, cardiac, and cutaneous fibrosis—most studies indicate that its aberrant activation acts as a promoter of organ fibrosis onset and progression.16,106,115,126,134 Thus, strategies targeting the cGAS-STING signaling pathway have demonstrated therapeutic potential for fibrotic diseases. For instance, Quzhou Fructus Arantii-nB, fluvoxamine, and honokiol have been shown to alleviate PF by inhibiting cGAS-STING signaling; naringenin disrupts this pathway to prevent HF; and H-151 treatment significantly preserves myocardial function and attenuates CF by specifically targeting STING.18,95,96,129,151 Consequently, these findings indicate that the cGAS-STING signaling pathway contributes to fibrotic processes, and its inhibition shows promising therapeutic potential for organ fibrosis. However, there is also evidence suggesting that cGAS-STING signaling may exert protective roles in certain contexts, such as PF and HF, highlighting the complexity and context-dependent nature of this pathway.97,121,122 Therefore, further in-depth research is required to elucidate the crosstalk between cGAS-STING signaling and other signaling pathways involved in fibrotic processes, as well as to advance its pharmaceutical translation into clinical applications.

    Pathological scars, such as hypertrophic scars and keloids, have long posed significant challenges for both patients and clinicians due to the lack of effective therapies—particularly in the case of keloids, which are characterized by a high recurrence rate.152 This is largely due to the unclear pathogenesis of hypertrophic scars and keloids. Nevertheless, inflammation within the microenvironment of skin wounds and scars plays a critical role in the processes of wound healing and scar formation.137 To date, no studies have specifically investigated the role of cGAS-STING signaling in the pathogenesis of hypertrophic scars or keloids. Given its established involvement in inflammation and fibrosis, this pathway may represent a promising therapeutic target, warranting further investigation.

    To sum up, the role of cGAS-STING signaling in fibrotic diseases is complex and multifaceted, influenced by various factors such as individual variability, tissue microenvironment, the extent and duration of pathway activation, the functional state of resident cells, and potential contributions from comorbid conditions. Despite growing interest, current evidence remains insufficient to clearly define how cGAS-STING signaling modulates fibrotic processes or whether targeting this pathway can effectively reverse or halt organ fibrosis. Therefore, future research should aim to elucidate the precise cellular and molecular mechanisms involved, develop selective and safe therapeutic agents, and evaluate their efficacy in clinical settings. Continued exploration of the cGAS-STING pathway holds promise for advancing treatment strategies and improving outcomes in fibrotic diseases.

    Conclusions

    This review provides a comprehensive overview of the effects and molecular mechanisms of the cGAS-STING signaling pathway in fibrotic diseases. It outlines the core components of the cGAS-STING cascade, summarizes the mechanisms by which its activation contributes to fibrosis across multiple organs, and catalogs recently developed cGAS and STING antagonists explored in fibrotic disease models. Furthermore, it discusses the therapeutic potential and translational prospects of targeting this pathway in clinical settings.

    Author Contributions

    All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

    Disclosure

    The authors report no conflicts of interest in this work.

    References

    1. Lurje I, Gaisa NT, Weiskirchen R, et al. Mechanisms of organ fibrosis: emerging concepts and implications for novel treatment strategies. Mol Aspects Med. 2023;92:101191. doi:10.1016/j.mam.2023.101191

    2. Weiskirchen R, Weiskirchen S, Tacke F. Organ and tissue fibrosis: molecular signals, cellular mechanisms and translational implications. Mol Aspects Med. 2019;65:2–15. doi:10.1016/j.mam.2018.06.003

    3. Henderson NC, Rieder F, Wynn TA. Fibrosis: from mechanisms to medicines. Nature. 2020;587(7835):555–566. doi:10.1038/s41586-020-2938-9

    4. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008;214(2):199–210. doi:10.1002/path.2277

    5. Antar SA, Ashour NA, Marawan ME, et al. Fibrosis: types, effects, markers, mechanisms for disease progression, and its relation with oxidative stress, immunity, and inflammation. Int J Mol Sci. 2023;24(4):4004. doi:10.3390/ijms24044004

    6. Huang E, Peng N, Xiao F, et al. The roles of immune cells in the pathogenesis of fibrosis. Int J Mol Sci. 2020;21(15):5203. doi:10.3390/ijms21155203

    7. Zhang WJ, Chen SJ, Zhou SC, et al. Inflammasomes and Fibrosis. Front Immunol. 2021;12:643149. doi:10.3389/fimmu.2021.643149

    8. Malaviya R, Kipen HM, Businaro R, et al. Pulmonary toxicants and fibrosis: innate and adaptive immune mechanisms. Toxicol Appl Pharmacol. 2020;409:115272. doi:10.1016/j.taap.2020.115272

    9. Siamwala JH, Zhao A, Barthel H, et al. Adaptive and innate immune mechanisms in cardiac fibrosis complicating pulmonary arterial hypertension. Physiol Rep. 2020;8(15):e14532. doi:10.14814/phy2.14532

    10. Khanam A, Chua JV, Kottilil S. Immunopathology of chronic hepatitis B infection: role of innate and adaptive immune response in disease progression. Int J Mol Sci. 2021;22(11):5497. doi:10.3390/ijms22115497

    11. Ishikawa H, Barber GN. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature. 2008;455(7213):674–678. doi:10.1038/nature07317

    12. Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol. 2015;15(12):760–770. doi:10.1038/nri3921

    13. Hopfner KP, Hornung V. Molecular mechanisms and cellular functions of cGAS-STING signalling. Nat Rev Mol Cell Biol. 2020;21(9):501–521. doi:10.1038/s41580-020-0244-x

    14. Chauvin SD, Stinson WA, Platt DJ, et al. Regulation of cGAS and STING signaling during inflammation and infection. J Biol Chem. 2023;299(7):104866. doi:10.1016/j.jbc.2023.104866

    15. Chen C, Xu P. Cellular functions of cGAS-STING signaling. Trends Cell Biol. 2023;33(8):630–648. doi:10.1016/j.tcb.2022.11.001

    16. Zhang J, Zhang L, Chen Y, et al. The role of cGAS-STING signaling in pulmonary fibrosis and its therapeutic potential. Front Immunol. 2023;14:1273248. doi:10.3389/fimmu.2023.1273248

    17. Luo W, Xu G, Song Z, et al. Licorice extract inhibits the cGAS-STING pathway and protects against non-alcoholic steatohepatitis. Front Pharmacol. 2023;14:1160445. doi:10.3389/fphar.2023.1160445

    18. Chen L, Xia S, Wang S, et al. Naringenin is a potential immunomodulator for inhibiting liver fibrosis by inhibiting the cGAS-STING pathway. J Clin Transl Hepatol. 2023;11(1):26–37. doi:10.14218/JCTH.2022.00120

    19. Zhang D, Liu Y, Zhu Y, et al. A non-canonical cGAS-STING-PERK pathway facilitates the translational program critical for senescence and organ fibrosis. Nat Cell Biol. 2022;24(5):766–782. doi:10.1038/s41556-022-00894-z

    20. Gekara NO, Jiang H. The innate immune DNA sensor cGAS: a membrane, cytosolic, or nuclear protein? Sci Signal. 2019;12(581):eaax3521. doi:10.1126/scisignal.aax3521

    21. Barnett KC, Coronas-Serna JM, Zhou W, et al. Phosphoinositide interactions position cGAS at the plasma membrane to ensure efficient distinction between self- and viral DNA. Cell. 2019;176(6):1432–1446.e1411. doi:10.1016/j.cell.2019.01.049

    22. Civril F, Deimling T, de Oliveira Mann CC, et al. Structural mechanism of cytosolic DNA sensing by cGAS. Nature. 2013;498(7454):332–337. doi:10.1038/nature12305

    23. Zhang X, Bai XC, Chen ZJ. Structures and mechanisms in the cGAS-STING innate immunity pathway. Immunity. 2020;53(1):43–53. doi:10.1016/j.immuni.2020.05.013

    24. Blest HTW, Chauveau L. cGAMP the travelling messenger. Front Immunol. 2023;14:1150705. doi:10.3389/fimmu.2023.1150705

    25. Zaver SA, Woodward JJ. Cyclic dinucleotides at the forefront of innate immunity. Curr Opin Cell Biol. 2020;63:49–56. doi:10.1016/j.ceb.2019.12.004

    26. Burdette DL, Monroe KM, Sotelo-Troha K, et al. STING is a direct innate immune sensor of cyclic di-GMP. Nature. 2011;478(7370):515–518. doi:10.1038/nature10429

    27. Ouyang S, Song X, Wang Y, et al. Structural analysis of the STING adaptor protein reveals a hydrophobic dimer interface and mode of cyclic di-GMP binding. Immunity. 2012;36(6):1073–1086. doi:10.1016/j.immuni.2012.03.019

    28. Shu C, Yi G, Watts T, et al. Structure of STING bound to cyclic di-GMP reveals the mechanism of cyclic dinucleotide recognition by the immune system. Nat Struct Mol Biol. 2012;19(7):722–724. doi:10.1038/nsmb.2331

    29. Shang G, Zhu D, Li N, et al. Crystal structures of STING protein reveal basis for recognition of cyclic di-GMP. Nat Struct Mol Biol. 2012;19(7):725–727. doi:10.1038/nsmb.2332

    30. Lu D, Shang G, Li J, et al. Activation of STING by targeting a pocket in the transmembrane domain. Nature. 2022;604(7906):557–562. doi:10.1038/s41586-022-04559-7

    31. Taguchi T, Mukai K, Takaya E, et al. STING operation at the ER/golgi interface. Front Immunol. 2021;12:646304. doi:10.3389/fimmu.2021.646304

    32. Ishikawa H, Ma Z, Barber GN. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature. 2009;461(7265):788–792. doi:10.1038/nature08476

    33. Dobbs N, Burnaevskiy N, Chen D, et al. STING activation by translocation from the ER is associated with infection and autoinflammatory disease. Cell Host Microbe. 2015;18(2):157–168. doi:10.1016/j.chom.2015.07.001

    34. Tanaka Y, Chen ZJ. STING specifies IRF3 phosphorylation by TBK1 in the cytosolic DNA signaling pathway. Sci Signal. 2012;5(214):ra20. doi:10.1126/scisignal.2002521

    35. Yum S, Li M, Fang Y, et al. TBK1 recruitment to STING activates both IRF3 and NF-κB that mediate immune defense against tumors and viral infections. Proc Natl Acad Sci U S A. 2021;118(14):e2100225118. doi:10.1073/pnas.2100225118

    36. Zhou X, Meng F, Xu B, et al. STING promotes invasion and migration of uveal melanoma through p38‑MAPK signaling. Oncol Rep. 2024;51(2):23. doi:10.3892/or.2023.8682

    37. Zhang W, Li G, Luo R, et al. Cytosolic escape of mitochondrial DNA triggers cGAS-STING-NLRP3 axis-dependent nucleus pulposus cell pyroptosis. Exp Mol Med. 2022;54(2):129–142. doi:10.1038/s12276-022-00729-9

    38. Gui X, Yang H, Li T, et al. Autophagy induction via STING trafficking is a primordial function of the cGAS pathway. Nature. 2019;567(7747):262–266. doi:10.1038/s41586-019-1006-9

    39. Wang L, Zhang S, Liu H, et al. STING activation in cardiomyocytes drives hypertrophy-associated heart failure via NF-κB-mediated inflammatory response. Biochim Biophys Acta Mol Basis Dis. 2023;1870(3):166997. doi:10.1016/j.bbadis.2023.166997

    40. Erttmann SF, Swacha P, Aung KM, et al. The gut microbiota prime systemic antiviral immunity via the cGAS-STING-IFN-I axis. Immunity. 2022;55(5):847–861.e810. doi:10.1016/j.immuni.2022.04.006

    41. Samson N, Ablasser A. The cGAS-STING pathway and cancer. Nat Cancer. 2022;3(12):1452–1463. doi:10.1038/s43018-022-00468-w

    42. Chin EN, Sulpizio A, Lairson LL. Targeting STING to promote antitumor immunity. Trends Cell Biol. 2023;33(3):189–203. doi:10.1016/j.tcb.2022.06.010

    43. Hu Y, Chen B, Yang F, et al. Emerging role of the cGAS-STING signaling pathway in autoimmune diseases: biologic function, mechanisms and clinical prospection. Autoimmun Rev. 2022;21(9):103155. doi:10.1016/j.autrev.2022.103155

    44. Paul BD, Snyder SH, Bohr VA. Signaling by cGAS-STING in neurodegeneration, neuroinflammation, and aging. Trends Neurosci. 2021;44(2):83–96. doi:10.1016/j.tins.2020.10.008

    45. Ferecskó AS, Smallwood MJ, Moore A, et al. STING-triggered CNS inflammation in human neurodegenerative diseases. Biomedicines. 2023;11(5):1375. doi:10.3390/biomedicines11051375

    46. Govindarajulu M, Ramesh S, Beasley M, et al. Role of cGAS-sting signaling in alzheimer’s disease. Int J Mol Sci. 2023;24(9):8151. doi:10.3390/ijms24098151

    47. Ma C, Liu Y, Li S, et al. Microglial cGAS drives neuroinflammation in the MPTP mouse models of parkinson’s disease. CNS Neurosci Ther. 2023;29(7):2018–2035. doi:10.1111/cns.14157

    48. Zamiri K, Kesari S, Paul K, et al. Therapy of autoimmune inflammation in sporadic amyotrophic lateral sclerosis: dimethyl fumarate and H-151 downregulate inflammatory cytokines in the cGAS-STING pathway. FASEB j. 2023;37(8):e23068. doi:10.1096/fj.202300573R

    49. Liu H, Zhang H, Wu X, et al. Nuclear cGAS suppresses DNA repair and promotes tumorigenesis. Nature. 2018;563(7729):131–136. doi:10.1038/s41586-018-0629-6

    50. Banerjee D, Langberg K, Abbas S, et al. A non-canonical, interferon-independent signaling activity of cGAMP triggers DNA damage response signaling. Nat Commun. 2021;12(1):6207. doi:10.1038/s41467-021-26240-9

    51. Gong J, Gao X, Ge S, et al. The role of cGAS-STING signalling in metabolic diseases: from signalling networks to targeted intervention. Int J Biol Sci. 2024;20(1):152–174. doi:10.7150/ijbs.84890

    52. Ma XM, Geng K, Law BY, et al. Lipotoxicity-induced mtDNA release promotes diabetic cardiomyopathy by activating the cGAS-STING pathway in obesity-related diabetes. Cell Biol Toxicol. 2023;39(1):277–299. doi:10.1007/s10565-021-09692-z

    53. Lv J, Xing C, Chen Y, et al. The STING in non-alcoholic fatty liver diseases: potential therapeutic targets in inflammation-carcinogenesis pathway. Pharmaceuticals. 2022;15(10):1241. doi:10.3390/ph15101241

    54. Zhang L, Pitcher LE, Yousefzadeh MJ, et al. Cellular senescence: a key therapeutic target in aging and diseases. J Clin Invest. 2022;132(15):e158450. doi:10.1172/JCI158450

    55. Schmitt CA, Wang B, Demaria M. Senescence and cancer – role and therapeutic opportunities. Nat Rev Clin Oncol. 2022;19(10):619–636. doi:10.1038/s41571-022-00668-4

    56. Glück S, Guey B, Gulen MF, et al. Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat Cell Biol. 2017;19(9):1061–1070. doi:10.1038/ncb3586

    57. Loo TM, Miyata K, Tanaka Y, et al. Cellular senescence and senescence-associated secretory phenotype via the cGAS-STING signaling pathway in cancer. Cancer Sci. 2020;111(2):304–311. doi:10.1111/cas.14266

    58. Bertheloot D, Latz E, Franklin BS. Necroptosis, pyroptosis and apoptosis: an intricate game of cell death. Cell Mol Immunol. 2021;18(5):1106–1121.

    59. Hadian K, Stockwell BR. The therapeutic potential of targeting regulated non-apoptotic cell death. Nat Rev Drug Discov. 2023;22(9):723–742. doi:10.1038/s41573-023-00749-8

    60. Xu Y, Chen C, Liao Z, et al. cGAS-STING signaling in cell death: mechanisms of action and implications in pathologies. Eur J Immunol. 2023;53(9):e2350386. doi:10.1002/eji.202350386

    61. Kesavardhana S, Malireddi RKS, Kanneganti TD. Caspases in cell death, inflammation, and pyroptosis. Annu Rev Immunol. 2020;38:567–595. doi:10.1146/annurev-immunol-073119-095439

    62. Liao Y, Liu C, Wang J, et al. Caspase-1 inhibits IFN-β production via cleavage of cGAS during M. bovis infection. Vet Microbiol. 2021;258:109126. doi:10.1016/j.vetmic.2021.109126

    63. Wang Y, Ning X, Gao P, et al. Inflammasome activation triggers caspase-1-mediated Cleavage of cGAS to regulate responses to DNA virus infection. Immunity. 2017;46(3):393–404. doi:10.1016/j.immuni.2017.02.011

    64. Ning X, Wang Y, Jing M, et al. Apoptotic caspases suppress type i interferon production via the cleavage of cGAS, MAVS, and IRF3. Mol Cell. 2019;74(1):19–31.e17. doi:10.1016/j.molcel.2019.02.013

    65. Kayagaki N, Warming S, Lamkanfi M, et al. Non-canonical inflammasome activation targets caspase-11. Nature. 2011;479(7371):117–121. doi:10.1038/nature10558

    66. Shi J, Zhao Y, Wang Y, et al. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature. 2014;514(7521):187–192. doi:10.1038/nature13683

    67. White MJ, McArthur K, Metcalf D, et al. Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell. 2014;159(7):1549–1562. doi:10.1016/j.cell.2014.11.036

    68. Nakhaei P, Sun Q, Solis M, et al. IκB kinase ε-dependent phosphorylation and degradation of X-linked inhibitor of apoptosis sensitizes cells to virus-induced apoptosis. J Virol. 2012;86(2):726–737. doi:10.1128/JVI.05989-11

    69. Hsu SK, Li CY, Lin IL, et al. Inflammation-related pyroptosis, a novel programmed cell death pathway, and its crosstalk with immune therapy in cancer treatment. Theranostics. 2021;11(18):8813–8835. doi:10.7150/thno.62521

    70. Liu J, Zhou J, Luan Y, et al. cGAS-STING, inflammasomes and pyroptosis: an overview of crosstalk mechanism of activation and regulation. Cell Commun Signal. 2024;22(1):22. doi:10.1186/s12964-023-01466-w

    71. Liu D, Wu H, Wang C, et al. STING directly activates autophagy to tune the innate immune response. Cell Death Differ. 2019;26(9):1735–1749. doi:10.1038/s41418-018-0251-z

    72. Zhang X, Liu J, Wang H. The cGAS-STING-autophagy pathway: novel perspectives in neurotoxicity induced by manganese exposure. Environ Pollut. 2022;315:120412. doi:10.1016/j.envpol.2022.120412

    73. Lu Q, Chen Y, Li J, et al. Crosstalk between cGAS-STING pathway and autophagy in cancer immunity. Front Immunol. 2023;14:1139595. doi:10.3389/fimmu.2023.1139595

    74. Lu QB, Ding Y, Liu Y, et al. Metrnl ameliorates diabetic cardiomyopathy via inactivation of cGAS/STING signaling dependent on LKB1/AMPK/ULK1-mediated autophagy. J Adv Res. 2023;51:161–179. doi:10.1016/j.jare.2022.10.014

    75. Chen YH, Chen HH, Wang WJ, et al. TRABID inhibition activates cGAS/STING-mediated anti-tumor immunity through mitosis and autophagy dysregulation. Nat Commun. 2023;14(1):3050. doi:10.1038/s41467-023-38784-z

    76. Liu M, Li Y, Han S, et al. Activin A alleviates neuronal injury through inhibiting cGAS-STING-mediated autophagy in mice with ischemic stroke. J Cereb Blood Flow Metab. 2023;43(5):736–748. doi:10.1177/0271678X221147056

    77. Zhu H, Zhang R, Yi L, et al. UNC93B1 attenuates the cGAS-STING signaling pathway by targeting STING for autophagy-lysosome degradation. J Med Virol. 2022;94(9):4490–4501. doi:10.1002/jmv.27860

    78. Ren C, Jin J, Li C, et al. Metformin inactivates the cGAS-STING pathway through autophagy and suppresses senescence in nucleus pulposus cells. J Cell Sci. 2022;135(15). doi:10.1242/jcs.259738

    79. Zhang L, Wei X, Wang Z, et al. NF-κB activation enhances STING signaling by altering microtubule-mediated STING trafficking. Cell Rep. 2023;42(3):112185. doi:10.1016/j.celrep.2023.112185

    80. Li R, Huang W. Yes-associated protein and transcriptional coactivator with PDZ-binding motif in cardiovascular diseases. Int J Mol Sci. 2023;24(2):1666.

    81. Meng Z, Moroishi T, Guan KL. Mechanisms of hippo pathway regulation. Genes Dev. 2016;30(1):1–17. doi:10.1101/gad.274027.115

    82. LaCanna R, Liccardo D, Zhang P, et al. Yap/Taz regulate alveolar regeneration and resolution of lung inflammation. J Clin Invest. 2019;129(5):2107–2122. doi:10.1172/JCI125014

    83. Plouffe SW, Lin KC, Moore JL, et al. The hippo pathway effector proteins YAP and TAZ have both distinct and overlapping functions in the cell. J Biol Chem. 2018;293(28):11230–11240. doi:10.1074/jbc.RA118.002715

    84. Hao F. An overview of the crosstalk between YAP and cGAS-STING signaling in non-small cell lung cancer: it takes two to tango. Clin Transl Oncol. 2022;24(9):1661–1672. doi:10.1007/s12094-022-02826-7

    85. Thannickal VJ, Toews GB, White ES, et al. Mechanisms of pulmonary fibrosis. Annu Rev Med. 2004;55(1):395–417. doi:10.1146/annurev.med.55.091902.103810

    86. Liu GY, Budinger GRS, Dematte JE. Advances in the management of idiopathic pulmonary fibrosis and progressive pulmonary fibrosis. BMJ. 2022;377:e066354.

    87. Podolanczuk AJ, Thomson CC, Remy-Jardin M, et al. Idiopathic pulmonary fibrosis: state of the art for 2023. Eur Respir J. 2023;61(4):2200957. doi:10.1183/13993003.00957-2022

    88. Koudstaal T, Funke-Chambour M, Kreuter M, et al. Pulmonary fibrosis: from pathogenesis to clinical decision-making. Trends Mol Med. 2023;29(12):1076–1087. doi:10.1016/j.molmed.2023.08.010

    89. Tsukui T, Wolters PJ, Sheppard D. Alveolar fibroblast lineage orchestrates lung inflammation and fibrosis. Nature. 2024;631(8021):627–634.

    90. Savin IA, Zenkova MA, Sen’kova AV. Pulmonary fibrosis as a result of acute lung inflammation: molecular mechanisms, relevant in vivo models, prognostic and therapeutic approaches. Int J Mol Sci. 2022;23(23):14959.

    91. Gifford AH, Matsuoka M, Ghoda LY, et al. Chronic inflammation and lung fibrosis: pleotropic syndromes but limited distinct phenotypes. Mucosal Immunol. 2012;5(5):480–484. doi:10.1038/mi.2012.68

    92. Zhang J, Du J, Liu D, et al. Polystyrene microplastics induce pulmonary fibrosis by promoting alveolar epithelial cell ferroptosis through cGAS/STING signaling. Ecotoxicol Environ Saf. 2024;277:116357. doi:10.1016/j.ecoenv.2024.116357

    93. Wu X, Jiang Y, Li R, et al. Ficolin B secreted by alveolar macrophage exosomes exacerbates bleomycin-induced lung injury via ferroptosis through the cGAS-STING signaling pathway. Cell Death Dis. 2023;14(8):577. doi:10.1038/s41419-023-06104-4

    94. Han B, Wang X, Wu P, et al. Pulmonary inflammatory and fibrogenic response induced by graphitized multi-walled carbon nanotube involved in cGAS-STING signaling pathway. J Hazard Mater. 2021;417:125984. doi:10.1016/j.jhazmat.2021.125984

    95. Zhou Q, Chang M, Guo S, et al. Honokiol ameliorates silica-induced lung fibrosis by inhibiting macrophage pyroptosis via modulating cGAS/STING signaling. Int Immunopharmacol. 2024;146:113812. doi:10.1016/j.intimp.2024.113812

    96. Xie X, Wu X, Zhao D, et al. Fluvoxamine alleviates bleomycin-induced lung fibrosis via regulating the cGAS-STING pathway. Pharmacol Res. 2023;187:106577. doi:10.1016/j.phrs.2022.106577

    97. Savigny F, Schricke C, Lacerda-Queiroz N, et al. Protective role of the nucleic acid sensor STING in pulmonary fibrosis. Front Immunol. 2020;11:588799. doi:10.3389/fimmu.2020.588799

    98. Nastase MV, Zeng-Brouwers J, Wygrecka M, et al. Targeting renal fibrosis: mechanisms and drug delivery systems. Adv Drug Deliv Rev. 2018;129:295–307. doi:10.1016/j.addr.2017.12.019

    99. Huang S, Lu H, Chen J, et al. Advances in drug delivery-based therapeutic strategies for renal fibrosis treatment. J Mater Chem B. 2024;12(27):6532–6549. doi:10.1039/D4TB00737A

    100. Davison SN, Pommer W, Brown MA, et al. Conservative kidney management and kidney supportive care: core components of integrated care for people with kidney failure. Kidney Int. 2024;105(1):35–45. doi:10.1016/j.kint.2023.10.001

    101. Liu Y. Renal fibrosis: new insights into the pathogenesis and therapeutics. Kidney Int. 2006;69(2):213–217. doi:10.1038/sj.ki.5000054

    102. Huang R, Fu P, Ma L. Kidney fibrosis: from mechanisms to therapeutic medicines. Signal Transduct Target Ther. 2023;8(1):129. doi:10.1038/s41392-023-01379-7

    103. Meng XM, Nikolic-Paterson DJ, Lan HY. Inflammatory processes in renal fibrosis. Nat Rev Nephrol. 2014;10(9):493–503. doi:10.1038/nrneph.2014.114

    104. Jiang A, Liu J, Wang Y, et al. cGAS-STING signaling pathway promotes hypoxia-induced renal fibrosis by regulating PFKFB3-mediated glycolysis. Free Radic Biol Med. 2023;208:516–529. doi:10.1016/j.freeradbiomed.2023.09.011

    105. Tsai YC, Hsieh TH, Liao YR, et al. METTL3-mediated N 6 -methyladenosine mRNA modification and cGAS-STING pathway activity in kidney fibrosis. J Am Soc Nephrol. 2024;35(10):1312–1329. doi:10.1681/ASN.0000000000000428

    106. Chung KW, Dhillon P, Huang S, et al. Mitochondrial damage and activation of the STING pathway lead to renal inflammation and fibrosis. Cell Metab. 2019;30(4):784–799.e785. doi:10.1016/j.cmet.2019.08.003

    107. Nanto-Hara F, Yamazaki M, Murakami H, et al. Chronic heat stress induces renal fibrosis and mitochondrial dysfunction in laying hens. J Anim Sci Biotechnol. 2023;14(1):81. doi:10.1186/s40104-023-00878-5

    108. Wang D, Li Y, Li G, et al. Inhibition of PKC-δ retards kidney fibrosis via inhibiting cGAS-STING signaling pathway in mice. Cell Death Discov. 2024;10(1):314. doi:10.1038/s41420-024-02087-z

    109. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol. 2021;18(3):151–166. doi:10.1038/s41575-020-00372-7

    110. Parola M, Pinzani M. Liver fibrosis: pathophysiology, pathogenetic targets and clinical issues. Mol Aspects Med. 2019;65:37–55. doi:10.1016/j.mam.2018.09.002

    111. Hernandez-Gea V, Friedman SL. Pathogenesis of liver fibrosis. Annu Rev Pathol. 2011;6(1):425–456. doi:10.1146/annurev-pathol-011110-130246

    112. Odenwald MA, Paul S. Viral hepatitis: past, present, and future. World J Gastroenterol. 2022;28(14):1405–1429. doi:10.3748/wjg.v28.i14.1405

    113. Russell RI. Hepatitis. Scott Med J. 1970;15(9):331–338. doi:10.1177/003693307001500905

    114. Suriawinata AA, Thung SN. Acute and chronic hepatitis. Semin Diagn Pathol. 2006;23(3–4):132–148. doi:10.1053/j.semdp.2006.11.001

    115. Luo S, Luo R, Lu H, et al. Activation of cGAS-STING signaling pathway promotes liver fibrosis and hepatic sinusoidal microthrombosis. Int Immunopharmacol. 2023;125(Pt B):111132. doi:10.1016/j.intimp.2023.111132

    116. Yong H, Wang S, Song F. Activation of cGAS/STING pathway upon TDP-43-mediated mitochondrial injury may be involved in the pathogenesis of liver fibrosis. Liver Int. 2021;41(8):1969–1971. doi:10.1111/liv.14895

    117. Arumugam S, Li B, Boodapati SLT, et al. Mitochondrial DNA and the STING pathway are required for hepatic stellate cell activation. Hepatology. 2023;78(5):1448–1461. doi:10.1097/HEP.0000000000000388

    118. Shen R, Yang K, Cheng X, et al. Accumulation of polystyrene microplastics induces liver fibrosis by activating cGAS/STING pathway. Environ Pollut. 2022;300:118986. doi:10.1016/j.envpol.2022.118986

    119. Zhang X, Du J, Huo S, et al. Hexafluoropropylene oxide trimer acid causes fibrosis in mice liver via mitochondrial ROS/cGAS-STING/NLRP3-mediated pyroptosis. Food Chem Toxicol. 2023;174:113706. doi:10.1016/j.fct.2023.113706

    120. Wang Q, Bu Q, Liu M, et al. XBP1-mediated activation of the STING signalling pathway in macrophages contributes to liver fibrosis progression. JHEP Rep. 2022;4(11):100555. doi:10.1016/j.jhepr.2022.100555

    121. Sun Y, Weng J, Chen X, et al. Oroxylin A activates ferritinophagy to induce hepatic stellate cell senescence against hepatic fibrosis by regulating cGAS-STING pathway. Biomed Pharmacother. 2023;162:114653. doi:10.1016/j.biopha.2023.114653

    122. Wu Q, Leng X, Zhang Q, et al. IRF3 activates RB to authorize cGAS-STING-induced senescence and mitigate liver fibrosis. Sci Adv. 2024;10(9):eadj2102. doi:10.1126/sciadv.adj2102

    123. Czubryt MP, Hale TM. Cardiac fibrosis: pathobiology and therapeutic targets. Cell Signal. 2021;85:110066. doi:10.1016/j.cellsig.2021.110066

    124. Kong P, Christia P, Frangogiannis NG. The pathogenesis of cardiac fibrosis. Cell Mol Life Sci. 2014;71(4):549–574. doi:10.1007/s00018-013-1349-6

    125. Lasrado N, Yalaka B, Reddy J. Triggers of inflammatory heart disease. Front Cell Dev Biol. 2020;8:192. doi:10.3389/fcell.2020.00192

    126. Hu D, Cui YX, Wu MY, et al. Cytosolic DNA sensor cGAS plays an essential pathogenetic role in pressure overload-induced heart failure. Am J Physiol Heart Circ Physiol. 2020;318(6):H1525–h1537. doi:10.1152/ajpheart.00097.2020

    127. Zhang C, Hao H, Wang Y, et al. Intercellular mitochondrial component transfer triggers ischemic cardiac fibrosis. Sci Bull. 2023;68(16):1784–1799. doi:10.1016/j.scib.2023.07.030

    128. Rech L, Abdellatif M, Pöttler M, et al. Small molecule STING inhibition improves myocardial infarction remodeling. Life Sci. 2022;291:120263. doi:10.1016/j.lfs.2021.120263

    129. Hu S, Gao Y, Gao R, et al. The selective STING inhibitor H-151 preserves myocardial function and ameliorates cardiac fibrosis in murine myocardial infarction. Int Immunopharmacol. 2022;107:108658. doi:10.1016/j.intimp.2022.108658

    130. Mizutani Y, Kanbe A, Ito H, et al. Activation of STING signaling accelerates skin wound healing. J Dermatol Sci. 2020;97(1):21–29. doi:10.1016/j.jdermsci.2019.11.008

    131. Watanabe T, Baker Frost DA, Mlakar L, et al. A human skin model recapitulates systemic sclerosis dermal fibrosis and identifies COL22A1 as a TGFβ early response gene that mediates fibroblast to myofibroblast transition. Genes. 2019;10(2):75. doi:10.3390/genes10020075

    132. Shook BA, Wasko RR, Rivera-Gonzalez GC, et al. Myofibroblast proliferation and heterogeneity are supported by macrophages during skin repair. Science. 2018;362(6417). doi:10.1126/science.aar2971

    133. Rosendahl AH, Schönborn K, Krieg T. Pathophysiology of systemic sclerosis (scleroderma). Kaohsiung J Med Sci. 2022;38(3):187–195. doi:10.1002/kjm2.12505

    134. Paul S, Kaplan MH, Khanna D, et al. Centromere defects, chromosome instability, and cGAS-STING activation in systemic sclerosis. Nat Commun. 2022;13(1):7074. doi:10.1038/s41467-022-34775-8

    135. Jeschke MG, Wood FM, Middelkoop E, et al. Scars. Nat Rev Dis Primers. 2023;9(1):64. doi:10.1038/s41572-023-00474-x

    136. Ogawa R. Keloid and hypertrophic scars are the result of chronic inflammation in the reticular dermis. Int J Mol Sci. 2017;18(3):606. doi:10.3390/ijms18030606

    137. Hong YK, Chang YH, Lin YC, et al. Inflammation in wound healing and pathological scarring. Adv Wound Care. 2023;12(5):288–300. doi:10.1089/wound.2021.0161

    138. Zhao W, Zhang H, Liu R, et al. Advances in immunomodulatory mechanisms of mesenchymal stem cells-derived exosome on immune cells in scar formation. Int J Nanomedicine. 2023;18:3643–3662. doi:10.2147/IJN.S412717

    139. Han C, Qian X, Ren X, et al. Inhibition of cGAS in paraventricular nucleus attenuates hypertensive heart injury via regulating microglial autophagy. Mol Neurobiol. 2022;59(11):7006–7024. doi:10.1007/s12035-022-02994-1

    140. Ren G, Lv W, Ding Y, et al. Ginseng saponin metabolite 20(S)-protopanaxadiol relieves pulmonary fibrosis by multiple-targets signaling pathways. J Ginseng Res. 2023;47(4):543–551. doi:10.1016/j.jgr.2023.01.002

    141. Shi W, Hao J, Wu Y, et al. Protective effects of heterophyllin B against bleomycin-induced pulmonary fibrosis in mice via AMPK activation. Eur J Pharmacol. 2022;921:174825. doi:10.1016/j.ejphar.2022.174825

    142. Deng J, He Y, Sun G, et al. Tanreqing injection protects against bleomycin-induced pulmonary fibrosis via inhibiting STING-mediated endoplasmic reticulum stress signaling pathway. J Ethnopharmacol. 2023;305:116071. doi:10.1016/j.jep.2022.116071

    143. Sun SC, Han R, Hou SS, et al. Juglanin alleviates bleomycin-induced lung injury by suppressing inflammation and fibrosis via targeting sting signaling. Biomed Pharmacother. 2020;127:110119. doi:10.1016/j.biopha.2020.110119

    144. Wang Y, He X, Wang H, et al. Qingfei xieding prescription ameliorates mitochondrial DNA-initiated inflammation in bleomycin-induced pulmonary fibrosis through activating autophagy. J Ethnopharmacol. 2024;325:117820. doi:10.1016/j.jep.2024.117820

    145. Haag SM, Gulen MF, Reymond L, et al. Targeting STING with covalent small-molecule inhibitors. Nature. 2018;559(7713):269–273. doi:10.1038/s41586-018-0287-8

    146. Mack M. Inflammation and fibrosis. Matrix Biol. 2018;68-69:106–121. doi:10.1016/j.matbio.2017.11.010

    147. Li Q, Wu P, Du Q, et al. cGAS-STING, an important signaling pathway in diseases and their therapy. MedComm. 2024;5(4):e511. doi:10.1002/mco2.511

    148. Decout A, Katz JD, Venkatraman S, et al. The cGAS-STING pathway as a therapeutic target in inflammatory diseases. Nat Rev Immunol. 2021;21(9):548–569. doi:10.1038/s41577-021-00524-z

    149. Liu Y, Pu F. Updated roles of cGAS-STING signaling in autoimmune diseases. Front Immunol. 2023;14:1254915. doi:10.3389/fimmu.2023.1254915

    150. Zhang Y, Zou M, Wu H, et al. The cGAS-STING pathway drives neuroinflammation and neurodegeneration via cellular and molecular mechanisms in neurodegenerative diseases. Neurobiol Dis. 2024;202:106710. doi:10.1016/j.nbd.2024.106710

    151. Wei X, Jing J, Huang R, et al. QFAE-nB alleviates pulmonary fibrosis by inhibiting the STING pathway in mice. J Ethnopharmacol. 2024;319(Pt 2):117295. doi:10.1016/j.jep.2023.117295

    152. Knowles A, Glass DA. Keloids and Hypertrophic Scars. Dermatol Clin. 2023;41(3):509–517. doi:10.1016/j.det.2023.02.010

    Continue Reading

  • Research Progress of Ketamine in Neuropathic Pain Comorbid Depression

    Research Progress of Ketamine in Neuropathic Pain Comorbid Depression

    1Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China; 2Department of Anesthesiology, The Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China; 3Department of Pain Medicine, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China; 4Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, 563000, People’s Republic of China; 5Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, 563000, People’s Republic of China

    Abstract: This review systematically summarizes the research progress of ketamine and its interaction with specific brain regions in the context of neuropathic pain comorbid with depression. As a non-competitive inhibitor of N-methyl-D-aspartate (NMDA) receptors, Ketamine exerts complex mechanisms and controversial topic. The structure and function of NMDA receptors, as well as the binding sites of ketamine, are summarized, and the hypothesis of disinhibition and neuroplasticity of ketamine’s antidepressant effect is elaborated. The mechanism of key brain regions, such as hippocampus and anterior cingulate cortex, is discussed in detail, and the antidepressant effect of ketamine is explored from the perspective of transcriptomics. Finally, this review integrates bioinformatics, molecular biology and other interdisciplinary approaches to elucidate the therapeutic effects and potential mechanisms of ketamine in treating neuropathic pain and depression comorbidity, providing a comprehensive theoretical basis, new directions for subsequent research and novel insights for the clinical application of ketamine.

    Introduction

    According to the International Association for the Study of Pain, pain is defined as an unpleasant feeling and emotional experience that is associated with or close to real or potential tissue damage. Pain results from the interplay of psychological, emotional, behavioral, and social factors. Mere activity of sensory neurons and neural pathways does not constitute pain. Although pain typically serves an adaptive and protective role, its presence may also lead to negative consequences for physical function, mental health, and social well-being. Verbal description is only one mean of pain expressing, and challenges in linguistic communication do not negate the presence of pain experience in individuals or animals.1,2

    Neuropathic pain (NP) is a type of chronic pain caused by damage to the somatosensory nervous system or harmful stimuli, and may be directly caused by an underlying disease. It is clinically manifested in the form of allodynia, hyperalgesia, and spontaneous pain. The mechanism of NP involves peripheral and central processes. The peripheral mechanisms include abnormal discharges from damaged peripheral sensory nerve fibers, discharges induced by aberrant neuronal interactions, and sympathetic nervous system hyperexcitability. The central mechanisms include the persistence and recurrence of pain that typically arises above the spinal cord.3,4

    Epidemiological studies show that the proportion of chronic pain with neuropathic characteristics is about 7%–10%. Chronic neuropathic pain is more common in females (8% vs 5.7% in males) and people above 50 (8.9% vs 5.6% in those under 49). Pain most frequently affects the lower back and limbs, including both the lower and upper extremities. Conventional pharmacotherapy relieves only 30%–40% of NP patients.5

    The pain-depression dyad (PDD) describes a pathological condition characterized by the co-occurrence of pain and depression in the same individual. Studies have indicated that the prolonged transmission of nociceptive signals leads to structural and functional plastic changes in higher brain centers, such as the prefrontal cortex (PFC),6 anterior cingulate cortex (ACC),7 amygdala,8 and hippocampus.9 These areas are also involved in regulating emotional and cognitive functions, suggesting that pain signals in higher brain centers encompass not only nociceptive signals but also emotion-related components.10 Clinical research has further highlighted that patients with neuropathic pain often exhibit mood disorders, including anxiety and depression, while patients with anxiety and depression are frequently accompanied by hypersensitivity to pain and hyperalgesia.11 This exacerbates the vicious cycle between pain and emotional disorders. This comorbidity not only intensifies the patient’s suffering but also poses significant challenges for treatment. However, the underlying mechanisms of this comorbidity remain under investigation. Therefore, identifying an appropriate entry point to explore the potential mechanisms between pain and depression could help in better understanding neuropathic pain.

    Ketamine, an NMDA receptor antagonist, exhibits potent analgesia and rapid-durable antidepressant effects. Low-dose ketamine induces such effects in both human patients and animal models.12,13 A clinical study14 has shown that six ketamine infusions yielded response and remission rates of 68.0% and 50.5%, respectively. The rapid, robust antidepressant and antisuicidal effects observed within four hours of infusion were sustained. Literature15 showed that ketamine will exhibit greater tolerability and safety in comparison to Electroconvulsive therapy (ECT). However, ketamine’s mechanisms in NP with comorbid anxiety and depression remain unclear. While current research focuses mainly on NMDA receptors, its effects via other mechanisms and brain regional roles in its treatment need further study. In conclusion, advancements in understanding ketamine’s application in NP with concurrent anxiety and depression are highly significant for clinical practice and research. A more thorough comprehension of ketamine’s mechanisms could result in the development of more effective treatment strategies, thereby enhancing patients’ quality of life.

    In recent years, bioinformatics has become an invaluable tool in the treatment of a wide range of diseases. Through the analysis of extensive genomic and transcriptomic data, bioinformatics methods can uncover new biomarkers and therapeutic targets, facilitating precise medical interventions. Additionally, these techniques enable the extraction of valuable insights from large-scale biological datasets, offering direction for future experimental designs.16

    Objective

    Ketamine has complex mechanisms, and previous studies have suggested that ketamine may improve depressive symptoms by enhancing the expression of brain-derived neurotrophic factor (BDNF), which is beneficial for the growth and connection of neurons.17 Research has also shown that ketamine can regulate the levels of monoamine neurotransmitters in the brain, such as dopamine, serotonin, and norepinephrine, thereby alleviating depressive symptoms.18 Additionally, ketamine may exert its therapeutic effects by modulating inflammatory responses and oxidative stress in the brain.19 Specific brain regions likely play significant roles in the treatment of NP comorbid with anxiety and depression through ketamine. Studies indicate that the lateral habenula (LHb), often referred to as the “anti-reward center” may be involved, where ketamine might act on NMDA receptors in this region to reduce cluster discharges, release inhibition on the reward center, and produce rapid antidepressant effects.20 Furthermore, regions such as the hippocampus and cortex may also contribute to ketamine’s antidepressant effects.21 However, the precise mechanisms of these brain regions in ketamine’s therapeutic action remain unclear.

    This review aims to elucidate the mechanisms of ketamine and its interaction with specific brain regions in neuropathic pain comorbid with anxiety and depression, further research is needed. On one hand, animal and cellular experiments can be conducted to explore ketamine’s impact on neuronal activity, neurotransmitter levels, inflammatory responses, and other factors in various brain regions, thereby revealing its mechanisms of action. From another perspective, clinical studies can evaluate the efficacy and safety of ketamine in regulating NP patients with anxiety and depression symptoms, and can also analyze functional changes in different brain regions, providing theoretical support for clinical application. In general, Understanding the role of ketamine and specific brain regions in neuropathic pain complicated with anxiety and depression is of key diagnostic and research value, which helps to formulate more effective treatment programs. This study aims to systematically dissect the antidepressant mechanisms of ketamine by integrating molecular, hypothesis level, brain region neurocircuits, regional specificity and transcriptomic perspectives.

    NMDA Receptor and Ketamine

    The NMDA receptor, an ionotropic glutamate receptor prevalent in the central nervous system (brain and spinal cord), features a complex structure permeable to K⁺, Na⁺, and Ca²⁺. A functional receptor requires the NR1 subunit, forming tetramers/pentamers with regulatory NR2 subunits. Different NR2 subtypes confer distinct regional distributions, physiological properties, and dual voltage- and ligand-gating mechanisms, rendering it a unique dual-gated channel.22 Mediating slow, Ca²⁺-permeable excitatory neurotransmission, NMDARs are implicated in neurodevelopmental, neuropsychiatric, neurological, and neurodegenerative disorders, making their modulation a therapeutic target for disease modification. During neurodevelopment, the receptors regulate neuronal survival, dendritic and axonal structural development, and participate in synaptic plasticity, playing a crucial role in the formation of neuronal circuits. They are critical receptors in learning and memory processes.

    Ketamine is an NMDA receptor antagonist. Although the pharmacological correlation with its rapid (within hours of administration) antidepressant effects remains unclear, this antidepressant action is thought to depend on mechanisms involving excitatory synaptic enhancement. Activation of synaptic NMDARs is essential for inducing typical long-term potentiation (LTP), which leads to sustained increases in synaptic strength. Studies have shown23 that through behavioral pharmacology, quantitative Western blotting of hippocampal synaptic proteins, and electrophysiological recordings from hippocampal slices, the hypothesis that rapid antidepressant effects require NMDAR activation has been validated. Fast-acting antidepressant compounds have a common downstream effect dependent on NMDAR activation, even though their initial pharmacological targets are not the same. Promoting the NMDAR signaling pathway or other strategies to enhance NMDAR dependent LTP-like synapses may be a useful antidepressant.

    “Disinhibition” Hypothesis

    The “disinhibition” hypothesis posits that ketamine’s ability to alleviate depression stems from its capacity to remove metaphorical “clouds” within the brain. By reducing the activity of overly active inhibitory systems—referred to as the brain’s “brakes”—ketamine helps to improve depressive symptoms. Specifically, at lower dosages, ketamine targets and blocks NMDA receptors located on GABAergic interneurons. This blockage decreases the inhibitory influence that these interneurons exert on glutamatergic pyramidal neurons, thereby promoting the release of glutamate (Figure 1A).

    Figure 1 Converging synaptic signaling pathways underlying ketamine action. (A) The antidepressant mechanism of ketamine predominantly depends on its antagonistic action on NMDARs located in GABAergic interneurons, blocking the release of GABA. By inhibiting the release of GABA, the inhibition of pyramidal glutamatergic neurons is averted. As a result, glutamate is released, leading to the subsequent downstream consequences triggered by a sudden increase in glutamate levels. Glutamate then binds to postsynaptic AMPARs, enabling calcium to flow in, results in the release of BDNF from the postsynaptic membrane, triggering TrkB receptor signaling. The TrkB pathway and resulting rapid homeostatic synaptic plasticity are crucial for both ketamine’s rapid and sustained actions. Activation of downstream mTOR causes structural plasticity, generating fast and long – lasting antidepressant effects. BDNF autocrine signaling directs downstream pathways like MAPK, PLC – γ, and PI3K – Akt. MAPK and PLC – γ are linked to synaptic plasticity, and PI3K – Akt to anti – apoptotic signaling and cell survival. (B) Ketamine is proposed to selectively block extrasynaptic GluN2B-containing NMDARs, which are tonically activated by low levels of ambient glutamate regulated by glutamate transporter 1 located on astrocytes. (C) The transient decrease in the excitatory drive of inhibitory interneuron-resident NMDARs mediated by ketamine is thought to inhibit the tonic release of GABA and relieve the activity of target excitatory neurons. The resulting increase in glutamatergic activity activates the downstream mammalian target of rapamycin (mTOR) function, leading to structural plasticity and producing rapid and sustained antidepressant effects. (D) Ketamine blocks NMDAR-mediated spontaneous neurotransmission, which inhibits eukaryotic elongation factor 2 kinase (eEF2K) activity, thus stopping the phosphorylation of its eEF2 substrate. (E) (2R,6R)-HNK increases AMPAR-dependent synaptic transmission. (F) A special firing pattern in the lateral habenula – burst firing is a sufficient condition for the occurrence of depression, and the effect of ketamine is to effectively prevent burst firing in this brain region. (G) T – VSCCs can serve as a novel antidepressant molecular target. (H) Another rapid antidepressant molecular target was revealed – the potassium ion channel Kir4.1 present in glial cells, which is crucial for triggering the burst firing of neurons. (I) The rapid elevation of NE by ketamine and the activation of astrocyte α1 – AR play an antidepressant role in sustained resilience. (J) Tiam1 links NMDARs stimulated by chronic pain to the activation of Rac1 in the ACC.

    This hypothesis focuses on glutamatergic neurotransmission, and it is thought that the effect of ketamine may be related to inhibiting gamma-Aminobutyric interneurons in the prefrontal cortex and hippocampus, then triggering downstream glutamatergic excitation and affecting synaptic plasticity. In mouse experiments,24 subanesthetic doses of ketamine rapidly increased glutamate content in the prefrontal cortex and hippocampus. This subanesthetic dose can enhance glutamate levels, but high anesthetic doses can cause a decrease in glutamate release, ketamine helps promote glutamate circulation in the PFC, and directly enhances glutamatergic neurotransmission in the medial prefrontal cortex (mPFC) and hippocampus. Inhibition of hippocampal GABAergic interneurons resulted in increased glutamate release,25,26 and this phenomenon in hippocampal CA1 region had a direct dose-dependent effect on BDNF surrender and TrkB receptor expression and translocation. These identifications demonstrated a causal relationship between synaptic glutamate increase and BDNF release, thereby supporting the disinhibition hypothesis.

    The mPFC, and its peak glutamate levels in the hippocampus are associated with postsynaptic alpha-amino-3-hydroxy-5-methyl-4-isoxazol-propionic acid receptors (AMPAR) activation, AMPARs are ionic glutamate receptors that are uniquely involved in rapid synaptic transmission, and are also involved in synaptic plasticity. In mouse models, Inhibition of GLT-1, a glutamate transporter in glial cells, can produce antidepressant effects similar to those of ketamine27 (Figure 1B); however, another study showed that inhibition of GLT-1 can hinder the antidepressant effects of ketamine and also affect downstream phosphorylation.28 This phenomenon may be caused by unbalanced glutamate circulation, which leads to hyperexcitability and excitotoxicity. Ketamine enhances synaptic transmission driven by AMPAR, especially in the hippocampus.29 The downstream response of AMPAR stimulation includes the release of BDNF, which interacts with the postsynaptic TrkB receptor. Current evidence suggests that even TrkB receptor antagonists can block the regulatory effects of ketamine.30 Some studies have shown that AMPAR activation is critical to ketamine’s depression-fighting effects. In a sample of mice with depression, AMPAR inhibition reduces or completely removes ketamine’s depression-fighting effects. Ketamine31 (10 mg/kg, i.p) produced a rapid (1 hour) anti-depression effect in mice with chronic adrenocorticotropin (ACTH) and chronic and unpredictable stress (CUMS) induced depression. These responses to depression are associated with the regular expression of glutamate transporter-1 (GLT-1), glial fibrillary acidic protein (GFAP), BDNF, and phosphorylated eukaryotic expansion factor 2 (p-eEF2) in PrL-PFC. Excitatory neurons in PrL are less responsive to peripheral glutamate synaptic stimulation.

    “Neuroplasticity” Hypothesis

    The “neuroplasticity” hypothesis suggests that antidepressant effects are due to the increase in substances or connections in the brain that promote feelings of happiness. Ketamine is believed to trigger the production of substances that support neuronal growth and synapse formation. Research has shown32 that the glutamate system and structural plasticity hypothesis are central to the rapid and long-lasting antidepressant effects of new antidepressants. Hippocampal plasticity is one of the important mechanisms for the sustained antidepressant effect of ketamine. Ketamine may improve depressive symptoms by increasing the expression of BDNF, which in turn promotes neuronal growth and association. A single dose of ketamine dramatically increases synaptic function and the number of pyramidal cells in the prefrontal cortex, rapidly changing the synapses lost by these neurons due to chronic stress. Ketamine33 enhances the synthesis and transport of AMPAR by activating the signaling pathway of BDNF and its receptor TrkB, thereby enhancing the excitability of synaptic transmission. It may improve depression by increasing the expression of neurotrophic factor BDNF in the brain to promote the growth and cohesion of neurons. The autocrine signaling of BDNF34 relies on MAPK, PLC-γ and PI3K-Akt signaling pathways to lead the downstream signaling pathway. The MAPK and PLC-γ pathways are associated with synaptic plasticity, the PI3K-Akt pathways are associated with anti-apoptotic signaling and cell survival, and the signals conveyed by mTORC1 are also associated with synaptic plasticity and neurogenesis (Figure 1A).

    Duman’s lab has done research, which shows that the action of ketamine involves blocking the action of NMDARs on inhibitory interneurons.35,36 For NMDARs expressed in these inhibitory neurons, their excitatory drive is temporarily weakened, and people feel that GABA tonic release is inhibited. The inhibitory activity of target excitatory neurons is stimulated, the glutamatergic activity is improved, the function of downstream mammalian target protein of rapamycin (mTOR) is activated, the formation of dendritic spines is increased, and the rapid and lasting antidepressant effect is not strong.37,38 One-time administration of ketamine can rapidly improve the synaptic function of prefrontal cortex pyramidal neurons. Increase their numbers, and immediately adjust the synaptic loss that occurs in these neurons as a result of chronic stress (Figure 1C).

    Detailed interpretation39 of the synaptic signaling activity associated with the NMDA receptor by ketamine provides evidence that ketamine inhibits the tension-stimulated NMDA receptor, resulting in the blocking of these static calcium signals and the suppression of eEF2K activity (Figure 1D), leading to dephosphorylation of eEF2 and lifting restrictions on the synthesis of dendritic proteins, especially BDNF. Instead, BDNF activates the post-synaptic TrkB receptor to induce the addition of the AMPA receptor, which in turn creates a new type of synaptic improvement in the hippocampus, which is the basis for the rapid antidepressant effect. Research indicates40 that astrocytes play a role in the pathophysiological process of major depression and the efficacy of antidepressant drugs through the regulation of synaptic plasticity supports the hypothesis that astrocyte atrophy is beneficial to the pathophysiological process of depression. The morphological changes of astrocytes may be one of the ways that ketamine rapidly improves depressive symptoms. The findings show41 that ketamine has an antidepressant effect, and that synaptic plasticity through presynaptic promotion promotes fear of memory loss, which may give new ideas for the treatment of post-traumatic stress disorder (PTSD).

    AMPAR and Ketamine

    Studies42 have shown that AMPAR promote rapid excitatory synaptic transmission in the central nervous system, and changes in synaptic plasticity of AMPAR are considered to be the basis for the long-term antidepressant effects of ketamine. Although ketamine and (2R,6R)-HNK do not alter the content of GluA1 and GluA2 AMPAR subunits in hippocampus 1 hour after treatment, both of them increase these subunits 24 hours after treatment in mice. These findings indicate that, the maintenance of AMPAR-induced synaptic strengthening—mediated by (2R,6R)-HNK via enhancing glutamate yield and AMPAR expression to boost AMPAR-dependent synaptic transmission—underlies the compound’s persistent antidepressant effects. (Figure 1E). Evidence also shows that (2R,6R)-HNK modulates metabolic glutamate (mGlu) receptor signaling, stimulates mTOR and BDNF pathways, and enhances release of other neurotransmitters (serotonin, norepinephrine). The compound promotes structural plasticity via dendritic remodeling and influences additional processes—including inflammatory responses and mitochondrial function.

    The Effects of Ketamine on Specific Brain Regions (Figure 2)

    Lateral Habenula (LHb): A Key Brain Region in Ketamine’s Antidepressant Action

    Research43 for the first time revealed that a special firing pattern in the LHb – the burst firing – is a sufficient condition for the development of depression, and the effect of ketamine is to effectively prevent burst firing in this brain region.(Figure 1F) In addition to relying on NMDARs, the burst firing in the LHb also requires the hyperpolarization of the neuronal membrane potential and the coordinated action of low – voltage – sensitive T-type calcium channels (T-VSCCs). Locally blocking T-VSCCs in the LHb also produced rapid antidepressant effects. This finding indicates that T-VSCCs can serve as a novel antidepressant molecular target (Figure 1G). The research team led by Hu Hailan44 further explored the molecular mechanisms leading to the hyperpolarization of LHb neurons and the increase in burst firing activity, revealing another rapid antidepressant molecular target the potassium ion channel Kir4.1 present in glial cells, which is crucial for triggering the burst firing of neurons (Figure 1H). When the potassium ion channel Kir4.1 is highly expressed in astrocytes, the ions released by neurons into the extracellular space are cleared more rapidly, leading to the hyperpolarization of neurons and subsequently triggering burst firing. A variety of typical depression-like behaviors were also observed in relevant mouse models. Subsequently, the researchers used RNA interference or dominant – negative mutations to specifically reduce the expression level of Kir4.1 or block its function in the astrocytes of the LHb, and found that the depressive behaviors were alleviated. The above findings demonstrate a causal relationship between the high expression of this potassium ion channel in the LHb and the formation of depression, and clarify a new molecular mechanism of depression onset – that is, the interaction between LHb neurons and glial cells is altered, triggering the burst firing of neurons and ultimately mediating depressive behaviors.

    Figure 2 The effects of ketamine on various brain regions of rats with comorbid chronic pain and depression. Adapted from Borsellino P, Krider RI, Chea D, et al. Ketamine and the Disinhibition Hypothesis: neurotrophic Factor-Mediated Treatment of Depression. J Pharmaceuticals. 2023;16(5):742. Under Creative Commons License https://creativecommons.org/licenses/by/4.0/.34

    Abbreviations: ACC, anterior cingulate cortex; HPC, hippocampus; LHb, lateral habenula; mPFC, medial prefrontal cortex; LC, locus coeruleus.

    Studies have shown that in models of depression in rats and mice, blocking the explosive activity of the “anti-reward center” (LHb) on which NMDA relies can produce the rapid antidepressant effect of ketamine. In animals with similar depression, LHb neurons show a large increase in explosive activity, and this phenomenon is altered by ketamine. The understanding that light stimulation triggers LHb explosion, which leads to the behavioral characteristics of despair and anhedonia, suggests a basic model in which ketamine alleviates the inhibition of the downstream monoaminergic reward center by blocking the NMDA-dependent explosion of LHb neurons, thereby improving mood at a very rapid rate. This gives a framework for developing new, fast-acting antidepressants. Research45 shows that the LHb is a brain region of great significance for the generation of stress and anxiety. Norepinephrine (NE) has always been associated with arousal, stress and anxiety, and it has been determined that the projection of NE to LHb comes from the locus coeruleus (LC). Current research results have established that the NE void inside LHb plays a role in arousal and anxiety. Calcium signaling in LHb astrocytes relies on α1A-adrenergic receptors and on a neural network between LHb and LC (Figure 2). LHb astrocytes mediate the second activation of local LHb neurons and the release of NE. Activating or inhibiting the calcium signals in LHb astrocytes respectively promoted or alleviated stress – induced depression – like behaviors. Research shows46 the crucial role of the rapid elevation of NE by ketamine and the activation of astrocyte α1 – AR in sustained resilience (Figure 1I), which may explain some alternative antidepressant interventions in rodent models and patients.

    Ketamine has fundamentally changed the approach to treating depression, offering not only rapid but also long-lasting antidepressant effects. While its half-life in mice is only 13 minutes, its antidepressant benefits can persist for over 24 hours, a discrepancy that presents an intriguing biological puzzle and carries significant clinical relevance. Research has shown46 that after a single systemic injection, ketamine continues to inhibit burst firing and block NMDARs in the LHb for up to a full day. This prolonged effect is not due to endocytosis, but instead arises from the use-dependent retention of ketamine within the NMDAR. By manipulating the interaction dynamics between ketamine and NMDARs at different plasma concentrations, we can control the duration of its antidepressant effects. These findings shed new light on the mechanisms responsible for ketamine’s sustained antidepressant action, and provide a promising avenue for enhancing its clinical application by regulating the length of its effects based on its biophysical interactions with NMDARs. Research by Professor Hu Hailan’s team at Zhejiang University has found that, once ketamine enters the depressed brain, it specifically targets the LHb, where NMDA receptors on LHb neurons serve as the initial target for ketamine’s effects. In the context of neuropathic pain comorbid with anxiety and depression, the LHb may also play a critical role in ketamine’s therapeutic effects. After administration of ketamine, neuronal activity in the LHb is initially suppressed, which may be a key step in alleviating anxiety and depressive symptoms. Furthermore, specific local knockout of NR1 (the NMDA receptor subunit) in the LHb of mice prevents the rapid antidepressant behavioral effects of ketamine, further confirming the crucial role of the LHb in ketamine’s antidepressant action.

    The influence of neural signaling on the LHb and depression: Under normal conditions, the LHb serves as the “anti-reward center” and its hyperactivity is linked to depressive-like behaviors. In the context of neuropathic pain, pain signals may exacerbate the abnormal activity in the LHb. Ketamine acts on the LHb, inhibiting the cluster firing of its neurons and thereby altering neural signal transmission. This not only alleviates depressive symptoms but may also impact comorbid anxiety symptoms. This mechanism of action could be a key pathway through which ketamine exerts its effects in cases of neuropathic pain comorbid with anxiety and depression.

    Hippocampus: Regulation of Neuroplasticity

    Ketamine is believed to promote neuroplasticity in the hippocampus. Studies have shown47 that even a single dose of ketamine can significantly improve anxiety-like symptoms induced by stress, possibly through modulation of the GSK-3β/GR signaling pathway to enhance hippocampal synaptic plasticity. Chronic pain can induce depression,48 severely affecting the patient’s life quality, although the underlying mechanisms remain unclear. Chronic neuropathic pain has been shown to regulate the DNA methylation of target genes associated with neuroplasticity and emotional regulation, a process induced by DNA methyltransferases (DNMTs). Methylation changes in the BDNF gene in the hippocampus are crucial for both neuropathic pain and depression. In the context of comorbid anxiety and depression associated with neuropathic pain, hippocampal neuroplasticity may be impaired. Ketamine can improve hippocampal neuroplasticity through the modulation of the glutamatergic system and upregulation of neurotrophic factor expression. This helps restore normal connections and function between neurons, alleviating anxiety and depressive symptoms. Research has found that ketamine treatment leads to changes in molecular events related to neuroplasticity in hippocampal neurons, such as alterations in the expression and phosphorylation levels of AMPA receptor subunits. These changes may represent a specific manifestation of ketamine’s effect on improving neuroplasticity. Studies have also shown that a single injection of ketamine selectively promotes neurogenesis in the ventral hippocampus of adult rats. Furthermore,49 the ventral dominance induction of GluN2B subunits of NMDARs, p-mTOR, GluA1 subunits of AMPARs, and BDNF in the hippocampus may form the basis of ketamine’s unique antidepressant effects.

    The hippocampus has extensive neural connections with other brain regions, including the prefrontal cortex and LHb. In cases of comorbid anxiety and depression associated with neuropathic pain, interactions between these brain regions may become dysregulated. Ketamine acts on the hippocampus and may modulate the neural activity of the entire brain by affecting its connections with other regions, thereby improving anxiety and depressive symptoms. Numerous studies have shown that BDNF is highly expressed in the hippocampus and is essential for processes such as neuronal growth, differentiation, regeneration, and maintenance of physiological functions. Chronic pain and depression are both associated with decreased levels of BDNF in the hippocampus.50 By pharmacologically inducing increased BDNF expression in the hippocampus, ketamine exerts both antidepressant and analgesic effects, and these actions may be linked to changes in other brain regions.

    Anterior Cingulate Cortex (ACC)

    The ACC51 is located on the medial surface of the cerebral hemisphere, above the corpus callosum, spanning its entire length, and includes subregions such as the subgenual, perigenual, and dorsal areas. The ACC is closely related to etiology, pathogenesis, and treatment of major depressive disorders.

    As an important component of the limbic system, the ACC has extensive fiber connections with many cortical and subcortical structures, and it plays a central role in regulating emotions, affect, motivation, and other functions. Animal studies52 have identified the ACC as a critical part of the medial pain system that mediates emotional responses. Additionally, the ACC receives nociceptive inputs from other pain-related cortical regions,53 such as the primary somatosensory cortex (S1) and the insular cortex. Not only does the ACC receive widespread afferent input, but its efferent fibers are also broadly distributed. Deep pyramidal cells project to many subcortical structures, including the hypothalamus and periaqueductal gray matter. Reports also indicate that deep pyramidal cells in the ACC send descending projections to the spinal cord. It can be inferred that these extensive fiber connections likely contribute to the complex role of the ACC in processing pain and associated emotional disorders.

    The Relationship Between the ACC and Chronic Pain-Related Anxiety and Depression

    ACC is a well-known region involved in processing and modulating the emotional components of pain. Several animal studies have observed the significance of excessive ACC activity in the context of chronic pain.54,55 Previous imaging studies56 have shown that patients with neuropathic pain exhibit excessive activation of the ACC. Additionally, optogenetic activation of the ACC in mice has been found to induce anxiety- and depression-like behaviors.57 When activated during chronic pain, the ACC serves as a key center for emotional disorders, making it an important target for understanding the underlying mechanisms of these conditions.

    Neurons in the ACC form bidirectional connections with the amygdala,58 which allows the ACC to receive input related to emotional fear and anxiety signals. This unique connectivity enables ACC neurons to integrate sensory input with anxiety signals. Chronic pain and anxiety may be mutually reinforcing, and the anterior cingulate cortex has an anatomical connection to the amygdala and other subcortical areas involved in emotional responses, which provides a theoretical basis for the anterior cingulate cortex’s response to anxiety and fear associated with painful stimuli or experiences.59 These findings support the idea that neuronal activity in the ACC can influence anxiety-related emotions. Research suggests58 that synaptic LTP in the presynaptic neurons of the ACC may be a synaptic mechanism underlying anxiety-like behaviors in the context of chronic pain. The presence of presynaptic LTP enhances the input from the thalamus to the ACC neurons involved in the chronic pain response, leading to pain-related anxiety. Additionally, the presence of postsynaptic LTP (post-LTP) results in an additive effect from both forms of LTP, which further promotes the interaction between chronic pain and anxiety. Under depressive conditions, the ACC can undergo functional and morphological changes, positioning it as a critical brain region in neuropathic pain-induced depression, closely related to the pathophysiology of depression.

    As a key processing hub in the limbic system, ACC receives nociceptive information projected from the thalamus60 and somatosensory cortex,61 as well as fear and anxiety-related signals from the amygdala.62 This distinctive characteristic enables ACC neurons to combine sensory inputs from pain signals with anxiety-related data, playing a crucial role in the processing of pain and the associated anxiety and depression-like behaviors. Clinical MRI studies have shown that patients with neuropathic pain and concomitant emotional dysfunction exhibit reduced gray matter volume in the ACC and enhanced hemodynamic signals.63,64 Furthermore, bilateral anterior cingulate corticectomy65 has been shown to alleviate both neuropathic pain and major depressive disorder in patients. In animal models of neuropathic pain, increased c-fos expression and synaptic plasticity in the ACC indicate abnormal activation of this region.66 Local silencing of the ACC or inhibition of ACC LTP67 has been demonstrated to effectively reduce neuropathic pain and its associated anxiety and depression-like behaviors.

    Both domestic and international studies have confirmed that the ACC is closely associated with pain and emotional disorders. However, the molecular mechanisms by which nociceptive signals activate the ACC, leading to synaptic plasticity changes, remain unclear. Studies have shown68 that a single dose of ketamine can persistently inhibit the overactivity of ACC neurons in chronic pain, and the antagonistic effect of NMDA receptors in ACC is beneficial to the inhibition of ketamine on aversive change in chronic pain. Seed stimulation analysis was used to explore the activation of subgenual anterior cingulate cortex (sgACC) dependent task. Assessment of inter-group differences and changes before and after convalescence,69 compared with the control group, patients with major depression showed higher sgACC activation levels for favorable and reverse monetary rewards, which were associated with anhedonia and anxiety, respectively, and major depressive disorder (MDD) patients showed higher functional connectivity of resting state between the hippocampus and sgACC. This was related to sgACC overactivation of favorable rewards, but not reverse rewards, and ultimately ketamine reduced sgACC overactivation of favorable rewards, not reverse rewards. These findings suggest a neural mechanism for ketamine’s antidepressant effects, namely the rapid reduction of abnormal sgACC’s hyperresponsiveness to positive incentives.

    Studies have shown12 that Tiam1 correlates chronic pain-stimulated NMDARs with Rac1 activation in ACC, which regulates synaptic structural plasticity through actin and spinal remodeling. Synaptic NMDAR stabilizes functional plasticity, which can lead to ACC overactivity and depression-like behavior (Figure 1J). Ketamine addresses depression-like behaviors associated with chronic pain by preventing maladaptive plasticity induced by tiam1 in the anterior cingulate cortex. Therefore, ketamine may promote its sustained antidepressant like effects by suppressing the structural and functional plasticity of synapses induced by Tiam1 in ACC neurons, which may be the basis for depression-like behaviors induced by chronic pain.

    Prefrontal Cortex (PFC)

    Glutamatergic signaling in the mPFC mediates ketamine-induced synaptic plasticity, which is critical for its rapid antidepressant effects. Subanesthetic amounts of ketamine can trigger abnormal glutamatergic explosion in mPFC, and inhibition of mPFC neurons can block the antidepressant effect of ketamine. More evidence of the critical significance of mPFC has been provided by photogenetic studies. It has been shown that light stimulation of pyramidal neurons expressing camk2a in mPFC can re-produce the rapid and long-lasting antidepressant behavior of ketamine.70 mPFC is a central hub that can shape activities in a distributed network of output structures, including stress-regulating behaviors and autonomic responses. Research71 suggests that the antidepressant effect of ketamine is due to the increase of glutamate in the medial prefrontal cortex, which stimulates the projection of the prefrontal lobe to the medial dorsal nucleus and locus coeruleus, thereby stimulating the release of serotonin and noradrenaline in the same region. The time frame for the effects of the two monoamines on the antidepressant response to ketamine appears to be different.

    Studies72 have shown that ketamine can rapidly improve the signaling within mPFC, even after ketamine is metabolized and cleaned, it still causes continuous morphological and physiological changes, subanesthetic doses of ketamine will increase the glutamate in mPFC, and then AMPA receptors are activated, BDNF is released, within 30 to 60 minutes. mTORC1 signaling is enhanced. Studies have also found73 that activation of specific neuronal types in the prefrontal cortex, such as DRD1-expressing neurons, can produce ketamine-like rapid antidepressant effects, suggesting that ketamine may interact with specific neuronal populations in the prefrontal cortex to exert its anxiolytic and antidepressant effects.

    Ketamine Antidepressant Effects: A Transcriptomic Perspective

    Transcriptomics, as a powerful research tool, provides new insights into understanding the mechanisms of ketamine in treating pain-related depression. A number of studies have suggested that transcriptomics plays a critical role in ketamine’s therapeutic effects for pain-related depression. Transcriptomics is the study of the complete set of transcripts in a specific cell or tissue at a particular time, offering valuable information about gene expression regulation mechanisms and biological processes. Studies74 explored the susceptibility of depressive-like behavior development under chronic pain conditions by identifying key genes or cellular mechanisms. The researchers used genome-wide RNA sequencing to detect transcriptomic signatures of the hippocampus, a region responsible for regulating mood and stress responses, in male mice that suffered from chronic inflammatory pain. Based on behavioral tests, pain-plagued animals were divided into two groups: “Tough” and “fragile”, as verified by RNA-seq bioinformatic interpretation and qPCR, hippocampus genes are implicated in neuroinflammation, cell delay/neurogenesis, and impaired blood-brain barrier integrity. Another study75 found that Sema4a was significantly upregulated in both male mice and humans under conditions of emotional changes, playing a crucial role in the onset of mood disorders. Overall, these results place the amygdala-cingulate pathway at the core of pain-depression comorbidity, highlighting the role of Sema4a and myelin damage in emotional regulation.

    According to the research results,76 KEGG analysis shows that cholinergic synapses and estrogen signaling pathways in the ACC play a key role in NP, and gradually more and more according to.77 The ACC region is enhanced by estrogen receptor-β / PKA and G protein-coupled estrogen receptor-1 / protein kinase B pathways that promote emotional distress, promote synaptic plasticity facilitated by NMDAR, and the cholinergic system78 modulates NP through restrained delivery of muscarinic M1 receptors facilitated by activation of GABA. In addition to calcium signaling, the GO analysis also focused on the activity of voltage-gated potassium channels (Kv). Gao79 found that the excessive movement of NP-related cingulate pyramidal neurons was related to the current reduction induced by Kv2, and the efficacy limit of Kv2 was able to reduce the excessive movement of nerves and produce analgesic consequences. It was also observed80 that there are many adhesion molecules and vesicles in the GO profile, and that synaptic adhesion molecules are important in the control of synaptic growth, neural circuits and behavior, and that in the NP case, activation of the anterior cingulate cortex changes the number of specific adhesion molecules. Nerve cell adhesion molecule-181 underpins behavioral sensitivities through spinal tissue and NMDAR-dependent LTP, Cav-1 plays a key role in synapse generation and plasticity. Yang82 demonstrated for the first time that Cav-1 in ACC neurons directly binds to N-methyl-D-aspartate receptor 2B subunit (NR2B) to improve the NR2B surface level in ACC, thereby stimulating ERK/CREB signaling channels and regulating chronic neuropathic pain.

    In the antidepressant process of ketamine, significant changes in the transcriptomics of the ACC have been observed. The transcriptomics of the ACC holds great potential as a therapeutic target. Through transcriptomic analysis, the molecular mechanisms of disease can be identified, and new drug targets can be discovered. In depression, dysfunction of the ACC is closely related to executive dysfunction and comorbid depression in chronic pain conditions. Therefore, intervening in the transcriptomic changes of the ACC may become a novel strategy for treating depression. Studies83 found that major depressive disorder is a disease associated with circadian rhythm disruption and a high suicide rate. Low-dose ketamine KT and sleep deprivation SD, two fast-acting antidepressants, can significantly reduce the depressive symptoms of patients within 24 hours. To solve this problem, we conducted a contrastive transcriptomic analysis to identify the candidate genes and associated channels shared by KT and SD. This investigation confirmed the potential efficacy of the biological clock in the rapid response of antidepressants. These distinctions may lead to new research directions that may be useful in planning chronopharmacological protocols for the regulation of major depression. In summary, the transcriptomics of the ACC provides new perspectives and potential therapeutic targets for the treatment of depression.

    Concluding Remarks

    In summary, ketamine holds significant research value and clinical importance in the treatment of neuropathic pain comorbid with anxiety and depression. A bibliometric analysis84 revealed that over the past two decades, research on ketamine and its enantiomers for antidepressant effects has surged, culminating in the approval of esketamine nasal spray for treatment-resistant depression. Ketamine’s rapid antidepressant action has prompted investigations into its mechanisms and the development of new antidepressants with reduced side effects. Future studies should focus on further investigating the mechanisms of ketamine’s action, developing safer and more effective novel antidepressants, and providing better treatment options for patients.

    In future research, it is crucial to explore the mechanisms of ketamine’s action in neuropathic pain comorbid with anxiety and depression. First, the specific relationships between ketamine and different brain regions need to be further clarified. Although the roles of the LHb and hippocampus in ketamine’s antidepressant effects are well-established, the mechanisms of action in other brain regions, including the PFC, ACC, and amygdala, as well as how they affect the functional connectivity between brain regions and the balance of neural circuits, remain to be further explored and discussed. To address these gaps, future studies should integrate functional MRI (fMRI) with viral tracing techniques to map both structural and functional connectivity of the PFC-ACC-amygdala circuit during ketamine treatment. Additionally, optogenetic or chemogenetic manipulation of specific neuronal populations in these regions will enable causal validation of how ketamine modulates neural circuit balance underlying its therapeutic effects.

    Secondly, research into the antidepressant mechanism of ketamine through ACC transcriptomics should continue, with a focus on more precisely understanding the relationship between ketamine and ACC transcriptomics. Although it is known that ketamine induces significant changes in ACC transcriptomics during the antidepressant process, the specific genes involved, how these genes interact, and how they influence the onset and development of depression remain unclear. Advanced transcriptomics technologies, such as single-cell RNA sequencing, can be employed for in-depth analysis of different cell types within the ACC to identify the specific cell types and gene targets affected by ketamine. Studies suggest that different cell types in the brain may play distinct roles in the onset and treatment of depression, and single-cell RNA sequencing can reveal ketamine’s effects on specific cell types, providing a basis for precision medicine.

    In conclusion, future research should focus on exploring ketamine’s mechanisms of action and clinical applications. Through interdisciplinary collaboration and innovation, more effective and safer treatment options can be provided for patients with neuropathic pain comorbid with anxiety and depression.

    Data Sharing Statement

    No datasets were generated or analyzed during the current study.

    Ethical Approval

    Given that this manuscript is a review, which does not entail any new experiments on human or animal subjects, the statement of ethical approval is not applicable.

    Acknowledgments

    The authors appreciate the support from the Guizhou Key Laboratory of Anesthesia and Organ Protection Zunyi Medical University.

    Author Contributions

    Conceptualization, QM L, SM L, XF L and BY Q; Study design, QM L, SM L; Execution, QM L, XF L; Analysis and Interpretation, QM L, SM L, XF L and BY Q; Writing-Original Draft, QM L and SM L; Writing-Review & Editing; QM L, XF L and BY Q; Funding Acquisition, BY Q, XF L. All authors contributed to the study conception and design. All authors edited and approved the final manuscript.

    Funding

    This work was supported by the National Natural Science Foundation of China (No. 82360233, 82360232), the Basic Research Plan of Guizhou Provincial (No. ZK[2024]307, qiankehejichu ZD[2025]055).

    Disclosure

    The authors report no conflicts of interest in this work.

    References

    1. IsHak WW, Wen RY, Naghdechi L, et al. Pain and Depression: a Systematic Review. J Harv Rev Psychiatry. 2018;26(6):352–363. doi:10.1097/HRP.0000000000000198

    2. Maallo AMS, Moulton EA, Sieberg CB, et al. A lateralized model of the pain-depression dyad. J Neurosci Biobehav Rev. 2021;127:876–883. doi:10.1016/j.neubiorev.2021.06.003

    3. Rosenberger DC, Blechschmidt V, Timmerman H, et al. Challenges of neuropathic pain: focus on diabetic neuropathy. J Neural Transm. 2020;127(4):589–624. doi:10.1007/s00702-020-02145-7

    4. Scholz J, Finnerup NB, Attal N, et al. The IASP classification of chronic pain for ICD-11: chronic neuropathic pain. J Pain. 2019;160(1):53–59. doi:10.1097/j.pain.0000000000001365

    5. Colloca L, Ludman T, Bouhassira D, et al. Neuropathic pain. J Nat Rev Dis Primers. 2017;3(1):17002. doi:10.1038/nrdp.2017.2

    6. Mecca C, Chao D, Yu G, et al. Dynamic Change of Endocannabinoid Signaling in the Medial Prefrontal Cortex Controls the Development of Depression after Neuropathic Pain. J Neurosci. 2021;41(35):7492–7508. doi:10.1523/JNEUROSCI.3135-20.2021

    7. Smith M, Asada N, Malenka R. Anterior cingulate inputs to nucleus accumbens control the social transfer of pain and analgesia. J Science. 2021;371(6525):153–159.

    8. Murugappan S, Xie L, Wong H, et al. Suppression of Pain in the Late Phase of Chronic Trigeminal Neuropathic Pain Failed to Rescue the Decision-Making Deficits in Rats. Int J Mol Sci. 2021;22(15):7846. doi:10.3390/ijms22157846

    9. Toledo R, Stein D, Stefani Sanches P, et al. Repetitive transcranial magnetic stimulation (rTMS) reverses the long-term memory impairment and the decrease of hippocampal interleukin-10 levels, both induced by neuropathic pain in rats. J Neurosci. 2021;472:51–59. doi:10.1016/j.neuroscience.2021.07.030

    10. De Ridder D, Adhia D, Vanneste S. The anatomy of pain and suffering in the brain and its clinical implications. J Neurosci Biobehav Rev. 2021;130:125–146.

    11. Corlier J, Tadayonnejad R, Wilson AC, et al. Repetitive transcranial magnetic stimulation treatment of major depressive disorder and comorbid chronic pain: response rates and neurophysiologic biomarkers. J Psychol Med. 2023;53(3):823–832. doi:10.1017/S0033291721002178

    12. Ru Q, Lu Y, Saifullah AB, et al. TIAM1-mediated synaptic plasticity underlies comorbid depression–like and ketamine antidepressant–like actions in chronic pain. J Clin Invest. 2022;132(24):e158545. doi:10.1172/JCI158545

    13. Humo M, Ayazgök B, lJ B, et al. Ketamine induces rapid and sustained antidepressant-like effects in chronic pain induced depression: role of MAPK signaling pathway. J Prog Neuropsychopharmacol Biol Psychiatry. 2020;8(100):10989.

    14. Zheng W, Zhou Y-L, Liu W-J, et al. Rapid and longer-term antidepressant effects of repeated-dose intravenous ketamine for patients with unipolar and bipolar depression. J Psychiatr Res. 2018;106:61–68. doi:10.1016/j.jpsychires.2018.09.013

    15. Huang X-B, Zheng W, et al. Ketamine and Electroconvulsive Therapy for Treatment-Refractory Depression. J Alpha Psychiatry. 2023;24(6):244–246. doi:10.5152/alphapsychiatry.2023.231358

    16. Shen Z, Bao N, Chen J, et al. Neuromolecular and behavioral effects of cannabidiol on depressive-associated behaviors and neuropathic pain conditions in mice. J Neuropharmacol. 2024;261:110153. doi:10.1016/j.neuropharm.2024.110153

    17. Chen D, Changming W, Xiangxin L, et al. Esketamine alleviates hippocampal neuronal injury in depressed rats by activating the CREB/BDNF signaling pathway [J]. Chin J Gerontol. 2024;44(2):433–437.

    18. Hess EM, Riggs LM, Michaelides M, et al. Mechanisms of ketamine and its metabolites as antidepressants. J Biochem Pharmacol. 2022;197:114892. doi:10.1016/j.bcp.2021.114892

    19. Zhang X, Cui Y, Song X, et al. Curcumin alleviates ketamine induced oxidative stress and apoptosis via Nrf2 signaling pathway in rats’ cerebral cortex and hippocampus. J Environ Toxicol. 2023;38(2):300–311. doi:10.1002/tox.23697

    20. Chen M, Ma S, Liu H, et al. Brain region-specific action of ketamine as a rapid antidepressant. J Science. 2024;385(6709):eado7010.

    21. Masuda FK, Aery Jones EA, Sun Y, et al. Ketamine evoked disruption of entorhinal and hippocampal spatial maps. J Nat Commun. 2023;14(1):6285. doi:10.1038/s41467-023-41750-4

    22. Ullman EZ, Perszyk RE, Paladugu S, et al. Mechanisms of Action Underlying Conductance-Modifying Positive Allosteric Modulators of the NMDA Receptor. J Mol Pharmacol. 2024;106(6):334–353. doi:10.1124/molpharm.124.001019

    23. Zanos P, Brown KA, Georgiou P, et al. NMDA Receptor Activation-Dependent Antidepressant-Relevant Behavioral and Synaptic Actions of Ketamine. J Neurosci. 2023;43(6):1038–1050. doi:10.1523/JNEUROSCI.1316-22.2022

    24. Chowdhury GMI, Zhang J, Thomas M, et al. Transiently Increased Glutamate Cycling in Rat PFC Is Associated with Rapid Onset of Antidepressant-like Effects. J Mol Psychiatry. 2017;22(1):120–126. doi:10.1038/mp.2016.34

    25. Grace Jang MBM, MacIver MB. Ketamine Produces a Long-Lasting Enhancement of CA1 Neuron Excitability. Int J Mol Sci. 2021;22(15):8091. doi:10.3390/ijms22158091

    26. Zhang B, Yang X, Luyu Y, et al. Ketamine Activated Glutamatergic Neurotransmission by GABAergic Disinhibition in the Medial Prefrontal Cortex. J Neuropharmacol. 2021;194:108382. doi:10.1016/j.neuropharm.2020.108382

    27. Pham TH, Defaix C, Nguyen TML, et al. Cortical and Raphe GABAA, AMPA Receptors and Glial GLT-1 Glutamate Transporter Contribute to the Sustained Antidepressant Activity of Ketamine. J Pharmacol Biochem Behav. 2020;192:172913. doi:10.1016/j.pbb.2020.172913

    28. Chen Y, Shen M, Liu X, et al. The Regulation of Glutamate Transporter 1 in the Rapid Antidepressant-Like Effect of Ketamine in Mice. J Front Behav Neurosci. 2022;2(16):789524. doi:10.3389/fnbeh.2022.789524

    29. Iskandrani KSE, Oosterhof CA, Mansari ME, et al. Impact of subanesthetic doses of ketamine on AMPA-mediated responses in rats: an in vivo electrophysiological study on monoaminergic and glutamatergic neurons. J Psychopharmacol. 2015;29(7):792–801. doi:10.1177/0269881115573809

    30. Youge Q, Shan J, Wang S, et al. Rapid-Acting and Long-Lasting Antidepressant-like Action of (R)-Ketamine in Nrf2 Knock-out Mice: a Role of TrkB Signaling. J Eur Arch Psychiatry Clin Neurosci. 2021;271(3):439–446.

    31. Xiaoyan M, Yang S, Zhang Z, et al. Rapid and Sustained Restoration of Astrocytic Functions by Ketamine in Depression Model Mice. J Biochem Biophys Res Commun. 2022;616:89–94. doi:10.1016/j.bbrc.2022.03.068

    32. Ardalan M, Wegener G, Polsinelli B, et al. Neurovascular plasticity of the hippocampus one week after a single dose of ketamine in genetic rat model of depression. J Hippocampus. 2016;26(11):1414–1423. doi:10.1002/hipo.22617

    33. Yang C, Yang J, Luo A, et al. Molecular and cellular mechanisms underlying the antidepressant effects of ketamine enantiomers and its metabolites. J Transl Psychiatry. 2019;9(1):280. doi:10.1038/s41398-019-0624-1

    34. Borsellino P, Krider RI, Chea D, et al. Ketamine and the Disinhibition Hypothesis: neurotrophic Factor-Mediated Treatment of Depression. J Pharmaceuticals. 2023;16(5):742. doi:10.3390/ph16050742

    35. Gerhard DM, Pothula S, Liu RJ, et al. GABA interneurons are the cellular trigger for ketamine’s rapid antidepressant actions. J Clin Invest. 2020;130(3):1336–1349. doi:10.1172/JCI130808

    36. Pothula S, Kato T, Liu RJ, et al. Cell-type specific modulation of NMDA receptors triggers antidepressant actions. J Mol Psychiatry. 2021;26(9):5097–5111. doi:10.1038/s41380-020-0796-3

    37. Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. J Science. 2010;329(5994):959–964.

    38. Moda-Sava RN, Murdock MH, Parekh PK, et al. Sustained rescue of prefrontal circuit dysfunction by antidepressant-induced spine formation. J Science. 2019;364(6436):eaat8078.

    39. Nosyreva E, Szabla K, Autry AE, et al. Acute suppression of spontaneous neurotransmission drives synaptic potentiation. J Neurosci. 2013;33(16):6990–7002. doi:10.1523/JNEUROSCI.4998-12.2013

    40. Ardalan M, Rafati AH, Nyengaard JR, et al. Rapid antidepressant effect of ketamine correlates with astroglial plasticity in the hippocampus. Br J Pharmacol. 2017;174(6):483–492. doi:10.1111/bph.13714

    41. Wei MD, Wang YH, Lu K, et al. Ketamine reverses the impaired fear memory extinction and accompanied depressive-like behaviors in adolescent mice. J Behav Brain Res. 2020;379:112342. doi:10.1016/j.bbr.2019.112342

    42. Zanos P, Moaddel R, Morris PJ, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolite. J Nature. 2016;533(7604):481–486. doi:10.1038/nature17998

    43. Yang Y, Cui Y, Sang K, et al. Ketamine blocks bursting in the lateral habenula to rapidly relieve depression. J Nature. 2018;554(7692):317–322. doi:10.1038/nature25509

    44. Ma S, Chen M, Jiang Y, et al. Sustained antidepressant effect of ketamine through NMDAR trapping in. the LHb J Nature. 2023;622(7984):802–809. doi:10.1038/s41586-023-06624-1

    45. Purvis EM, Klein AK, Ettenberg A. Lateral habenular norepinephrine contributes to states of arousal and anxiety in male rats. J Behav Brain Res. 2018;347:108–115. doi:10.1016/j.bbr.2018.03.012

    46. Duque M, Chen AB, Hsu E, et al. Ketamine induces plasticity in a nore-pinephrine-astroglial circuit to promote behavioral perseverance. J Neuron. 2025;113(3):426–443.e5. doi:10.1016/j.neuron.2024.11.011

    47. Zhu X, Zhang F, You Y, et al. S-Ketamine Exerts Antidepressant Effects by Regulating Rac1 GTPase Mediated Synaptic Plasticity in the Hippocampus of Stressed Rats. J Cell Mol Neurobiol. 2023;43(1):299–314. doi:10.1007/s10571-021-01180-6

    48. Liu R, Wu XM, He X, et al. Contribution of DNA methyltransferases to spared nerve injury induced depression partially through epigenetically repressing Bdnf in hippocampus: reversal by ketamine. J Pharmacol Biochem Behav. 2021;200:173079. doi:10.1016/j.pbb.2020.173079

    49. Yamada J, Jinno S. Potential link between antidepressant-like effects of ketamine and promotion of adult neurogenesis in the ventral hippocampus of mice. J Neuropharmacol. 2019;158:107710. doi:10.1016/j.neuropharm.2019.107710

    50. Huanli Z, Wei W. Research Progress on the Role of Hippocampus in the Pathogenesis of Depressive Disorder and Chronic Pain [J]. Chin J Modern Doctors. 2017;55(05):151–154.

    51. Alexander L, Jelen LA, Mehta MA, et al. The anterior cingulate cortex as a key locus of ketamine’s antidepressant action. J Neurosci Biobehav Rev. 2021;127:531–554. doi:10.1016/j.neubiorev.2021.05.003

    52. Zhao M, Wang Z, Weng Z, et al. Electroacupuncture improves IBS visceral hypersensitivity by inhibiting the activation of astrocytes in the medial thalamus and anterior cingulate cortex. J Evid Based Complement Alternat Med. 2020;12(2020):2562979. doi:10.1155/2020/2562979

    53. Kummer KK, Miodrag M, Kalpachidou T, et al. The medial prefrontal cortex as a central hub for mental comorbidities associated with chronic pain. Int J Mol Sci. 2020;21(10):3440. doi:10.3390/ijms21103440

    54. Zhang Q, Manders T, Tong AP, et al. Chronic pain induces generalized enhancement of aversion. J Elife. 2017;6:e25302. doi:10.7554/eLife.25302

    55. Sellmeijer J, Mathis V, Hugel S, et al. Hyperactivity of anterior cingulate cortex areas 24a/24b drives chronic pain-induced anxiodepressive-like consequences. J Neurosci. 2018;38(12):3102–3115. doi:10.1523/JNEUROSCI.3195-17.2018

    56. Malfliet A, Coppieters I, Wilgen PV, et al. Brain changes associated with cognitive and emotional factors in chronic pain: a systematic review. Eur J Pain. 2017;21(5):769–786. doi:10.1002/ejp.1003

    57. Barthas F, Sellmeijer J, Hugel S, et al. The anterior cingulate cortex is a critical hub for pain? Induced depression. J BiolPsychiatry. 2015;77(3):236–245.

    58. Zhuo M. Neural mechanisms underlying anxiety chronic pain interactions. J Trends Neurosci. 2016;39(3):136–145. doi:10.1016/j.tins.2016.01.006

    59. Bliss TV, Collingridge GL, Kaang BK, et al. Synaptic plasticity in the anterior cingulate cortex in acute and chronic pain. J Nat Rev Neurosci. 2016;17(8):485–496. doi:10.1038/nrn.2016.68

    60. Wang Y, Wang J, Xia S, et al. Neuropathic pain generates silent synapses in thalamic projection to anterior cingulate cortex. J Pain. 2021;162(5):1322–1333. doi:10.1097/j.pain.0000000000002149

    61. Mengual UM, Wybo WAM, Spierenburg LJE, et al. Efficient Low-Pass Dendro-Somatic Coupling in the Apical Dendrite of Layer 5 Pyramidal Neurons in the Anterior Cingulate Cortex. J Neurosci. 2020;40(46):8799–8815. doi:10.1523/JNEUROSCI.3028-19.2020

    62. Shao D, Cao Z, Fu Y, et al. Projection from the basolateral amygdala to the anterior cingulate cortex facilitates the consolidation of long-term withdrawal memory. J Addiction Biology. 2021;26(6):e13048. doi:10.1111/adb.13048

    63. McIlwrath SL, Montera MA, Gott KM, et al. Manganese-enhanced MRI reveals changes within brain anxiety and aversion circuitry in rats with chronic neuropathic pain and anxiety-like behaviors. J NeuroImage. 2020;223:117343. doi:10.1016/j.neuroimage.2020.117343

    64. Da Silva JT, Tricou C, Zhang Y, et al. Brain networks and endogenous pain inhibition are modulated by age and sex in healthy rats. J Pain. 2020;161(6):1371–1380. doi:10.1097/j.pain.0000000000001810

    65. Deng Z, Pan Y, Li D, et al. Effect of Bilateral Anterior Cingulotomy on Chronic Neuropathic Pain with Severe Depression. J World Neurosurgery. 2019;121:196–200. doi:10.1016/j.wneu.2018.10.008

    66. Zhu X, Tang H, Dong W, et al. Distinct thalamocortical circuits underlie allodynia induced by tissue injury and by depression-like states. J Nat Neurosci. 2021;24(4):542–553. doi:10.1038/s41593-021-00811-x

    67. Li X, Matsuura T, Xue M, et al. Oxytocin in the anterior cingulate cortex attenuates neuropathic pain and emotional anxiety by inhibiting presynaptic long-term potentiation. J Cell Rep. 2021;36(3):109411. doi:10.1016/j.celrep.2021.109411

    68. Zhou H, Zhang Q, Martinez E, et al. Ketamine reduces aversion in rodent pain models by suppressing hyperactivity of the anterior cingulate cortex. J Nat Commun. 2018;9(1):3751. doi:10.1038/s41467-018-06295-x

    69. Morris LS, Costi S, Tan A, et al. Ketamine normalizes subgenual cingulate cortex hyper-activity in depression. J Neuropsychopharmacol. 2020;45(6):975–981. doi:10.1038/s41386-019-0591-5

    70. Fuchikami M, Thomas A, Liu R, et al. Optogenetic stimulation of infralimbic PFC reproduces ketamine’s rapid and sustained antidepressant actions. J Proc Natl Acad Sci USA. 2015;112(26):8106–8111. doi:10.1073/pnas.1414728112

    71. López-Gil X, Jiménez-Sánchez L, Campa L, et al. Role of Serotonin and Noradrenaline in the Rapid Antidepressant Action of Ketamine. J ACS Chem Neurosci. 2019;10(7):3318–3326. doi:10.1021/acschemneuro.9b00288

    72. Hare BD, Ghosal S, Duman RS. Rapid acting antidepressants in chronic stress models: molecular and cellular mechanisms. J Chronic Stress. 2017;1:2470547017697317. doi:10.1177/2470547017697317

    73. Hare BD, Shinohara R, Liu RJ, et al. Optogenetic stimulation of medial prefrontal cortex Drd1 neurons produces rapid and long-lasting antidepressant effects. J Nat Commun. 2019;10(1):223. doi:10.1038/s41467-018-08168-9

    74. Garman A, Ash AM, Kokkinos EK, et al. Novel hippocampal genes involved in enhanced susceptibility to chronic pain-induced behavioral emotionality. Eur J Pharmacol. 2024;964:176273. doi:10.1016/j.ejphar.2023.176273

    75. Becker LJ, Fillinger C, Waegaert R, et al. The basolateral amygdala anterior cingulate pathway contributes to depression-like behaviors and comorbidity with chronic pain behaviors in male mice. J Nat Commun. 2023;14(1):2198. doi:10.1038/s41467-023-37878-y

    76. Qiu X-T, Guo C, Li-Tian M, et al. Transcriptomic and proteomic profiling of the anterior cingulate cortex in neuropathic pain model rats. J Front Mol Neurosci. 2023;16:1164426. doi:10.3389/fnmol.2023.1164426

    77. Zang KK, Xiao X, Chen LQ, et al. Distinct function of estrogen receptors in the rodent anterior cingulate cortex in pain related aversion. J Anesthesiol. 2020;133(1):165–184. doi:10.1097/ALN.0000000000003324

    78. Koga K, Matsuzaki Y, Migita K, et al. Stimulating muscarinic M (1) receptors in the anterior cingulate cortex reduces mechanical hypersensitivity via GABAergic transmission in nerve injury rats. J Brain Res. 2019;1704:187–195. doi:10.1016/j.brainres.2018.10.013

    79. Gao SH, Shen LL, Wen HZ, et al. Inhibition of metabotropic glutamate receptor subtype 1 alters the excitability of the commissural pyramidal neuron in the rat anterior cingulate cortex after chronic constriction injury to the sciatic nerve. J Anesthesiol. 2017;127(3):515–533. doi:10.1097/ALN.0000000000001654

    80. Kurshan PT, Shen K. Synaptogenic pathways. J Curr Opin Neurobiol. 2019;57:156–162. doi:10.1016/j.conb.2019.03.005

    81. Hyoung-GonKo J-HC, Choi J-H, Park DI, et al. Rapid Turnover of Cortical NCAM1 Regulates Synaptic Reorganization after Peripheral Nerve Injury. J Cell Rep. 2018;22(3):748–759. doi:10.1016/j.celrep.2017.12.059

    82. Yang J-X, Hua L, Yan-Qiang L, et al. Caveolin-1 in the anterior cingulate cortex modulates chronic neuropathic pain via regulation of NMDA receptor 2B subunit. J Neurosci. 2015;35(1):36–52. doi:10.1523/JNEUROSCI.1161-14.2015

    83. Orozco-Solis R, Montellier E, Aguilar-Arna L, et al. A Circadian Genomic Signature Common to Ketamine and Sleep Deprivation in the Anterior Cingulate Cortex. J Biol Psychiatry. 2017;82(5):351–360. doi:10.1016/j.biopsych.2017.02.1176

    84. Zhao L-Y, Zhang G-F, Lou X-J, et al. Ketamine and its enantiomers for depression: a bibliometric analysis from 2000 to 2023. J Eur Arch Psychiatry Clin Neurosci. 2024;2024:1.

    Continue Reading

  • Efficacy of Spesolimab in Treating Generalized Pustular Psoriasis with

    Efficacy of Spesolimab in Treating Generalized Pustular Psoriasis with

    Introduction

    Generalized pustular psoriasis (GPP) is a rare and chronic disease that can be life-threatening. It is marked by repeated episodes of painful redness, sterile pustules, and “lakes of pus”. These episodes may occur with or without systemic inflammation or plaque psoriasis.1 The clinical course of GPP is highly variable, occurring either de novo or triggered by various factors. Current guidelines for the treatment of GPP are lacking. Traditional systemic treatments, including corticosteroids, acitretin, cyclosporine, and methotrexate, are often used as first-line therapies for GPP. However, there is limited evidence supporting their effectiveness. Several biologics, including TNF inhibitors, IL-17/IL-17R inhibitors and IL-23 inhibitors, have been used for the treatment of GPP. However the utilization of various non-specific treatments for GPP usually results in only partial control of the disease. Recently, biologic agents that inhibit the IL-36 pathway have demonstrated efficacy and safety in patients with GPP.2 Spesolimab, an IL-36R antagonist, is currently the only approved treatment for generalized pustular psoriasis in China.1,2 Previous studies on these treatments have been limited, and their therapeutic effects across different genotypes of pustular psoriasis have not been adequately assessed. In these cases. In this case series, we evaluate the therapeutic efficacy of spesolimab in five GPP patients, focusing on the influence of IL-36RN gene mutations and the presence of concomitant plaque psoriasis.

    Case Report

    Patient 1 is a 28-year-old Chinese man who has had GPP since he was 3 years old. Genetic analysis confirmed a homozygous IL36RN mutation (c.115 + 6T > C). He experienced recurrent severe flares with fever, leading to multiple hospitalizations, despite receiving intermittent low-dose prednisolone and traditional Chinese medicine. For the past decade, he used only topical traditional Chinese medicine and steroid ointments, yet he still had episodes of pustular eruptions. One month before his hospital admission, a severe flare prompted him to seek treatment. Physical examination revealed extensive erythema, severe scaling, yellow pustular crusts on the scalp, trunk, and limbs, and dystrophic nails covered with pustules (Figure 1A1–3 and Table 1). Laboratory tests showed leukocytosis, elevated platelets (587 × 10⁹/L), ESR, CRP (40.03 mg/L), and IL-6 (56.24 pg/mL). GPPASI, GPPGA, and JDA-GPPSI scores3 were 51.6, 3, and 12, respectively. After administering a single intravenous dose of 900 mg spesolimab, the patient experienced a 51% reduction in GPPASI scores and a 33.3% reduction in GPPGA scores within one week. Yellow crusts shed within 12 hours, and nails improved significantly by 4 weeks. Only vitamin E ointment was used for moisturizing after discharge. Platelet levels returned to normal (269.5 × 109/L), with no new pustules observed at 4 months (Figure 1A4–6), and the patient has remained in complete remission for 12 months.

    Table 1 The Demographic and Clinical Characteristics of GPP Patients with or Without IL-36RN Gene Mutations and Coexisting Plaque Psoriasis

    Figure 1 Efficacy of Spesolimab in Treating GPP patients with or without Concomitant Plaque Psoriasis. The skin lesions of GPP patient 1, who did not have coexisting plaque psoriasis prior to treatment (A13), exhibited notable improvement four months after receiving spesolimab therapy (A46). In contrast, the skin lesions of GPP patient 5, who presented with coexisting plaque psoriasis before treatment (B13), displayed worsening of symptoms within one week of spesolimab administration (B46). Table 1 has been revised regarding the genetic mutations of Case 2 and Case 3. Case 1 (IL36RN homozygous mutation) and Case 2 (IL36RN/CARD14 compound mutations), Cases 3, 4, and 5 did not show IL-36RN gene mutations.

    Patient 2 is a 35-year-old Chinese woman with a 30-year history of intermittent erythema and pustules, recently experiencing a one-month uncontrolled flare of GPP. Previous treatments, including acitretin, cyclosporine, corticosteroids, and traditional Chinese medicine, provided only temporary relief. One month ago, extensive erythema and pustules reappeared on her trunk and limbs, accompanied by itching, pain, and bilateral knee joint pain, with poor response to cyclosporine. Physical examination revealed diffuse erythema and dense pustules on the face, trunk, and extremities (Table 1). Laboratory tests showed elevated cytokine levels (eg, IL-6596.73 pg/mL, IL-10 80.89 pg/mL, IL-8333.28 pg/mL) and CRP (150.43 mg/L). Genetic analysis identified IL36RN (c.227C>T, p.P76L) and multiple CARD14 mutations (eg, p.R547S, p.R820W, p.R883H). A single 900 mg intravenous dose of spesolimab was administered, leading to significant improvement within 10 hours as pustules subsided. One week post-treatment, the GPPASI and GPPGA scores were reduced by 83.2% and 66.7%, respectively. The patient has remained in complete remission for 4 months.

    Patient 3 is a 62-year-old Chinese man with a 40-year history of GPP unresponsive to various treatments. Symptoms improved with secukinumab since December 2022 but worsened after titanium dental implants, causing a high fever, widespread erythema, pustules, and swelling. Genetic testing revealed no mutations (Table 1), and laboratory tests showed elevated CRP (65.43 mg/L) and hypoalbuminemia (23 g/L). After immunoglobulin therapy, he received a single 900 mg dose of spesolimab, leading to significant improvement within 18 hours. One week later, GPPASI and GPPGA scores were reduced by 60% and 33.3%. The patient has remained in complete remission for 6 months.

    Patient 4, a 25-year-old female with a 3-year history of erythematous plaques and scales, developed recurrent pustules one month prior. She experienced recurrent fever. Treatment with prednisone, diphenhydramine, and cyclosporine was ineffective. The patient has a history of type I diabetes. Examination revealed widespread pustules, and pathology confirmed pustular psoriasis. Genetic testing showed no mutations, and cytokine levels were elevated (IL-17: 50.26 pg/mL, IL-8: 151.13 pg/mL) (Table 1). A single 900 mg dose of spesolimab showed slight improvement after one week but was unsatisfactory. Switched to ixekizumab in the second week, pustules resolved within one week, and no recurrence was observed during four months of follow-up.

    Patient 5 is a 44-year-old Chinese woman with a 10-year history of plaque psoriasis and psoriatic arthritis, effectively managed with secukinumab since January 2023 after unsuccessful treatments with adalimumab, cyclosporine, MTX, and topical therapies. Two months prior, she discontinued secukinumab for pregnancy preparation and developed uncontrolled GPP, with widespread pustules, erythema, severe edema, and a high fever of 40°C (Figure 1B1–3 and Table 1). Genetic testing showed no mutations, and cytokine levels were elevated (IL-17: 73.16 pg/mL, IL-8: 134.61 pg/mL, IL-6: 78.33 pg/mL (Table 1). A single 900 mg dose of spesolimab was administered, but one week later, symptoms persisted, including high fever and new pustules (Figure 1B4–6). Slight improvement was noted in pustules on the abdomen and upper limbs, but the patient refused a second injection. She was given 1 mL of betamethasone, resulting in rapid improvement within three days, with resolution of pustules and erythema. Fever subsided, and she was discharged after resuming secukinumab and topical treatments.

    Discussion

    The case reports presented herein highlight the significant challenges and successes in managing GPP, particularly in the context of varying IL36RN gene mutations. The importance of these cases lies not only in the individual patient experiences but also in their contributions to the broader understanding of GPP management, treatment efficacy, and the implications of genetic factors on clinical outcomes. Current literature emphasizes the pivotal role of IL-36 signaling in the pathogenesis of GPP, where dysregulated immune responses lead to severe inflammatory episodes characterized by pustulation and systemic symptoms.3

    Genetic analysis in our patients has revealed various mutations, particularly in the IL36RN gene, which are associated with the clinical severity and treatment response. The rapid pustular resolution in Case 1 and Case 2 (detectable IL36RN gene mutations) supports canonical IL-36–dependent inflammation. The IL36RN defects likely drive uncontrolled NF-κB/MAPK signaling cascades, consistent with prior reports linking IL36RN mutations to amplified IL-36 ligand activity.4,5 The data from these cases, including cytokine normalization post-treatment, underscore spesolimab’s efficacy in interrupting the autocrine loop. Case 3 (no detectable mutations) achieving complete remission within 18 hours highlights non-genetic IL-36 pathway activation. Despite the absence of IL36RN mutations, spesolimab effectively suppressed IL-36 signaling, evidenced by post-treatment declines in CRP and IL-6/IL-8 levels. Spesolimab direct targeting of IL-36 receptor (downstream effector molecules) to block IL-36 α / β / γ signaling, regardless of the presence of IL36RN mutations upstream.6,7 Conversely, Patient 4’s limited response to spesolimab suggests that not all GPP cases are responsive to this treatment, highlighting the complexity of the disease. Rising IL-17 levels (50.26→63.75 pg/mL) post-spesolimab, whereas switching to ixekizumab (anti-IL-17A) resolved pustules within one week. IL-17 enhances keratinocyte production of IL-36 ligands, creating a feedforward loop that likely diminishes spesolimab’s efficacy in Th17-driven disease.8 Besides, the treatment failure in Case 5 (plaque psoriasis with PsA) suggests IL-23/Th17 axis dominance may override IL-36-targeted therapy. Prior secukinumab efficacy and rapid relapse after spesolimab indicate IL-36 acts merely as a downstream effector in mixed phenotypes, where single-pathway blockade is insufficient.9 IL-17 levels remained elevated post-spesolimab, further supporting Th17 pathway resilience. May be anti-spesolimab antibodies could explain poor response.

    In conclusion, spesolimab has proven effective in treating generalized pustular psoriasis (GPP), but its efficacy is not universal. IL36RN genotyping does not influence its therapeutic effect, and IL-36 inhibition alone may not be sufficient for all GPP phenotypes. However, the presence of concomitant plaque psoriasis does impact treatment outcomes. This variability highlights the need for multi-pathway stratification in GPP treatment, rather than relying solely on IL36RN mutations. Furthermore, cytokine measurements—such as IL-6, IL-8, and IL-17—appeared to correlate with treatment responses in our study, highlighting the potential of cytokine profiling as a predictive tool for monitoring treatment efficacy.

    Consent for Publication

    We have confirmed with the patients that the details of any images, videos, recordings, etc can be published, and patients informed consent for publication of their case details and images was obtained in written form. Institutional approval was not required to publish the case details.

    Acknowledgments

    The authors express profound appreciation to all study investigators and patients for their invaluable contributions to this research.

    Funding

    This study was supported by Guangzhou Basic Research Plan Jointly Funded by the City, School (Hospital), and/or Enterprise (2024A03J0478); The Bethune Charitable Foundation Immunoinflammatory Disease Research Support Project (J202301E036).

    Disclosure

    The authors report no conflicts of interest in this work.

    References

    1. Choon SE, Navarini AA, Pinter A. Clinical course and characteristics of generalized pustular psoriasis. Am J Clin Dermatol. 2022;23:21–29. doi:10.1007/s40257-021-00654-z

    2. Rivera-Diaz R, Dauden E, Carrascosa JM, Cueva P, Puig L. Generalized pustular psoriasis: a review on clinical characteristics, diagnosis, and treatment. Dermatol Ther. 2023;13:673–688. doi:10.1007/s13555-022-00881-0

    3. Choon SE, Tok PSK, Wong KW, et al. Clinical profile of patients with acute generalized pustular psoriasis with and without IL36RN mutations in multi-ethnic Johor Bahru, Malaysia. Exp Dermatol. 2023;32:1263–1271. doi:10.1111/exd.14776

    4. Marrakchi S, Guigue P, Renshaw BR, et al. Interleukin-36-receptor antagonist deficiency and generalized pustular psoriasis. N Engl J Med. 2011;365:620–628. doi:10.1056/NEJMoa1013068

    5. Fujita H, Terui T, Hayama K, et al. Japanese guidelines for the management and treatment of generalized pustular psoriasis: the new pathogenesis and treatment of GPP. J Dermatol. 2018;45:1235–1270. doi:10.1111/1346-8138.14523

    6. Bachelez H, Choon SE, Marrakchi S, et al. Trial of spesolimab for generalized pustular psoriasis. N Engl J Med. 2021;385:2431–2440. doi:10.1056/NEJMoa2111563

    7. Morita A, Choon SE, Bachelez H, et al. Design of Effisayil 2: a randomized, double-blind, placebo-controlled study of spesolimab in preventing flares in patients with generalized pustular psoriasis. Dermatol Ther. 2023;13:347–359. doi:10.1007/s13555-022-00835-6

    8. Pathak GN, Wang E, Dhillon J, et al. Spesolimab: a review of the first IL-36 blocker approved for generalized pustular psoriasis. Ann Pharmacother. 2025;59:174–183. doi:10.1177/10600280241252688

    9. Hsieh CY, Tsai TF. Clinical advances in biological therapy for generalized pustular psoriasis: a review. Expert Opin Biol Ther. 2024;24:37–50. doi:10.1080/14712598.2024.2309301

    Continue Reading

  • Article – Philanthropy News Digest

    Article – Philanthropy News Digest

    1. Article  Philanthropy News Digest
    2. Kennedy Cancels Nearly $500 Million in mRNA Vaccine Contracts  The New York Times
    3. RFK Jr. pulls $500 million in funding for vaccine development  AP News
    4. Episode 189: Are We at Risk of Losing Our Vaccines?  CIDRAP
    5. On Monday’s show: Vax pullback affects local drug trials; raw milk sickens many  WJCT News 89.9

    Continue Reading

  • Australian researchers use sunlight to turn “forever chemicals” into fluoride-Xinhua

    CANBERRA, Aug. 9 (Xinhua) — Australian researchers have developed a sunlight-activated material that transforms toxic “forever chemicals” (PFAS) in water into harmless fluoride.

    The innovation offers a low-energy approach to tackling toxic PFAS (per- and polyfluoroalkyl substances) contamination, which has been linked to cancer, infertility, and developmental disorders, according to a statement released Friday by the University of Adelaide in South Australia.

    PFAS are synthetic chemicals in cookware, firefighting foams and water-repellent fabrics that resist breakdown and build up in the environment and the human body, with over 85 percent of Australians carrying them in their blood, it said.

    New drinking water guidelines have cut safe PFAS limits to mere nanograms per liter, researchers said.

    “PFAS contamination continues to pose a global health risk, and this research represents a critical step toward safer communities and cleaner ecosystems,” said the study’s lead researcher Cameron Shearer from the University of Adelaide.

    The team redesigned a catalyst to target PFAS fluorine atoms, achieving complete breakdown and enabling recovered fluoride to be reused in products like toothpaste or fertilizers, according to the study published in Small, a nanoscience & nanotechnology journal in Germany.

    The new materials could be integrated into treatment systems that capture and concentrate PFAS in water, which can then be degraded through exposure to the light-activated materials, the authors said, adding that work is now underway to improve the material’s stability for large-scale applications.

    Continue Reading

  • Bacterial profile and antimicrobial resistance patterns among pediatrics patients with suspected bloodstream infections in Ethiopia: a systematic review and meta-analysis | BMC Infectious Diseases

    Bacterial profile and antimicrobial resistance patterns among pediatrics patients with suspected bloodstream infections in Ethiopia: a systematic review and meta-analysis | BMC Infectious Diseases

    Bacterial bloodstream infections (BSIs) represent a major clinical challenge for both adult and pediatric hospital admissions. This underscores the critical need to consider BSI in the differential diagnosis, especially for febrile patients. In Ethiopia, the pooled prevalence of culture-confirmed pediatric BSIs is 30.66% (95% CI: 27.18–34.15%), reflecting a substantial public health concern. The analysis of 23 studies revealed considerable heterogeneity (I² = 91.3%, p < 0.001). In contrast, a separate meta-analysis focused exclusively on neonates hospitalized with clinical sepsis, which included nine studies, reported a higher pooled prevalence of bacteremia at 40. % (95% CI: 34.0–46.0%). This study also exhibited substantial heterogeneity (I² = 92.19%, p < 0.01) [37].

    Compared to previous global and regional studies, Ethiopia’s current pooled prevalence of pediatric BSIs is much higher. The review of Community-Acquired BSIs in Africa found only 8.2% in children [38], whereas Community-Onset BSIs in Africa and Asia had a median frequency of 12.5% [39]. Similarly, the Pediatric BSIs in LMICs analysis found a positive rate of 19.1%, which is still significantly lower than Ethiopia’s result [40]. In comparison, in Africa, the overall frequency of healthcare-associated infections was 12.76%, with BSIs accounting for only 17.07% [40]. The higher prevalence of pediatric BSIs in Ethiopia compared to global and regional studies may be due to limited healthcare access, poor infection prevention, and diagnostic challenges. Additionally, high antimicrobial resistance especially among Klebsiella and E. coli reflects gaps in antibiotic stewardship and infection control, exacerbating the burden in the Ethiopian setting.

    Reviews on antimicrobial resistance, such as those on carbapenem resistant Gram-Negative Bacteria in African neonates and carbapenem resistant Enterobacteriaceae in West Africa, consistently identified K. pneumoniae and E. coli as leading resistant pathogens, mirroring findings in Ethiopia where Klebsiella spp. was the most frequently isolated organism (30.6%) with a high MDR rate (72.2%). Lastly, the impact of antibiotic resistant BSIs in LMICs meta-analysis revealed increased mortality, prolonged hospital stays, and elevated healthcare costs associated with resistant infections, reinforcing the urgency of the current study’s call for enhanced infection prevention, diagnostics, and antimicrobial stewardship in Ethiopia.

    In this review, when examining the types of bacterial pathogens causing bloodstream infections in Ethiopia, Gram-negative bacteria were found to be more prevalent, comprising 56.65% (95% CI: 43.23–70.07%) of the isolates. This aligns with global trends where Gram-negative pathogens are often more difficult to treat due to their higher rates of multidrug resistance. The most common Gram-negative pathogen identified was Klebsiella spp., which accounted for 30.6% (95% CI: 20.76–40.45%) of the infections, posing significant treatment challenges due to its resistance to multiple classes of antibiotics. In comparison to studies conducted in Africa [38, 41], where Klebsiella spp. has been reported with a pooled prevalence of 2.2–28% in Asia [42], this prevalence in Ethiopia reflects a more considerable burden of infection, which might be associated with regional epidemiological factors such as higher HIV prevalence and hospital-acquired infections.

    On the other hand, Gram-positive bacteria accounted for 44.51% (95% CI: 32.68–56.34%) of the isolates, with Staphylococcus aureus being the second most common pathogen, responsible for 20.51% (95% CI: 13.63–27.39%) of infections. This is comparable to other studies in Africa [38, 41], where S. aureus is also a predominant cause of bloodstream infections. Additionally, coagulase-negative Staphylococci (CONS) made up 18.24% (95% CI: 10.77–25.71%) of the isolates. Both S. aureus and CONS are of concern due to their potential for multidrug resistance, especially in the case of methicillin-resistant S. aureus (MRSA), which complicates treatment options. This finding aligns with reports from other regions, where MRSA remains a significant challenge in managing bloodstream infections.

    Overall, the high prevalence of both Gram-negative and Gram-positive bacteria, particularly multidrug-resistant strains like Klebsiella spp. and S. aureus, underscores the urgent need for targeted antimicrobial stewardship, improved diagnostic strategies, and strengthened infection control measures to mitigate the impact of bloodstream infections in Ethiopia. The escalating rates of multidrug resistance are further complicating treatment protocols and intensifying the burden of infection in the region. Moreover, the inconsistency in the measurement and reporting of antimicrobial resistance data hampers meaningful comparisons across different countries and even within regions of the same country. This inconsistency underscores the critical need for reliable and standardized data on pathogen resistance. Access to both routine and research data on resistance patterns is essential to developing focused and effective strategies to address the growing global AMR crisis. This review highlights the alarmingly high prevalence of resistant sepsis-causing pathogens in Ethiopia, calling for urgent action to confront this escalating threat to public health.

    The pooled prevalence of MDR in bacterial blood stream infections in Ethiopia is alarmingly high, with the overall pooled MDR estimate reaching 80.54% (95% CI: 77.24–83.85%), indicating significant resistance to commonly used antibiotics. The variation in MDR rates across studies is substantial, ranging from 65.0% (95% CI: 58.82–71.18%) to 91.3% (95% CI: 87.84–94.76%), with high heterogeneity (I² = 90.4%, p < 0.001), reflecting the complexity of the issue and suggesting regional or methodological factors at play. When comparing Gram-positive and Gram-negative bacteria, it is clear that Gram-negative pathogens exhibit much higher levels of resistance. Klebsiella, Acinetobacter, and Serratia species showed a 100% MDR rate, signaling complete resistance to multiple antibiotic classes, which poses a severe treatment challenge. Other Gram-negative bacteria, such as Salmonella (98.43%), Proteus (90.72%), and Enterobacter (90.12%), also demonstrate alarmingly high resistance. In contrast, Gram-positive bacteria like Coagulase-negative Staphylococci (CONS) and Enterococcus species exhibit lower but still concerning MDR rates of 66.46%, while S. aureus and S. pyogenes both show 58.77% MDR. S. pneumoniae has a somewhat lower MDR rate of 51.08%, though it remains a significant concern. Overall, the Gram-negative bacteria exhibit higher and more worrisome resistance compared to Gram-positive bacteria, particularly with the 100% resistance seen in certain species. Implementing antimicrobial stewardship programs in hospital and pediatric care settings is critical to address the burden of MDR pathogens.

    Among Gram-positive bacteria, Coagulase-negative Staphylococci (CoNS) exhibited notable resistance to trimethoprim-sulfamethoxazole (72.05%) and penicillin (71.5%), but displayed lower resistance to amoxicillin (12.3%) and vancomycin (13%). S. aureus was highly resistant to ampicillin (85.2%) and penicillin (81.92%), but had relatively lower resistance to vancomycin (21.27%) and ciprofloxacin (21.34%). S. pyogenes showed high resistance to SXT (91.7%) and penicillin (75%), but lower resistance to ciprofloxacin, vancomycin, and ceftriaxone (16.7% for each). Enterococcus species demonstrated resistance to amoxicillin (35.13%) and ceftriaxone (44.36%), with vancomycin resistance at 26.27%. S. pneumoniae exhibited resistance to penicillin (80%), amoxicillin (38.3%), and ceftriaxone (21.7%), indicating considerable but lower resistance compared to other Gram-positive pathogens. These patterns are consistent with findings from other regions, including Africa and Sub-Saharan Africa, where similar resistance to penicillin and ampicillin has been reported in S. aureus and CoNS.

    In Gram-negative bacteria, the resistance profiles are more alarming, particularly among the most common pathogens associated with sepsis. E. coli exhibited 100% resistance to ampicillin, along with 93.75% resistance to tetracycline and 93.05% resistance to SXT. Klebsiella species showed 100% resistance to ampicillin, erythromycin, doxycycline, and cefotaxime, with complete resistance to ceftriaxone. Salmonella species exhibited 98.43% resistance to ampicillin, 93.73% resistance to tetracycline, and high resistance to chloramphenicol (93.73%), but lower resistance to ciprofloxacin (40.65%). Acinetobacter species demonstrated significant resistance to amoxicillin (71.65%), gentamicin (74.66%), and ceftriaxone, with complete resistance to doxycycline. Similarly, Serratia species showed resistance to amoxicillin, gentamicin, and tetracycline. These high levels of resistance across Gram-negative pathogens underscore the challenges in treating sepsis effectively with first-line antibiotics like ampicillin, tetracycline, and SXT.

    The resistance rates found in Ethiopia are consistent with reports from Africa [41] and Sub-Saharan Africa [38], where high resistance to penicillin, ampicillin, tetracycline, and SXT has been observed in both Gram-positive and Gram-negative bacteria. However, discrepancies are noted in certain antibiotic-bacterium combinations. For example, S. pyogenes isolates in our analysis showed a higher resistance to penicillin than previously reported in Africa [41], Asia [42] and Salmonella spp. showed higher resistance to ceftriaxone compared to previous studies in Africa [41] and Sub-Saharan Africa [38]. These differences may arise from variations in antimicrobial resistance testing methodologies and highlight the need for standardized and harmonized testing protocols across regions.

    This study employed sensitivity analysis, subgroup analysis, and meta-regression to identify potential sources of heterogeneity in the data. Subgroup analyses in Ethiopia demonstrated significant variation in the prevalence of pediatric BSIs and MDR pathogens based on demographic, clinical, and geographic characteristics. Neonatal and NICU patients had the greatest BSI rates (34.07% and 35.83%, respectively), with regional peaks in Sidama and Tigray. Gram-negative bacteria were more prevalent, and studies with bigger sample sizes revealed increased BSI prevalence, presumably due to improved identification. MDR prevalence was disturbingly high at 80.54%, with no significant differences observed across age groups, hospital wards, research periods, or sample sizes, indicating widespread and durable resistance (76.75%). resistant pediatric bloodstream infections in Ethiopia. These findings highlight the urgent need for targeted, region-specific infection control and antimicrobial stewardship efforts to curb the burden of resistant pediatric BSIs in Ethiopia.

    The sensitivity analysis demonstrated that excluding any single study had minimal impact on the pooled estimate, confirming the robustness and reliability of the overall result. The studies included in the analysis had prevalence estimates ranging from 26.54 to 34.76%, with most estimates falling between 27% and 31%. The combined estimate for these studies was 30.66% (95% CI: 27.18–34.15%), showing consistency across the included studies. Notably, the pooled estimate remained within the 95% confidence interval of the overall estimate, confirming that no single study significantly influenced the pooled prevalence of bacterial infections in Ethiopia. This reinforces the stability of the pooled estimate and indicates a consistent pattern across the studies.

    The potential impact of small-study effects and publication bias on the pooled prevalence estimate of bloodstream infections was evaluated. While the visual inspection of funnel plots suggested some degree of asymmetry, the Egger’s test results indicated that there was no significant publication bias, with a p-value greater than or equal to 0.05. This suggests that small-study effects did not notably influence the overall prevalence estimate of bloodstream infections in Ethiopia. To further assess the effect of potential publication bias, a trim-and-fill analysis was conducted. Initially, the pooled estimate from the 23 studies included in the analysis was 30.66% (95% CI: 27.18–34.15%). After performing the trim-and-fill procedure to adjust for publication bias, the adjusted pooled prevalence of bloodstream infections in Ethiopia was found to be 29.52% (95% CI: 28.52–30.53%). These results indicate that publication bias had a minor influence on the pooled estimate, and the adjustments made were relatively small.

    This study is limited by potential publication bias due to the inclusion of only published articles, excluding gray literature. All included data were phenotypic, lacking genotypic analysis of resistance mechanisms. Additionally, variability in local AMR patterns and non-standardized diagnostic methods across studies may have influenced the pooled estimates.

    Continue Reading

  • Causal Relationships between Abdominal Obesity, Type 2 Diabetes, Fasti

    Causal Relationships between Abdominal Obesity, Type 2 Diabetes, Fasti

    Introduction

    Abdominal obesity and type 2 diabetes (T2D) are among the most prevalent public health challenges today, serving as significant metabolic contributors and central components of metabolic syndrome.1 Over the last four decades, the global health landscape has transitioned from having twice as many underweight individuals as those classified as obese to a scenario where the number of obese individuals now exceeds the underweight population. This shift has positioned obesity as a critical global health issue associated with high prevalence and increased mortality rates.2 Recent estimates indicate that approximately 2 billion people worldwide are overweight or obese, accounting for 30% of the global population.3 Projections suggest that by 2030, nearly half of the adult population in the United States will be obese, with no state reporting an obesity rate below 35%, and some states nearing 60%.4

    T2D is one of the most prevalent and severe chronic metabolic diseases worldwide. Shifts in living environments, lifestyles, and dietary habits have contributed to the rise in overweight and obesity, fueling the global T2D epidemic.5 As of 2021, more than 500 million individuals worldwide were living with diabetes, with a prevalence rate exceeding 10%, and diabetes-related healthcare costs estimated at $966 billion. By 2045, these figures are expected to increase to 783 million individuals with diabetes and expenditures surpassing $1.054 trillion, creating a substantial burden on both society and families.6

    Fasting insulin (FI) serves as a key intermediary between obesity and T2D. Increased body fat content has long been recognized as a major pathogenic factor in insulin resistance. In the presence of obesity, insulin resistance and fasting hyperinsulinemia often coexist, even in the absence of hyperglycemia.7

    Orthopedic diseases, such as cervical disc disorders, osteoporosis (OP), and rheumatoid arthritis (RA), are leading causes of disability globally, placing a significant economic burden on society. Cervical disc disorders, which are prevalent degenerative conditions, are primarily responsible for neck pain and nerve root-related discomfort.8 OP is a common metabolic disorder characterized by decreased bone mass and altered bone microarchitecture.9 RA is an autoimmune disease characterized by synovitis, cartilage damage, and bone erosion in peripheral joints.10 Identifying potential risk and protective factors for orthopedic diseases is crucial for their prevention, particularly those that can be controlled through interventions.

    Population-based cross-sectional and cohort studies have demonstrated that abdominal obesity and T2D increase the risk of RA and cervical disc disorders, while T2D may enhance bone mineral density (BMD), acting as a protective factor against OP.11–13 However, discrepancies in the data persist.14 Clarifying the causal relationships between metabolic factors and orthopedic diseases is essential for providing valuable insights into clinical treatment. Previous evidence primarily stems from observational studies, and the complex pathogenic factors associated with abdominal obesity and T2D make these studies susceptible to confounding factors, leading to uncertain causal links between metabolic factors and orthopedic diseases. Unlike traditional observational studies, Mendelian randomization (MR) employs genetic variants as instrumental variables (IVs) to assess the association between exposures and outcomes. MR minimizes confounding factors by utilizing single nucleotide polymorphisms (SNPs) which are randomly assigned at conception and are not influenced by environmental or behavioral factors. This approach can effectively eliminate confounding factors and reverse causality, making MR a valuable tool for exploring the associations between complex diseases and their risk factors.15

    Given the uncertainty regarding the causal impact of metabolic factors on orthopedic diseases, an MR design was applied to evaluate the potential causal relationships. This MR analysis explores the possible causal links between metabolic factors (abdominal obesity, FI, and T2D) and orthopedic conditions including cervical disc disorders, OP, and RA.

    Materials and Methods

    Study Design

    This study utilized genome-wide association studies (GWAS) summary data from public databases for MR analysis to explore the causal relationships between metabolic factors (waist-hip ratio (WHR), FI, and T2D) and orthopedic diseases (OP, cervical disc disorders, and RA). Initially, a two-sample MR approach was utilized, incorporating various GWAS summary datasets to investigate the causal relationships between metabolic factors and the three orthopedic diseases in European populations. Subsequently, body mass index (BMI), as a measure of general obesity, was included as a potential confounder in multivariate Mendelian randomization (MVMR) to account for horizontal pleiotropy. To ensure the validity of the MR analysis, three key principles must be satisfied: (1) a strong correlation between the instrumental variable (IV) and the exposure factor; (2) the IV must be independent of any confounding factors; and (3) the IV should influence the specific disease solely through the exposure factor. This study adhered to the STROBE-MR guidelines to ensure that every step of the Mendelian randomization analysis was conducted appropriately. The specific study design process is illustrated in Figure 1.

    Figure 1 Diagrams illustrating associations examined in this study.

    Abbreviations: MR, Mendelian randomization; SNP, single nucleotide polymorphism; WHR, waist-hip ratio; FI, fasting insulin; T2D, type 2 diabetes; MVMR, Multivariate Mendelian randomization; BMI, body mass index.

    Data Sources

    Data on BMI and WHR were derived from a large GWAS involving individuals of European ancestry.16 The study combined data from the Genetic Investigation of Anthropometric Traits (GIANT) consortium (n = 212,248) and the UK Biobank (UKB) (n = 485,486), performing a meta-analysis of genome-wide studies. In this GWAS, WHR represented abdominal fat content (abdominal obesity), and BMI served as an indicator of general obesity, comprising a total of 697,734 samples. Data on the FI metabolic phenotype were obtained from the Meta-Analyses of Glucose and Insulin-Related Traits Consortium (MAGIC), comprising up to 281,416 non-diabetic individuals of European ancestry, including 151,013 samples.17 T2D data were sourced from a large-scale GWAS meta-analysis, combining three European-ancestry datasets: the DIAbetes Genetics Replication and Meta-analysis (DIAGRAM), Genetic Epidemiology Research on Aging (GERA), and UKB.18 This dataset included 62,892 cases and 596,424 controls.

    Outcome data were obtained from the latest whole-genome genetic datasets from the Finnish database R10 (https://r10.finngen.fi/), which integrates imputed genotype data generated from both new and legacy samples collected from Finnish biobanks and national health registries dating back to 1969. These data, derived from Finnish residents of European ancestry, included outcomes for OP (8,017 cases and 391,037 controls), cervical disc disorders (14,670 cases and 294,770 controls), and RA (13,621 cases and 262,844 controls) (Table 1).

    Table 1 Characteristics of the GWAS Summary Data

    Selection of Instrumental Variables

    For WHR and FI, the IVs reported in the original literature were directly utilized. This included 316 WHR-related SNPs (P < 5.0 × 10⁹) and 43 FI-related SNPs (P < 5.0 × 10⁸). For T2D, the genome-wide statistical significance threshold was set at P < 5.0 × 10⁸, with parameters set at R² < 0.001 and kb = 10,000 to mitigate the effects of linkage disequilibrium (LD). SNPs with a minor allele frequency (MAF) of less than 0.01 were excluded. To minimize bias from weak IVs, the F-statistic were used to assess the strength of the association between the IVs and metabolic factors, calculated as: F=[R2×(N-1-K)]/[K×(1-R2)], where R22×2×MAF×(1-MAF), β represents the effect size of the genetic variant related to the exposure factor, MAF represents minor allele frequency, R² indicates the proportion of exposure explained by genetic variation, N denotes the sample size, and K is the number of genetic variants. An F-statistic greater than 10 is considered indicative of a strong IV, with a higher F-statistic suggesting a lower likelihood of weak IVs.19,20

    MR Study

    The study employed a range of MR techniques, to assess causal relationships, including random or fixed-effects inverse-variance weighted (IVW) analysis, MR-Egger regression, and the weighted median (WM) method. IVW was used as the primary analysis approach. IVW does not account for an intercept term and provides a combined estimate of the causal effect of the exposure on the outcome when the genetic variants satisfy the assumptions of valid IVs.21 However, IVW results overlook the potential presence of gene-level pleiotropy. Therefore MR-Egger and WM were applied as supplementary methods to verify the validity and consistency of the findings.22 MR-Egger regression permits pleiotropy for all SNPs, detecting horizontal heterogeneity via intercept testing and providing estimates after adjusting for pleiotropy.23 WM method weights the causal effect value of each SNP according to its cluster size, yielding a temporary estimate weighted by the most significant SNPs.24

    A causal estimate is considered significant if the IVW method P < 0.05, and the direction is consistent with the MR-Egger and WM results. In the multivariate MVMR analysis, IVW was the primary method. To mitigate data bias from multiple measurements, the Bonferroni correction was applied to adjust the P-values. A total of 12 corrections were performed, resulting in a Bonferroni-corrected significance threshold of α < P/12 = 0.0042. Therefore, P < 0.0042 is regarded as a significant impact, 0.0042 < P < 0.05 indicates a suggestive association, and P > 0.05 suggests no significant effect.

    Quality Analysis

    To ensure the reliability and stability of the study results, several quality control measures were implemented, including sensitivity analyses, heterogeneity tests, and gene-level pleiotropy assessments. Sensitivity analyses included leave-one-out analyses, where each IV was excluded individually, and the results of the remaining IVs were recalculated to evaluate their impact on the outcomes. The Cochran Q-test was used to assess heterogeneity among IVs.25 If the P > 0.05 with no evidence of heterogeneity, a fixed-effects IVW model was employed as the primary analysis method. When the P < 0.05, indicating significant heterogeneity, a random-effects IVW model was used. MR-Egger regression was utilized to assess horizontal pleiotropy, with a significant deviation of the intercept term from zero indicating the presence of pleiotropy. In these cases, MR-pleiotropy residual sum and outlier (MR-PRESSO) was applied to remove outlier SNPs and eliminate horizontal pleiotropy, after which MR analysis was repeated.26

    All analyses in this study were conducted using the TwoSampleMR and MVMR packages in R software (version 4.3.1).

    Results

    Instrumental Variables

    Based on the selection criteria for IVs, SNPs for metabolic-related factors were chosen from GWAS data, resulting in 316 SNPs for WHR, 43 for FI, and 139 for T2D. The strength of these IVs was evaluated using the F-statistic. Among the selected SNPs, 13 with an F-statistic < 10, which were significantly associated with FI, were excluded. All remaining SNPs were used for MR analysis. The characteristics of the final selected SNPs are detailed in Supplementary Table S1.

    Two-Sample MR Analysis

    The MR analysis results suggest a causal relationship between genetically predicted WHR levels and cervical disc disorders, indicating a suggestive association and significant finding. Specifically, each one standard deviation (SD) increase in WHR was associated with a 1.147-fold increased risk of cervical disc disorders (IVW, p = 0.030, OR = 1.147, 95% CI: 1.014–1.297) (Figure 2). Similarly, genetically predicted FI levels show a causal relationship with cervical disc disorders, with each one SD increase in FI increasing the risk by 1.534 (IVW, p = 0.004, OR = 1.534, 95% CI: 1.145–2.055) (Figure 2), indicating a significant result and suggestive association.

    Figure 2 Mendelian randomization association of genetically determined metabolism-related traits with 3 kinds of orthopedic diseases.

    Abbreviations: SNP, single nucleotide polymorphism; WHR, waist-hip ratio; FI, fasting insulin; T2D, type 2 diabetes; OR, odds ratio; CI, confidence interval.

    Additionally, genetically predicted T2D levels are causally associated with cervical disc disorders, identifying T2D as a risk factor. Each one SD increase in the Genetic Risk Score (GRS) for T2D raised the risk of cervical disc disorders by 1.080 (IVW, p < 0.001, OR = 1.080, 95% CI: 1.037–1.125) (Figure 2). This finding is significant and indicates a suggestive association. Genetically predicted WHR levels also show a causal relationship with RA, with each one SD increase in WHR increasing the risk of RA by 1.260 (IVW, p < 0.001, OR = 1.260, 95% CI: 1.113–1.426) (Figure 2), signifying a significant result and suggestive association.

    Furthermore, genetically predicted T2D levels exhibited a causal relationship with OP, with T2D acting as a protective factor. Each one SD increase in the GRS for T2D reduced the risk of OP by 0.925 (IVW, p = 0.010, OR = 0.925, 95% CI: 0.873–0.981) (Figure 2), indicating a significant and suggestive association. Additionally, the analysis of FI-related SNPs (n = 43) without excluding weak IVs reveals a significant causal relationship between genetically predicted FI levels and cervical disc disorders. Each one SD increase in FI raised the risk of cervical disc disorders by 1.469 (IVW, p = 0.032, OR = 1.469, 95% CI: 1.034–2.087) (Figure 2), providing further evidence of a significant result and suggestive association. Supplementary Table S2 presents the results of four MR analysis methods, all of which showed consistent Beta directions across the analyses.

    MVMR Analysis

    In the MVMR analysis, BMI was adjusted as a confounder. After considering the influence of BMI, MVMR was applied to observe the independent effects of exposures on outcomes. The results of the MVMR analysis indicate that the causal relationship between WHR levels and cervical disc disorders disappeared after adjusting for BMI (IVW, p = 0.107, OR = 1.118, 95% CI: 0.976–1.281). However, FI levels continued to show a positive causal effect on cervical disc disorders (IVW, p < 0.001, OR = 1.716, 95% CI: 1.271–2.317). Similarly, T2D levels maintained a positive causal effect on cervical disc disorders (IVW, p < 0.001, OR = 1.092, 95% CI: 1.048–1.137) (Figure 2).

    WHR levels continued to show a positive causal effect on RA (IVW, p = 0.011, OR = 1.203, 95% CI: 1.044–1.385). T2D levels continued to have a protective effect on OP (IVW, p < 0.001, OR = 0.912, 95% CI: 0.867–0.959). Furthermore, after adjusting for BMI, a causal relationship between T2D levels and RA emerged, indicating that T2D is a risk factor for RA. Each unit increase in T2D increased the risk of RA by 1.062 times (IVW, p = 0.016, OR = 1.062, 95% CI: 1.012–1.116) (Figure 3 and Supplementary Table S3).

    Figure 3 Forest plot for the MVMR adjusted for BMI.

    Abbreviations: WHR, waist-hip ratio; FI, fasting insulin; T2D, type 2 diabetes; OR, odds ratio; CI, confidence interval.

    Sensitivity and Pleiotropy Analyses

    The MR-Egger intercept tests revealed P > 0.05 (Table 2), suggesting no evidence of horizontal pleiotropy between the selected SNPs and outcome factors, confirming the validity of the MR methods for causal inference in this study. Cochran’s Q-test for heterogeneity among SNPs showed no significant heterogeneity p > 0.05 for FI after excluding weak IVs (Table 2). Consequently, a fixed-effects IVW model was applied for analysis. For other tests showing significant heterogeneity (P < 0.05) (Table 2), a random-effects IVW model was employed.

    Table 2 Sensitivity Analyses with Directional Pleiotropy Test and Heterogeneity Test

    Leave-one-out sensitivity analyses yielded results consistent with the analysis of all included SNPs, with no SNPs significantly influencing causal associations (Supplementary Table S4). These findings confirm stable causal relationships between the three metabolic factors (WHR, FI, and T2D) and the three orthopedic diseases (cervical disc disorders, osteoporosis, and rheumatoid arthritis).

    Discussion

    This study utilized publicly available GWAS data and employed both Univariate MR (UVMR) and MVMR methods to explore the causal relationships between three metabolic factors (WHR, FI, and T2D) and various orthopedic diseases (OP, cervical disc disorders, and RA). The UVMR results indicated that T2D played a protective role against OP, while WHR, FI, and T2D appeared to be risk factors for cervical disc disorders, and WHR was a risk factor for RA. The findings from the MVMR analysis, which adjusted for BMI as a confounder, indicated that T2D remained protective against OP, while FI and T2D remained risk factors for cervical disc disorders. Notably, the causal relationship between WHR and cervical disc disorders disappeared when accounting for BMI. Additionally, WHR remained a risk factor for RA, but the positive causal relationship between T2D and RA was attenuated after adjusting for BMI.

    Cervical disc disorders are common orthopedic diseases caused by degenerative changes in the cervical intervertebral discs, resulting in neck pain and nerve root-related discomfort.8 This disorder is characterized by a detrimental cycle involving altered cell metabolism, matrix, and biomechanics, ultimately leading to mechanical instability and loss of the shock-absorbing function of the intervertebral discs.27 Over the past 30 years, the prevalence and disability rates of this disease have significantly increased. By 2015, more than one-third of the global population had experienced neck pain lasting longer than three months.28

    Most existing literature investigates the relationship between obesity and lumbar diseases, with fewer studies focusing on obesity and cervical diseases, typically using BMI as a measure of obesity levels. However, actual fat distribution varies significantly among individuals, leading to the growing recognition that WHR better reflects fat distribution and central obesity. This makes WHR a more accurate indicator of metabolic status than BMI.29

    The UVMR results indicate that WHR is a risk factor for cervical disc disorders, while MVMR analysis suggests that this relationship may be attributed to BMI. Sun et al also found that BMI negatively affects the risk of cervical spondylosis in MR studies, demonstrating that general obesity (as indicated by BMI) is a risk factor for cervical spondylosis (IVW, OR = 1.166, 95% CI: 1.052–1.292, P = 0.003).30 However, observational studies regarding whether obesity causes cervical spondylosis have been inconsistent.

    Teraguchi et al used MRI to examine the prevalence and distribution of intervertebral disc degeneration across the entire spine in a small population (n = 975) and identified a significant correlation between obesity (assessed by BMI) and intervertebral disc degeneration in all regions (cervical, thoracic, and lumbar).11 Sheng et al, however, found no significant association between overweight and obesity (assessed by BMI) and cervical diseases in a cross-sectional sample from the 2014 Medical Expenditure Panel Study (MEPS) (n = 23,048), which is considered representative of the US population.14 Their study further confirmed the absence of a significant association between obesity and cervical diseases, aligning with the prevailing opinion that no significant relationship exists between obesity and cervical spondylosis. Interestingly, studies on orthopedic diseases in children have shown a high correlation between obesity and neck pain.31,32 Dianat et al found that children with a BMI below 17.33 were less likely to report neck pain (OR = 0.63, 95% CI: 0.42–0.95), suggesting that lower weight may reduce the risk of neck pain.33 Azabagic et al found that the most common chronic pain site associated with overweight and obesity (assessed by BMI) was the neck (OR = 1.212), and Krul et al reported a higher frequency of chronic neck pain in overweight or obese children (assessed by BMI) (OR = 2.60; 95% CI: 1.30–5.19).34,35

    The MVMR results show that after excluding the effect of BMI, BMI-adjusted WHR (WHRadjBMI), which directly measures abdominal fat content and is independent of obesity level, is not associated with cervical disc disorders, consistent with previous observational studies.14,36 This may be because mechanical factors are the primary pathogenic contributors to disc diseases, and the cervical spine bears less body weight than the lumbar spine, reducing the impact of abdominal obesity on the cervical spine.37 In children, head weight (indicated by head circumference) is positively correlated with newborn BMI, with the head accounting for 20 to 25% of total body weight at birth, which decreases to 4 to 6% by adulthood.38 Consequently, children’s cervical spines bear greater head weight than those of adults. To more accurately assess local obesity levels in the head and neck region, some scholars have proposed the concept of subcutaneous fat tissue thickness (SFTT) at the cervical level. A retrospective study based on this concept found a close correlation between SFTT at the C3 level and intervertebral disc degeneration, with females having an SFTT > 9.64mm being more likely to develop spinal degeneration, and males showing a similar trend with SFTT > 8.21mm.39 Unfortunately, GWAS data related to neck circumference or neck fat distribution could not be obtained.

    In addition to mechanical factors, significant fat infiltration around the cervical spine creates an inflammatory environment within disc tissues and induces abnormal adipokine production. On one hand, the excessive production of inflammatory factors such as IL-1β, TNF-α, and IL-6 accelerates disc matrix degradation, cell senescence, and cell mortality.40 On the other hand, adipokines like leptin (primarily produced by white adipose tissue) and resistin (linked to insulin resistance) activate immune regulation and inflammatory signaling pathways within disc tissues, further enhancing the expression of inflammatory factors.41

    Most existing studies examine the relationship between T2D and lumbar diseases, commonly suggesting that obesity induced by T2D is the primary pathogenic factor.42 The current study, however, indicates that both T2D and FI are risk factors for cervical disc disorders. The high-glucose environment in patients with T2D accelerates disc degeneration, with advanced glycation end-products (AGEs) associated with diabetes accumulating in the annulus fibrosus, nucleus pulposus, and cartilage endplate, ultimately contributing to disc degeneration independent of obesity.43 In vitro experiments reveal that elevated blood glucose concentrations inhibit nucleus pulposus cell proliferation and disrupt disc cell homeostasis.44 Furthermore, since insulin shares structural similarities with Insulin-Like Growth Factor-1 (IGF-1) and IGF-2, hyperinsulinemia may interfere with IGF signaling pathways, accelerating disc degeneration.45,46

    RA is a chronic inflammatory disease characterized by symmetric synovitis, cartilage damage, and bone erosion in peripheral joints. Its global prevalence ranges from approximately 0.25% to 1.0%, affecting 1 in every 200 individuals. The incidence is 2 to 3 times higher in women than in men, with peak onset occurring between ages 50 and 59.10,47

    Previous MR studies have confirmed that WHR is a risk factor for RA (OR = 1.41; 95% CI, 1.22–1.62).48 Similarly, observational studies have shown that abdominal obesity increases the risk of RA compared to general obesity (HR = 1.22, 95% CI: 1.06–1.41), aligning with the UVMR results.49 When individuals first exhibit symptoms of RA, their immune system may have already been in a state of chronic inflammation for years. Obesity itself is a chronic inflammatory state, with adipose tissue, particularly visceral fat, promoting the production of various inflammatory factors and adipokines such as IL-1, IL-6, IL-17, TNF-α, interferon-γ, and leptin. This induces systemic inflammation, increases peripheral tissue damage, and triggers autoimmune responses, which may lead to and exacerbate systemic inflammatory conditions, including inflammatory arthritis.40,41,48 Martin et al proposed new expressions for different body fat distributions: favorable adiposity (FA), which has higher subcutaneous fat but lower visceral fat, and unfavorable adiposity (UFA), characterized by higher subcutaneous and visceral fat.50 They used MR studies to confirm that both FA and UFA are risk factors for RA, consistent with the UVMR and MVMR results.

    T2D and RA share common inflammatory pathological mechanisms. Studies have shown that abnormal glucose metabolism, insulin resistance, and T2D are frequently observed in patients with RA.51 Zhang et al confirmed through MR studies that RA is a risk factor for T2D (OR = 1.04; 95% CI, 1.02–1.05).52 However, limited evidence suggests that the risk of RA increases after a T2D diagnosis. Lu et al found that female patients with T2D, especially younger women, had a significantly increased risk of RA (OR = 1.46, 95% CI: 1.24–1.72), while no such association was observed in men.12 Lahiri et al used data from the European Prospective Investigation of Cancer-Norfolk and the Norfolk Arthritis Register Study (EPIC-2-NOAR Study) and found that T2D (HR = 2.54, 95% CI: 1.26–5.09) and obesity (HR = 2.75, 95% CI: 1.39–5.46) were both associated with an increased risk of inflammatory polyarthritis.53 This finding provides new insights into the MR research, suggesting that T2D and RA may have a mutual causal relationship.

    A high-glucose environment enhances the interaction between thioredoxin-interacting protein (TXNIP) and the NOD-like receptor protein 3 (NLRP3) inflammasome, activating caspase 1 and promoting IL-1β release. Additionally, hyperglycemia directly induces β-cell apoptosis through Fas receptors, further upregulating IL-1β. Synovial fluid of patients with RA contains high levels of IL-1β, which regulates leukocyte recruitment, induces matrix metalloproteinases (MMPs), promotes cartilage degradation, inhibits new matrix synthesis, causes joint damage, and induces bone erosion by stimulating osteoclast differentiation and activation.51,54 Studies have shown that IL-1 blockers (eg, Anakinra and Canakinumab) can simultaneously improve the progression of both diseases.55

    OP is a prevalent metabolic disorder characterized by reduced bone mass, altered bone microarchitecture, increased bone fragility, and an elevated risk of fractures. Due to its significant public health impact, it is often referred to as the “silent epidemic of the 21st century”.9 The gold standard for diagnosing OP remains dual-energy X-ray absorptiometry for measuring BMD.

    MA et al performed a meta-analysis on the association between T2D and BMD involving 3437 patients with diabetes and 19139 controls.13 The analysis found significantly higher BMD at the femoral neck, hip, and spine in patients with T2D compared to non-diabetic individuals. However, this study did not include subgroup analyses. Qiu et al conducted a meta-analysis examining the relationship between diabetes and low bone mass, which included 4599 patients with diabetes and 19741 controls.56 They found no association between T2D and low bone mass, but identified a significant link between type 1 diabetes and low bone density. These results remained consistent in subgroup analyses. Zhou et al utilized data from the US National Health and Nutrition Examination Survey (NHANES) for a cross-sectional study and European GWAS summary statistics for a MR study.57 They found a positive correlation between fasting glucose levels and BMD at the hip, femoral neck, and first lumbar vertebra (L1) in patients with T2D. Additionally, FI was positively correlated with hip BMD in patients with T2D, who had higher hip BMD than non-diabetic individuals.

    Obesity associated with T2D was previously thought to indirectly increased BMD. However, numerous recent MR studies have confirmed a direct protective role of T2D against OP.58–60 Ma et al demonstrated, through mediated MR analysis, that while BMI increases the risk of OP, this effect is mediated by T2D.61 The MR results further suggest that T2D acts as a direct protective factor against OP, while FI does not show a significant association with OP, possibly due to the inclusion of non-diabetic individuals in the GWAS. Notably, MR findings suggest FI may be a potential protective factor against OP, though the results were not significant [UVMR: OR = 0.648, P = 0.154; MVMR: OR = 0.813, P = 0.272], which is generally consistent with previous studies. High glucose levels in T2D reduce RANKL levels, inhibiting the RANKL/RANK/OPG pathway, which disrupts osteoclast differentiation and function, thus preventing bone matrix degradation and increasing BMD.62 Elevated insulin levels in circulation may also contribute to the higher BMD in patients with T2D, as insulin, structurally similar to insulin-like growth factor 1 (IGF-1), can bind to IGF-1 receptors on osteoblasts, facilitating bone anabolic processes.45

    This study has several limitations. First, the results may not be generalizable to non-European populations, as the GWAS dataset were of European ancestry. Differences in genetics, cultural practices, and lifestyles may influence the observed associations. Future studies should incorporate more diverse cohorts to validate the generalizability of these results across various ethnic and demographic groups. Second, due to limitations within the dataset, we were unable to perform subgroup analyses based on age, sex, or disease severity, which may have provided further insight into differential risk profiles and disease mechanisms. Further subgroup analyses of orthopedic disease datasets could be performed. Third, the complex pathogenesis of abdominal obesity and T2D complicates the complete exclusion of confounding factors, which may account for the variability in results observed across different MR studies.63 Additional studies incorporating multi-omics data and pathway-specific analyses may better elucidate these relationships and clarify causal mechanisms.

    Conclusions

    In conclusion, this study suggests that abdominal obesity and T2D are associated with an increased risk of RA, with T2D exhibiting a causal relationship with a heightened risk of cervical disc disorders and a reduced risk of OP. These findings enhance the understanding of the relationships between metabolic factors and common orthopedic diseases, providing valuable insights for clinical practice and disease management.

    Data Sharing Statement

    The datasets presented in this study can be found in online repositories. The names of the repository/repositories and accession number(s) can be found in the article.

    Ethics Approval

    This study was conducted in accordance with the declaration of Helsinki. This study was conducted with approval from the Ethics Committee of Affiliated Hospital of Shandong Traditional Chinese Medicine University.

    Acknowledgments

    All data used in this study were obtained from openly available databases and consortiums. We express our sincere appreciation to them.

    Funding

    This work was supported by the Shandong Provincial Natural Science Foundation (No. ZR20210H229) and the Science and Technology Plan Projects of Shandong Provincial Administration of Traditional Chinese Medicine (No. 2021Q107).

    Disclosure

    The authors declare no competing interests in this work.

    References

    1. Chandrasekaran P, Weiskirchen R. The Role of Obesity in Type 2 Diabetes Mellitus-An Overview. Int. J Mol Sci. 2024;25(3):1882. doi:10.3390/ijms25031882

    2. Boutari C, Mantzoros CS. A 2022 update on the epidemiology of obesity and a call to action: as its twin COVID-19 pandemic appears to be receding, the obesity and dysmetabolism pandemic continues to rage on. Metabolism. 2022;133:155217. doi:10.1016/j.metabol.2022.155217

    3. Caballero B. Humans against Obesity: who Will Win? Adv Nutr. 2019;10(suppl_1):S4–s9. doi:10.1093/advances/nmy055

    4. Ward ZJ, Bleich SN, Cradock AL, et al. Projected U.S. State-Level Prevalence of Adult Obesity and Severe Obesity. New Engl J Med. 2019;381(25):2440–2450. doi:10.1056/NEJMsa1909301

    5. Tinajero MG, Malik VS. An Update on the Epidemiology of Type 2 Diabetes: a Global Perspective. Endocrinol Metab Clinics North Am. 2021;50(3):337–355. doi:10.1016/j.ecl.2021.05.013

    6. Sun H, Saeedi P, Karuranga S, et al. IDF Diabetes Atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183:109119. doi:10.1016/j.diabres.2021.109119

    7. Abdul-Ghani M, DeFronzo RA. Fasting Hyperinsulinemia and Obesity: cause or Effect. J Clin Endocrinol Metab. 2023;108(10):e1151–e2. doi:10.1210/clinem/dgad118

    8. Qiu Y, Wei X, Tao Y, et al. Causal association of leisure sedentary behavior and cervical spondylosis, sciatica, intervertebral disk disorders, and low back pain: a Mendelian randomization study. Front Public Health. 2024;12:1284594. doi:10.3389/fpubh.2024.1284594

    9. Aibar-Almazán A, Voltes-Martínez A, Castellote-Caballero Y, Afanador-Restrepo DF, Carcelén-Fraile MDC, López-Ruiz E. Current Status of the Diagnosis and Management of Osteoporosis. Int J Mol Sci. 2022;23(16):9465. doi:10.3390/ijms23169465

    10. Di Matteo A, Bathon JM, Emery P. Rheumatoid arthritis. Lancet. 2023;402(10416):2019–2033. doi:10.1016/S0140-6736(23)01525-8

    11. Ali D, Tencerova M, Figeac F, Kassem M, Jafari A. The pathophysiology of osteoporosis in obesity and type 2 diabetes in aging women and men: the mechanisms and roles of increased bone marrow adiposity. Front Endocrinol. 2022;13:981487. doi:10.3389/fendo.2022.981487

    12. Lu MC, Yan ST, Yin WY, Koo M, Lai NS. Risk of rheumatoid arthritis in patients with type 2 diabetes: a nationwide population-based case-control study. PLOS ONE. 2014;9(7):e101528. doi:10.1371/journal.pone.0101528

    13. Ma L, Oei L, Jiang L, et al. Association between bone mineral density and type 2 diabetes mellitus: a meta-analysis of observational studies. Eur J Epidemiol. 2012;27(5):319–332. doi:10.1007/s10654-012-9674-x

    14. Sheng B, Feng C, Zhang D, Spitler H, Shi L. Associations between Obesity and Spinal Diseases: a Medical Expenditure Panel Study Analysis. Int J Environ Res Public Health. 2017;14(2):183. doi:10.3390/ijerph14020183

    15. Tin A, Köttgen A. Mendelian Randomization Analysis as a Tool to Gain Insights into Causes of Diseases: a Primer. J. Am Soc Nephrol. 2021;32(10):2400–2407. doi:10.1681/ASN.2020121760

    16. Pulit SL, Stoneman C, Morris AP, et al. Meta-analysis of genome-wide association studies for body fat distribution in 694649 individuals of European ancestry. Human Molecular Genetics. 2019;28(1):166–174. doi:10.1093/hmg/ddy327

    17. Chen J, Spracklen CN, Marenne G, et al. The trans-ancestral genomic architecture of glycemic traits. Nature Genet. 2021;53(6):840–860. doi:10.1038/s41588-021-00852-9

    18. Xue A, Wu Y, Zhu Z, et al. Genome-wide association analyses identify 143 risk variants and putative regulatory mechanisms for type 2 diabetes. Nat Commun. 2018;9(1):2941. doi:10.1038/s41467-018-04951-w

    19. Pritchard JK, Przeworski M. Linkage disequilibrium in humans: models and data. Am J Hum Genet. 2001;69(1):1–14. doi:10.1086/321275

    20. Burgess S, Thompson SG. Avoiding bias from weak instruments in Mendelian randomization studies. Int J Epidemiol. 2011;40(3):755–764. doi:10.1093/ije/dyr036

    21. Pagoni P, Dimou NL, Murphy N, Stergiakouli E. Using Mendelian randomisation to assess causality in observational studies. Evidence Mental Health. 2019;22(2):67–71. doi:10.1136/ebmental-2019-300085

    22. Burgess S, Butterworth A, Thompson SG. Mendelian randomization analysis with multiple genetic variants using summarized data. Geneti Epidemiol. 2013;37(7):658–665. doi:10.1002/gepi.21758

    23. Hemani G, Bowden J, Davey Smith G. Evaluating the potential role of pleiotropy in Mendelian randomization studies. Human Molecular Genetics. 2018;27(R2):R195–r208. doi:10.1093/hmg/ddy163

    24. Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent Estimation in Mendelian Randomization with Some Invalid Instruments Using a Weighted Median Estimator. Geneti Epidemiol. 2016;40(4):304–314. doi:10.1002/gepi.21965

    25. Greco MF, Minelli C, Sheehan NA, Thompson JR. Detecting pleiotropy in Mendelian randomisation studies with summary data and a continuous outcome. Stat Med. 2015;34(21):2926–2940. doi:10.1002/sim.6522

    26. Hemani G, Zheng J, Elsworth B, et al. The MR-Base platform supports systematic causal inference across the human phenome. eLife. 2018;7. doi:10.7554/eLife.34408

    27. Vergroesen PP, Kingma I, Emanuel KS, et al. Mechanics and biology in intervertebral disc degeneration: a vicious circle. Osteoarthritis Cartilage. 2015;23(7):1057–1070. doi:10.1016/j.joca.2015.03.028

    28. Hurwitz EL, Randhawa K, Yu H, Côté P, Haldeman S. The Global Spine Care Initiative: a summary of the global burden of low back and neck pain studies. Eur Spine J. 2018;27(Suppl 6):796–801. doi:10.1007/s00586-017-5432-9

    29. Åberg F, Färkkilä M, Salomaa V, et al. Waist-Hip ratio is superior to BMI in predicting liver-related outcomes and synergizes with harmful alcohol use. Communicat Med. 2023;3(1):119. doi:10.1038/s43856-023-00353-2

    30. Sun Y, Jin M, Yu T, Zhang J. Cardiovascular risk factors mediating the protective effect of education on cervical spondylosis risk. Sci Rep. 2023;13(1):936. doi:10.1038/s41598-023-28153-7

    31. Singh S, Kumar D, Kumar S. Risk factors in cervical spondylosis. J Clinl Orthopaedics Trauma. 2014;5(4):221–226. doi:10.1016/j.jcot.2014.07.007

    32. Samal S, Panigrahi P, Dutta A. Social epidemiology of excess weight and central adiposity in older Indians: analysis of Study on global AGEing and adult health (SAGE). BMJ open. 2015;5(11):e008608. doi:10.1136/bmjopen-2015-008608

    33. Dianat I, Alipour A, Asgari Jafarabadi M. Risk factors for neck and shoulder pain among schoolchildren and adolescents. J Paediatr Child Health. 2018;54(1):20–27. doi:10.1111/jpc.13657

    34. Azabagic S, Pranjic N. The Site of Musculoskeletal Pain in School Children with Excessive Body Weight and Obesity in Bosnia and Herzegovina. Materia Socio-Medica. 2019;31(2):88–92. doi:10.5455/msm.2019.31.88-92

    35. Krul M, van der Wouden JC, Schellevis FG, van Suijlekom-Smit LW, Koes BW. Musculoskeletal problems in overweight and obese children. Ann Family Med. 2009;7(4):352–356. doi:10.1370/afm.1005

    36. Yuan C, Jian Z, Feng S, et al. Do Obesity-Related Traits Affect Prostate Cancer Risk through Serum Testosterone? A Mendelian Randomization Study. Cancers. 2023;15(19):4884. doi:10.3390/cancers15194884

    37. Gandhi R, Woo KM, Zywiel MG, Rampersaud YR. Metabolic syndrome increases the prevalence of spine osteoarthritis. Orthopaedic Surg. 2014;6(1):23–27. doi:10.1111/os.12093

    38. Makhoul IR, Abulhija H, Smolkin T, Shehadeh N, Hochberg Z. Modified anthropometry in prepubertal Israeli children while excluding the head’s weight and height. Acta paediatrica. 2012;101(11):e496–9. doi:10.1111/j.1651-2227.2012.02792.x

    39. Cao J, Sun D, Guo L, Wang R, Liu P. A novel radiological index for the evaluation of cervical posterior subcutaneous fat tissue thickness and cervical spine degeneration: a retrospective study. Medicine. 2023;102(29):e34355. doi:10.1097/MD.0000000000034355

    40. Francisco V, Pino J, González-Gay M, et al. A new immunometabolic perspective of intervertebral disc degeneration. Nat Rev Rheumatol. 2022;18(1):47–60. doi:10.1038/s41584-021-00713-z

    41. Shin JH, Park S, Cho H, Kim JH, Choi H. Adipokine human Resistin promotes obesity-associated inflammatory intervertebral disc degeneration via pro-inflammatory cytokine cascade activation. Sci Rep. 2022;12(1):8936. doi:10.1038/s41598-022-12793-2

    42. Agius R, Galea R, Fava S. Bone mineral density and intervertebral disc height in type 2 diabetes. J Diabet Complicat. 2016;30(4):644–650. doi:10.1016/j.jdiacomp.2016.01.021

    43. Yang F, Zhu D, Wang Z, et al. Role of Advanced Glycation End Products in Intervertebral Disc Degeneration: mechanism and Therapeutic Potential. Oxid Med Cell Longev. 2022;2022(1):7299005. doi:10.1155/2022/7299005

    44. Gorth DJ, Shapiro IM, Risbud MV. Discovery of the drivers of inflammation induced chronic low back pain: from bacteria to diabetes. Discov Med. 2015;20(110):177–184.

    45. Conte C, Epstein S, Napoli N. Insulin resistance and bone: a biological partnership. Acta diabetologica. 2018;55(4):305–314. doi:10.1007/s00592-018-1101-7

    46. Lin H, Tian S, Peng Y, et al. IGF Signaling in Intervertebral Disc Health and Disease. Front Cell Develop Biol. 2021;9:817099. doi:10.3389/fcell.2021.817099

    47. Smith MH, Berman JR. What Is Rheumatoid Arthritis? JAMA. 2022;327(12):1194. doi:10.1001/jama.2022.0786

    48. Li X, Zhu J, Zhao W, et al. The Causal Effect of Obesity on the Risk of 15 Autoimmune Diseases: a Mendelian Randomization Study. Obesity Facts. 2023;16(6):598–605. doi:10.1159/000534468

    49. Marchand NE, Sparks JA, Tedeschi SK, et al. Abdominal Obesity in Comparison with General Obesity and Risk of Developing Rheumatoid Arthritis in Women. J Rheumatol. 2021;48(2):165–173. doi:10.3899/jrheum.200056

    50. Martin S, Tyrrell J, Thomas EL, et al. Disease consequences of higher adiposity uncoupled from its adverse metabolic effects using Mendelian randomisation. eLife. 2022;11:1.

    51. Di Muzio C, Cipriani P, Ruscitti P. Rheumatoid Arthritis Treatment Options and Type 2 Diabetes: unravelling the Association. BioDrugs. 2022;36(6):673–685. doi:10.1007/s40259-022-00561-7

    52. Zhang K, Jia Y, Wang R, et al. Rheumatoid arthritis and the risk of major cardiometabolic diseases: a Mendelian randomization study. Scand J Rheumatol. 2023;52(4):335–341. doi:10.1080/03009742.2022.2070988

    53. Lahiri M, Luben RN, Morgan C, et al. Using lifestyle factors to identify individuals at higher risk of inflammatory polyarthritis (results from the European Prospective Investigation of Cancer-Norfolk and the Norfolk Arthritis Register–the EPIC-2-NOAR Study). Ann Rheumatic Dis. 2014;73(1):219–226. doi:10.1136/annrheumdis-2012-202481

    54. Galicia-Garcia U, Benito-Vicente A, Jebari S, et al. Pathophysiology of Type 2 Diabetes Mellitus. Int J Mol Sci. 2020;21(17):6275. doi:10.3390/ijms21176275

    55. Giacomelli R, Ruscitti P, Alvaro S, et al. IL-1β at the crossroad between rheumatoid arthritis and type 2 diabetes: may we kill two birds with one stone? Expert Rev Clinical Immunol. 2016;12(8):849–855. doi:10.1586/1744666X.2016.1168293

    56. Qiu J, Li C, Dong Z, Wang J. Is diabetes mellitus a risk factor for low bone density: a systematic review and meta-analysis. BMC Endocr Disord. 2021;21(1):65. doi:10.1186/s12902-021-00728-3

    57. Zhou H, Li C, Song W, et al. Increasing fasting glucose and fasting insulin associated with elevated bone mineral density-evidence from cross-sectional and MR studies. Osteoporosis Int. 2021;32(6):1153–1164. doi:10.1007/s00198-020-05762-w

    58. Ahmad OS, Leong A, Miller JA, et al. A Mendelian Randomization Study of the Effect of Type-2 Diabetes and Glycemic Traits on Bone Mineral Density. J Bone Mineral Res. 2017;32(5):1072–1081. doi:10.1002/jbmr.3063

    59. Huang G, Chen X, Chen Y, et al. Causal relationship between type 2 diabetes mellitus and bone mineral density: a Mendelian randomization study in an East Asian population. Osteoporosis Int. 2023;34(10):1719–1727. doi:10.1007/s00198-023-06807-6

    60. Cheng L, Wang S, Tang H. Type 2 diabetes mellitus plays a protective role against osteoporosis –Mendelian randomization analysis. BMC Musculoskel Disorders. 2023;24(1):444. doi:10.1186/s12891-023-06528-1

    61. Ma W, Zhou X, Huang X, Xiong Y. Causal relationship between body mass index, type 2 diabetes and bone mineral density: Mendelian randomization. PLOS ONE. 2023;18(10):e0290530. doi:10.1371/journal.pone.0290530

    62. Sassi F, Buondonno I, Luppi C, et al. Type 2 diabetes affects bone cells precursors and bone turnover. BMC Endocr Disord. 2018;18(1):55. doi:10.1186/s12902-018-0283-x

    63. Inamo J, Kochi Y, Takeuchi T. Is type 2 diabetes mellitus an inverse risk factor for the development of rheumatoid arthritis? J Human Genet. 2021;66(2):219–223. doi:10.1038/s10038-020-00837-2

    Continue Reading