Blog

  • Court orders detention of 3 over alleged TSMC trade secret theft

    Court orders detention of 3 over alleged TSMC trade secret theft

    Taipei, Sept. 1 (CNA) The Intellectual Property and Commercial Court (IPCC) has ordered one former employee and two current employees of Taiwan Semiconductor Manufacturing Co. (TSMC) be detained and held incommunicado for allegedly stealing sensitive trade secrets involving the company’s advanced 2 nanometer process.

    The ruling was handed down after the three suspects — Chen Li-ming (陳力銘), an ex-TSMC engineer, Wu Ping-chun (吳秉駿) and Ko Yi-ping (戈一平), who currently worked as engineers for the chipmaker — were referred to the IPCC on Monday morning.

    A three-judge panel at the IPCC ruled that the three deleted their communications records after their conduct involving the alleged theft of TSMC’s advanced 2nm process was discovered, so there are reasonable fears they could continue to destroy evidence and collude with one another in making false statements.

    The judges said that as the three suspects’ conduct potentially harmed national security and affected market competition, they should be detained to make sure the future court hearings proceed smoothly.

    On Aug. 27, the Taiwan High Prosecutors Office Intellectual Property Branch indicted the three suspects for their roles in the alleged theft of trade secrets and violation of the National Security Act by obtaining national core technology secrets for use abroad.

    Prosecutors are seeking prison terms of 14 years, nine years, and seven years, respectively for Chen, Wu and Ko.

    Chen is a former TSMC engineer working at Tokyo Electron Ltd. (TEL), a Japan-based supplier to TSMC. During the second half of 2024 and the first half of 2025, Chen asked Wu and Ko to provide trade secrets they had access to, under the guise of helping TEL secure more TSMC supplying contracts, according to prosecutors.

    TSMC detected the irregularity and filed a lawsuit against the three in early July. Prosecutors launched searches and raids related to the case from July 25-28 and secured approval from the IPPC to detain them men the investigation.

    The case was referred to the IPCC on Monday, and a hearing was needed to decide whether the three suspects could continue to be detained.

    TEL said in late August that an internal investigation has so far found no evidence that confidential information about TSMC’s 2nm was leaked to a third party.

    TSMC is developing the 2nm process, which is scheduled to start mass production in the second half of this year. Currently, the 3nm process is the latest technology for TSMC to begin commercial production.

    (By Lin Chang-shun and Frances Huang)

    Enditem/AW

    Continue Reading

  • Rate of Post-Stroke Depression and Associated Factors in Saudi single

    Rate of Post-Stroke Depression and Associated Factors in Saudi single

    Turki Aljuhani,1,2 Shahd Alsubaie,3,4 Abrar M Al-Mutairi,5 Abdulmajeed S Altheyab,1,2 Abdulrahman M Alsahali,1 Abdulrahman S Alhamdan,1 Falah M Alqahtani,1 Lafi H Olayan,2,6 Mohammed Senitan7

    1Department of Occupational Therapy, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, 11481, Saudi Arabia; 2King Abdullah International Medical Research Center, Riyadh, 11481, Saudi Arabia; 3Department of Rehabilitation, Ministry of National Guard – Heath Affairs, Riyadh, 11481, Saudi Arabia; 4Saudi Central Board for Accreditation for Healthcare Institution, Riyadh, 12264, Saudi Arabia; 5Research Unit, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, 11481, Saudi Arabia; 6Anesthesia Technology Department, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 11481, Saudi Arabia; 7Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh, 11673, Saudi Arabia

    Correspondence: Turki Aljuhani, Department of Occupational Therapy, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, 11481, Saudi Arabia, Tel +96611429999, Email [email protected]

    Purpose: Stroke is a significant global health concern, with post-stroke depression (PSD) affecting approximately 30% of patients and contributing to reduced quality of life and increased mortality. In Saudi Arabia, data on PSD frequency and associated factors remain limited in relation to the rehabilitation of stroke patients, highlighting the need for further investigation. The study’s aims to investigate the rate of PSD and the factors that influence PSD.
    Methods: This feasibility study was conducted at the Neurorehabilitation Unit of King Abdulaziz Medical City in Riyadh, Saudi Arabia (October 2023–October 2024), and included stroke patients aged 18– 80 years. Data on stroke severity (NIHSS), functional independence (FIM), Hospital Anxiety and Depression Scale (HADS), and Short-Form Health Survey (SF-36) were collected using validated Arabic tools. All the analyses were performed with the significance level set at p < 0.05.
    Results: Out of the 37 participants, the frequency of anxiety and depression was 59.5% at admission, and it decreased to 40.5% at discharge from rehabilitation services. Functional independence improved significantly, with a 9.5-point increase in FIM scores. The mean differences (- 1.54 ± 4.3 p=0.03) and categorical differences between the initial and discharge HADS scores were significant (p=0.02).
    Conclusion: We found a high rate of depression and anxiety among stroke patients at admission. Rehabilitation services can lead to the improvement of depression and anxiety in stroke patients, from initial admission to discharge, with emotional health as a factor for better outcomes.

    Keywords: anxiety, depression, rehabilitation, stroke, functional independent measure, emotional health

    Introduction

    Stroke is a significant global health concern, affecting approximately 12.2 million individuals globally resulting in 6.6 million deaths in 2019.1 Recent data indicate that stroke prevalence has increased among both males and females with advancing age, with around 9.4 million Americans self-reporting a history of stroke between 2017 and 2020.2

    In Saudi Arabia, the prevalence of stroke shows considerable variation across studies, ranging from 29 to 57.6 per 100,000 populations.3,4 This disparity may reflect differences in study methodologies or population demographics. With an aging population, the incidence of stroke in Saudi Arabia is expected to rise further, increasing the burden on healthcare systems.5 Stroke can lead to significant physical, sensory, and cognitive impairments, often resulting in diminished quality of life.

    One of the most frequent and debilitating complications after stroke is post-stroke depression (PSD). PSD is associated with reduced functional ability, poorer quality of life, and increased mortality.6 Globally, PSD affects approximately 30% of stroke patients, persisting for up to five years’ post stroke.7,8 A recent systematic review estimated the prevalence of PSD to be around 27%.9 However, this prevalence has varied across regions; for example, it was 47% in Iran but 31% in Sub-Saharan Africa.10,11 Furthermore, post stroke anxiety (PSA) affects around 20 to 25% of stroke patients and can negatively impact the rehabilitation process and quality of life for patients.12,13 Depression and anxiety post stroke are highly related condition and can coexist in many stroke patients.14 In Saudi Arabia, data on PSD remain limited. Only two studies have reported PSD rates of 70.6% and 76.5%, but these studies had small sample sizes, did not follow up with participants, and were completed at one point only.15,16 Another study was conducted during the COVID-19 pandemic, and it reported a lower PSD prevalence of 36%, though the pandemic’s impact on mental health might have influenced these findings.17

    Mechanism that associated stroke with PSD are investigated with hypotheses related the cause to; stroke lesion site, amino acid neurotransmitters, and neuroinflammation.18 As for the stroke lesion location and its relation to PSD evidence suggested that specific brain regions such as the prefrontal cortex, limbic area, and basal ganglia can disturb the pathway which may lead to PSD.19 The monoamine neurotransmitters is considered the main biological factor that link PSD to stroke. If distributed occur at the hypothalamic-pituitary-adrenal axial, glutamate and gut-brain circuits this can lead to PSD.20,21 Lastly, studies proposed that elevated levels of inflammatory mediators are associated with PSD.22,23

    PSD contributes to increased physical disability, cognitive impairment, higher mortality risk, and a greater likelihood of falls.6,24,25 It also impedes rehabilitation progress, resulting in poorer quality of life and challenges in returning to work.17,25–27 Common risk factors for PSD include stroke severity, cognitive impairment, physical disability, and functional dependency.17,28,29 Patients with aphasia are at particularly high risk, with more severe aphasia correlating with more pronounced PSD symptoms.30,31 Additionally, a history of depression, psychiatric illness, and living alone further increase the likelihood of developing PSD.32

    This study aims to shed light on the current practice of stroke rehabilitation in Saudi Arabia and identify barriers that may hinder recovery—particularly post-stroke depression. Our objective is to track patients throughout their rehabilitation admission and at discharge to better understand how PSD evolves and impacts recovery.

    Although some studies in Saudi Arabia have reported on PSD, there is still a lack of data regarding its frequency and associated factors within rehabilitation settings. A deeper understanding of PSD and its predictors is crucial for designing individualized and effective interventions in neurorehabilitation. Furthermore, no prior study in the region has included a follow-up to examine changes in depression levels throughout the rehabilitation process. Therefore, this study investigated the prevalence of PSD and explored clinical factors linked to its occurrence. To our knowledge, this is the first study in Saudi Arabia—and the broader Middle East—to examine PSD and its associated factors within a neurorehabilitation context.

    Materials and Methods

    Study Design

    This predictive correlational feasibility study was conducted at the Neurorehabilitation Unit (NRU) of King Abdulaziz Medical City (KAMC) in Riyadh, Saudi Arabia, between October 1, 2023, and October 31, 2024, focusing on stroke patients admitted to the NRU with pre and post assessments of rehabilitation outcomes.

    A non-probability convenience sampling method was used to recruit adult stroke survivors aged 18 to 80 years who met the inclusion criteria Figure 1

    Figure 1 Flowchart of the recruitment process in the Neurorehabilitation Unit.

    Eligibility Criteria

    Patients were excluded if they had severe cognitive impairment (defined as a Montreal Cognitive Assessment [MoCA] score of ≤10), recurrent stroke or dementia diagnosed before the stroke, surgically treatable lesions on CT scans (eg, brain tumors), other central nervous system (CNS) conditions causing depression (eg, Parkinson’s disease), severe aphasia, severe comorbidities (eg, end-stage renal disease), or a prior history of psychiatric disturbances. Clinical data were extracted from electronic health records using the BestCare health record system to ensure accuracy and reliability.

    Ethical considerations were prioritized, with data collection initiated after obtaining Institutional Review Board (IRB) approval from the King Abdullah International Medical Research Center (KAIMRC) (Reference Number: SP34R/055/05). Informed consent was secured from patients and their families, ensuring confidentiality, privacy, and adherence to ethical research standards.

    Outcome Measurement

    All participants were assessed for the following: 1) stroke severity at the time of admission using the National Institute of Health Stroke Scale (NIHSS),33,34 2) functional independence at admission and discharge using the Functional Independence Measure (FIM), focusing on items such as eating, grooming, bathing, toileting, and upper and lower body dressing,35 3) self-reported depression and anxiety using the Hospital Anxiety and Depression Scale (HADS) at both admission and discharge,36 and 4) emotional and physical well-being at discharge using the 36-item Short Form Health Survey (SF-36).37 Questions related to the physical well-being asked about activities such as claiming stairs, walking. While mental well-being asked questions regarding the person’s such as if they feel clam, cheerful.

    Both the Hospital Anxiety and Depression Scale (HADS) and the 36-item Short Form Health Survey (SF-36) were administered using their validated Arabic versions. The Arabic-translated form of the HADS has been utilized in this study due to its validation across various medical settings.38,39 The HADS consists of an anxiety subscale (7 items, assessed on a 4-point Likert scale) and a depression subscale (7 items, assessed similarly). The Arabic version demonstrates strong internal consistency, with Cronbach’s alpha values ranging from 0.70 to 0.835. Scores for each subscale were calculated by summing the relevant items, with a maximum score of 21 for each subscale. A score of 0–7 is categorized as normal, 8–10 as mild, 11–14 as moderate, and 15–21 as severe for each subscale for depression and anxiety.40 Both the HADS and SF-36 initial assessment were administered in day 5 or 6 after admission, while the discharge assessments were completed after 20 days on average.

    Statistical Analysis

    Descriptive analyses were performed, presenting categorical variables as frequencies and percentages. A one-way ANOVA test was used to assess the association between depression levels, as measured using the HADS, and variables including age, length of stay, NIHSS, FIM, SF-36 physical health, and emotional health. Spearman’s rank correlation test was used to assess the correlation between emotional well-being and physical functioning with depression. The McNemar test was used to assess changes in HADS classifications between admission and discharge (pre–post assessment). Lastly, multivariable logistic regression was conducted to examine the association between HADS scores at discharge, adjusting for relevant covariates including age and NIHSS scores, with adjusted odd ratio (AORs) and 95% confidence intervals (CI). A p-value <0.05 was considered statistically significant. All the statistical analyses were performed using the Statistical Package for the Social Sciences (SPSS V.25.0, SPSS Inc., Chicago, IL).

    Results

    Participants Demographics

    A total of 37 participants were included in the current study. The most common exclusion criteria were low cognitive ability and aphasia. The majority of participants were male (62.2%), and 83.8% had ischemic stroke. Most participants had comorbidities, including hypertension (75.7%) and diabetes (73.0%). The average age of participants was 59 years, with an average length of stay of 22 days in the NRU. Participant characteristics are presented in Table 1.

    Table 1 Participants Characteristics

    Initial and Discharge Hospital Anxiety and Depression Scale & Short Form Health Survey Correlation with Hospital Anxiety and Depression Scale

    There was a statistically significant difference between the initial and discharge scores of the HADS (- 1.54 ± 4.3 p=0.03). The mean difference in total scores decreased by 1.54 at discharge (Table 2). In Addition, a Spearman correlation was conducted between the HADS scores at discharge and emotional and physical Short Form Health Survey SF-36. There was a moderate significant correlation between the SF-36 emotion and HADS at discharge, with the increase of emotional well-being correlated to decrease of HADS scores (p= 0.03, CI: −0.76, −0.17). Moreover, a weak negative non-significant correlation was found between SF-36 physical and HADS scores at discharge (Table 3).

    Table 2 Initial and Discharge Hospital Anxiety and Depression Scale

    Table 3 Spearman Correlation Between SF-36 and Discharge Hospital Anxiety and Depression Scale Scores

    Rate and Severity Level of Hospital Anxiety and Depression Scale

    The rate of depression or anxiety was 59.5% at admission among all participants, which decreased to 40.5% at discharge, with seven participants reporting no issues by the end of the rehabilitation sessions. In addition, the severity of depression and anxiety varies initially and at discharge with lower rate of severity at discharge (53.5% mild and 33.3% moderate in compared to 45.5% mild and 45.5% moderate cases) (Supplementary Table 1). To test the significant change in the proportion of patients with each category initially and at discharge we used a paired chi-square test) (Table 4). There was a significant difference between the HADS initial, and discharge based on the present of depression and anxiety and the severity levels.

    Table 4 Initial and Discharge Hospital Anxiety and Depression Scale Based on the Severity Level

    Functional Independent Measure Scores

    Scores of the FIM initial assessment to discharge was statistically significant (p= 0.001), with an increase of 9.5 points between the initial and discharge scores (mean initial= 17.76, mean discharge= 27.32).

    Factors Associated with Initial and Discharge Hospital Anxiety and Depression Scale Scores

    The HADS scores at both initial assessment and discharge were grouped into categories: normal (no depression or anxiety), mild, moderate, and severe depression or anxiety. There was no association between the initial HADS scores and other variables (age, FIM, and initial NIHSS) (Table 5). However, the SF-36 emotional health score was significantly associated with HADS scores at discharge (p = 0.01) (Table 6).

    Table 5 Association Between Initial Hospital Anxiety and Depression Scale Scores and Variables

    Table 6 Association Between Discharge Hospital Anxiety and Depression Scale Scores and Variable

    Factors Influencing Discharge Hospital Anxiety and Depression Scale Scores

    A binary logistic regression was used to determine factors that may increase the likelihood of depression or anxiety based on the HADS discharge scores. Table 7 illustrates the variables, showing that SF-36 emotional health scores significantly increased the likelihood of depression or anxiety (OR = 0.93, CI = 0.89–0.98, p= 0.01).

    Table 7 Variables Influencing the Hospital Anxiety and Depression Scale

    Discussion

    This study explored the frequency and severity of depression and anxiety among stroke patients admitted to a neurorehabilitation unit, alongside factors associated with these mental health outcomes at discharge. Consistent with the previous literature, we observed a high prevalence of mood disorders at admission, with 59.5% of patients exhibiting symptoms of depression and/or anxiety.24,41 This aligns with previous research that identified stroke survivors as being at an elevated risk of mood disorders due to the physical and psychological burden of the condition.27,28 The previous literature showed that the prevalence of PSD varies widely, ranging between 20% and 65%, depending on the population studied, the assessment measures used, and the definitions of depression applied.9,41,42 However, significant improvements were observed in anxiety and depression levels, with a reduction in the admission to discharge rate in our findings. Initially, most participants reported mild to moderate symptoms, which shifted to predominantly mild symptoms by discharge. Furthermore, functional independence measure scores showed significant improvement during rehabilitation. While no associations were found between initial HADS scores and variables such as age or stroke severity, emotional health, as measured by the SF-36, was significantly correlated with HADS scores at discharge.

    Our findings are particularly consistent with studies conducted in the Saudi Arabia context, where high rates of PSD have been reported. For example, Abuadas et al42 reported that more than two-thirds (70.6%) of a sample of stroke survivor patients in Saudi Arabia experienced depression, and Alharbi et al43 reported comparable results. These findings underscore the significant psychological burden experienced by stroke survivors in the region and highlight the importance of culturally contextualized research and interventions.

    Rehabilitation was associated with a statistically significant reduction in depression and anxiety symptoms, with the rate dropping to 40.5% at discharge. Most participants moved from moderate to mild severity levels, suggesting that structured neurorehabilitation can effectively mitigate mood disturbances. Improvements in functional independence likely contributed to this positive trend, with an average gain of 9.5 points from admission to discharge.

    This is in line with the work of a recent study that involved 1,440 stroke survivors and demonstrated that moderate- or high-intensity physical activity correlates with lower levels of depressive symptoms in stroke survivors. This suggests that non-pharmacological approaches, particularly those emphasizing physical and social engagement, may therefore play a pivotal role in emotional recovery.44

    Interestingly, our study found no significant association between initial HADS scores and clinical factors such as age, NIHSS, or initial FIM scores. This enhancement in physical functionality likely contributes to the reduction in depression and anxiety symptoms, as greater independence can foster a sense of accomplishment and control, which are critical for mental well-being., However, emotional health, as assessed by the SF-36, emerged as a predictor of depression and anxiety outcomes at discharge. Binary logistic regression analysis confirmed that higher SF-36 emotional well-being scores were associated with a lower likelihood of depressive and anxious symptoms. However, this finding should not be a surprise, given that the SF-36 emotional items have some similarity with the HADS items, which can cause correlation between both outcomes.

    Multiple studies reported that the rehabilitation program helps alleviate depressive symptoms, with better functional outcomes leading to improvement in PSD.45,46 However, evidence has illustrated that the efficiency of functional recovery for stroke patients with PSD is poorer than for those without depression. This suggests that PSD has an impact on functional recovery.47,48 In addition, the severity of depression measure via HADS can contribute to the functional outcomes with better functional outcomes in patients with mild or low severity scores.49

    In terms of clinical implications, our results emphasize the need for routine screening and the management of depression and anxiety in stroke patients, particularly during admission and discharge from rehabilitation programs. Implementing targeted psychological interventions, such as cognitive-behavioral therapy (CBT) or stress management techniques, alongside physical rehabilitation, could further enhance patient outcomes. Moreover, the significant role of emotional health as a predictor of mood disturbances underscores the value of integrating psychosocial support into stroke care plans. Moreover, it is recommended to train rehabilitation staff on how to identify signs of depression and or anxiety as well as the impact on mental well-being on the client’s performance in rehabilitation sessions.

    This study has some strengths: it examined patients at an early rehabilitation stage, followed up with participants pre and post rehabilitation, and administrated self-reported assessments at two points in time.

    Limitation

    This study also has several limitations. The small sample size (n=37) limits the generalizability of its findings. The use of a non-probability convenience sampling method may have introduced selection bias. The study’s strict inclusion criteria contributed to the small sample size and excluded stroke patients who may have PSD. The correlation between SF-36 emotion and HADS scores can overlap; the similarity of the items in both assessments may limit the impact of the findings. Another limitation is the lack of consideration of participants past medical and mental health history, social support networks, medication use (including antidepressants), lesion location, or stroke laterality, all of which are known to influence the development and severity of post-stroke depression, which could have influenced the rate and severity of depression and anxiety. Furthermore, this study was conducted in a single tertiary rehabilitation center, which may have limited the generalizability of the findings across other healthcare settings, including rural or non-specialized facilities. Future research should include larger, more diverse samples and adopt longitudinal designs to better understand the temporal dynamics of mood disorders in stroke survivors.

    Past medical history can play an important part in the results, for example insomnia and the history of lack of sleep are linked to PDS. Evidence supports the link that pre-stroke insomnia may predict PSD.50,51 Moreover, patients with insomnia are more likely to have a more depression symptoms post stroke.52,53 Thus, future studies should explore the association between insomnia pre, post stroke and PSD in the Saudi population.

    Our results suggest the importance of screening stroke patients for depression and anxiety prior to rehabilitation to optimize stroke recovery in patients in Saudi Arabia. In addition, these results support enhancing the rehabilitation team’s awareness of depression and/or anxiety for early identification and potential referral to a specialist.

    Conclusion

    In conclusion, this study highlights the high prevalence of depression and anxiety among stroke patients upon admission and the significant improvements associated with inpatient rehabilitation. Emotional health emerged as a key factor influencing mental health outcomes, reinforcing the need for integrated, holistic approaches to stroke care. Routine screening, early intervention, and enhanced team awareness are recommended to optimize both emotional and functional recovery.

    Abbreviations

    PSD, Post-Stroke Depression; PSA, Post Stroke Anxiety; NRU, Neurorehabilitation Unit; NIHSS, National Institute of Health Stroke Scale; FIM, Functional Independence Measure; SF-36, 36-item Short Form Health Survey; HDAS, Hospital Anxiety and Depression Scale; CBT, Cognitive-Behavioral Therapy.

    Data Sharing Statement

    The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

    Ethical Approval and Consent to Participation

    All methods were performed in accordance with the ethical standards established in the Declaration of Helsinki and its subsequent amendments or comparable ethical standards. This study was approved with approval and consent of the Ethics Committee of King Abdullah International Medical Research Center (IRB approval number: SP34R/055/05). Informed consent was obtained from all participants before their involvement in the study, ensuring that they were fully aware of the study’s purpose, procedures, potential risks, and benefits. Informed consent was obtained from all subjects involved in the study.

    Acknowledgments

    The authors thank all the patients, physical therapists, and occupational therapists who contributed to the data collection.

    Funding

    This research received no external funding.

    Disclosure

    The authors report no conflicts of interest in this work.

    References

    1. Feigin VL, Stark BA, Johnson CO, et al. Global, regional, and national burden of stroke and its risk factors, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol. 2021;20(10):795–820. doi:10.1016/S1474-4422(21)00252-0

    2. Bryan S, Afful J, Carroll M, et al. NHSR 158. In: National Health and Nutrition Examination Survey 2017–March 2020 Pre-Pandemic Data Files. Hyattsville, MD; 2021.

    3. Alhazzani AA, Mahfouz AA, Abolyazid AY, et al. Study of Stroke Incidence in the Aseer Region, Southwestern Saudi Arabia. Int J Environ Res Public Health. 2018;15(2):215. doi:10.3390/ijerph15020215

    4. Alqahtani BA, Alenazi AM, Hoover JC, et al. Incidence of stroke among Saudi population: a systematic review and meta-analysis. Neurological Sci. 2020;41(11):3099–3104. doi:10.1007/s10072-020-04520-4

    5. Basri R, Issrani R, Hua Gan S, Prabhu N, Khursheed Alam M. Burden of stroke in the Kingdom of Saudi Arabia: a soaring epidemic. Saudi Pharm J. 2021;29(3):264–268. doi:10.1016/j.jsps.2021.02.002

    6. Towfighi A, Ovbiagele B, El Husseini N, et al. Poststroke Depression: a Scientific Statement for Healthcare Professionals From the American Heart Association/American Stroke Association. Stroke. 2017;48(2):e30–e43. doi:10.1161/STR.0000000000000113

    7. Hackett ML, Pickles K. Part I: frequency of depression after stroke: an updated systematic review and meta-analysis of observational studies. Int J Stroke. 2014;9(8):1017–1025. doi:10.1111/ijs.12357

    8. Ayerbe L, Ayis S, Wolfe CDA, Rudd AG. Natural history, predictors and outcomes of depression after stroke: systematic review and meta-analysis. Br J Psychiatry. 2013;202(1):14–21. doi:10.1192/bjp.bp.111.107664

    9. Liu L, Xu M, Marshall IJ, Wolfe CDA, Wang Y, O’Connell MD. Prevalence and natural history of depression after stroke: a systematic review and meta-analysis of observational studies. PLoS Med. 2023;20(3):e1004200–e1004200. doi:10.1371/journal.pmed.1004200

    10. Dalvand S, Gheshlagh RG, Kurdi A. Prevalence of poststroke depression in Iranian patients: a systematic review and meta-analysis. Neuropsychiatr Dis Treat. 2018;14:3073–3080. doi:10.2147/NDT.S184905

    11. Ojagbemi A, Akpa O, Elugbadebo F, Owolabi M, Ovbiagele B. Depression after Stroke in Sub-Saharan Africa: a Systematic Review and Meta-Analysis. Behav Neurol. 2017;2017:4160259. doi:10.1155/2017/4160259

    12. Knapp P, Dunn-Roberts A, Sahib N, et al. Frequency of anxiety after stroke: an updated systematic review and meta-analysis of observational studies. Int J Stroke. 2020;15(3):244–255. doi:10.1177/1747493019896958

    13. Donnellan C, Hickey A, Hevey D, O’Neill D. Effect of mood symptoms on recovery one year after stroke. Int J Geriatr Psychiatry. 2010;25(12):1288–1295. doi:10.1002/gps.2482

    14. Li X, Wang X. Relationships between stroke, depression, generalized anxiety disorder and physical disability: some evidence from the Canadian Community Health Survey-Mental Health. Psychiatry Res. 2020;290:113074. doi:10.1016/j.psychres.2020.113074

    15. Abuadas FH, Ayasrah SM, Ahmad MM, Abu‐Snieneh HM, Obiedallah HF, Basheti IA. Prevalence of depression and its associated factors among stroke survivors in Saudi Arabia. Nursing Open. 2023;10(3):1629–1638. doi:10.1002/nop2.1417

    16. Al-Busaidi IS, Alamri Y. Poststroke Anxiety and Depression: findings from Saudi Arabia. J Stroke Cerebrovascular Dis. 2016;25(7):1653–1654. doi:10.1016/j.jstrokecerebrovasdis.2016.03.003

    17. Ahmed ZM, Khalil MF, Kohail AM, Eldesouky IF, Elkady A, Shuaib A. The Prevalence and Predictors of Post-Stroke Depression and Anxiety During COVID-19 Pandemic. J Stroke Cerebrovascular Dis. 2020;29(12):105315. doi:10.1016/j.jstrokecerebrovasdis.2020.105315

    18. Zhan Q, Kong F. Mechanisms associated with post-stroke depression and pharmacologic therapy. Front Neurol. 2023;14:1274709. doi:10.3389/fneur.2023.1274709

    19. Robinson RG, Jorge RE. Post-stroke depression: a review. Am J Psychiatry. 2016;173(3):221–231. doi:10.1176/appi.ajp.2015.15030363

    20. Sun M, Chen H, Dong S, Zhang G, Zhou X, Cheng H. Alteration of gut microbiota in post-stroke depression patients with Helicobacter pylori infection. Neurobiol Dis. 2024;193:106458. doi:10.1016/j.nbd.2024.106458

    21. Gao H, Sai Y, Shang R, et al. Post stroke depression: pathogenesis and molecular mechanisms of natural product-based interventions. Front Pharmacol. 2025;16:1595160. doi:10.3389/fphar.2025.1595160

    22. Korostynski M, Hoinkis D, Piechota M, et al. Toll-like receptor 4-mediated cytokine synthesis and post-stroke depressive symptoms. Transl Psychiatry. 2021;11(1):246. doi:10.1038/s41398-021-01359-x

    23. Li R, Fan W, Li D, Liu X. Correlation of common inflammatory cytokines with cognition impairment, anxiety, and depression in acute ischemic stroke patients. Braz J Med Biol Res. 2022; 55:e11517.

    24. Robinson RG, Jorge RE, Starkstein SE. Poststroke Depression: an Update. J Neuropsychiatry Clin Neurosci. 2024;36(1):22–35. doi:10.1176/appi.neuropsych.21090231

    25. Lenzi GL, Altieri M, Maestrini I. Post-stroke depression. Revue neurologique. 2008;164(10):837–840. doi:10.1016/j.neurol.2008.07.010

    26. Trusova NA, Levin OS. Clinical significance and possibilities of therapy of post-stroke depression. Zhurnal nevrologii i psikhiatrii imeni SS Korsakova. 2019;119(9. Vyp. 2):60–67. doi:10.17116/jnevro201911909260

    27. Shewangizaw S, Fekadu W, Gebregzihabhier Y, Mihretu A, Sackley C, Alem A. Impact of depression on stroke outcomes among stroke survivors: systematic review and meta-analysis. PLoS One. 2023;18(12):e0294668–e0294668. doi:10.1371/journal.pone.0294668

    28. Babkair LA. Risk Factors for Poststroke Depression: an Integrative Review. J Neurosci Nurs. 2017;49(2):73–84. doi:10.1097/JNN.0000000000000271

    29. De Ryck A, Fransen E, Brouns R, et al. Poststroke depression and its multifactorial nature: results from a prospective longitudinal study. J Neurol Sci. 2014;347(1–2):159–166. doi:10.1016/j.jns.2014.09.038

    30. Zanella C, Laures-Gore J, Dotson VM, Belagaje SR. Incidence of post-stroke depression symptoms and potential risk factors in adults with aphasia in a comprehensive stroke center. Topic Stroke Rehabil. 2023;30(5):448–458. doi:10.1080/10749357.2022.2070363

    31. Kao S-K, Chan C-T. Increased risk of depression and associated symptoms in poststroke aphasia. Sci Rep. 2024;14(1):21352. doi:10.1038/s41598-024-72742-z

    32. De Ryck A, Fransen E, Brouns R, et al. Psychosocial problems associated with depression at 18 months poststroke. International Journal of Geriatric Psychiatry. 2014;29(2):144–152. doi:10.1002/gps.3974

    33. Spilker J, Kongable G, Barch C, et al. Using the NIH Stroke Scale to assess stroke patients. The NINDS rt-PA Stroke Study Group. J Neurosci Nurs. 1997;29(6):384–392. doi:10.1097/01376517-199712000-00008

    34. Brott T, Adams HP, Olinger CP, et al. Measurements of acute cerebral infarction: a clinical examination scale. Stroke. 1989;20(7):864–870. doi:10.1161/01.STR.20.7.864

    35. Heinemann AW, Linacre JM, Wright BD, Hamilton BB, Granger C. Relationships between impairment and physical disability as measured by the functional Independence measure. Arch Phys Med Rehabil. 1993;74(6):566–573. doi:10.1016/0003-9993(93)90153-2

    36. Zigmond AS, Snaith RP. The Hospital Anxiety and Depression Scale. Acta psychiatrica Scandinavica. 1983;67(6):361–370. doi:10.1111/j.1600-0447.1983.tb09716.x

    37. Ware JE, Sherbourne CD. The MOS 36-item short-form health survey (SF-36). I. Conceptual framework and item selection. Med Care. 1992;30(6):473–483. doi:10.1097/00005650-199206000-00002

    38. Al Aseri ZA, Suriya MO, Hassan HA, et al. Reliability and validity of the Hospital Anxiety and Depression Scale in an emergency department in Saudi Arabia: a cross-sectional observational study. BMC Emergency Medicine. 2015;15(1):28. doi:10.1186/s12873-015-0051-4

    39. Al-Azzam S, Alzoubi KH, Ayoub NM, et al. An audit on public awareness of depression symptoms in Jordan. Int J Occup Med Environ Health. 2013;26(4):545–554. doi:10.2478/s13382-013-0128-9

    40. Stern AF. The Hospital Anxiety and Depression Scale. Occupational Med. 2014;64(5):393–394. doi:10.1093/occmed/kqu024

    41. De Ryck A, Brouns R, Geurden M, Elseviers M, De Deyn PP, Engelborghs S. Risk Factors for Poststroke Depression. J Geriatric Psychiatry Neurol. 2014;27(3):147–158. doi:10.1177/0891988714527514

    42. Abuadas FH, Ayasrah SM, Ahmad MM, Abu-Snieneh HM, Obiedallah HF, Basheti IA. Prevalence of depression and its associated factors among stroke survivors in Saudi Arabia. Nursing Open. 2023;10(3):1629–1638.

    43. Alharbi NA, Aydan NA, Alhamzah SA. Prevalence of Poststroke Depression among Saudi Patients in Tertiary Medical Centers: a Cross-Sectional Study. Journal of Nature and Science of Medicine. 2023;6(2):77–83. doi:10.4103/jnsm.jnsm_120_22

    44. Wang Y, Chen J, Zou Y, et al. Relationship between physical activity and depressive symptoms in stroke survivors: a cross-sectional study of 1,140 individuals. J Rehabil Med. 2025;57:jrm41272–jrm41272.

    45. Butsing N, Zauszniewski JA, Ruksakulpiwat S, Griffin MTQ, Niyomyart A. Association between post-stroke depression and functional outcomes: a systematic review. PLoS One. 2024;19(8):e0309158. doi:10.1371/journal.pone.0309158

    46. Khanna M, Sivadas D, Gupta A, Haldar P, Prakash NB. Impact of inpatient rehabilitation on quality of life among stroke patients. J Neurosci Rural Pract. 2022;13(4):800–803. doi:10.25259/JNRP-2022-1-18-R1-(2322)

    47. Wada Y, Otaka Y, Yoshida T, et al. Effect of Post-stroke Depression on Functional Outcomes of Patients With Stroke in the Rehabilitation Ward: a Retrospective Cohort Study. Arch Rehabil Res Clin Transl. 2023;5(4):100287. doi:10.1016/j.arrct.2023.100287

    48. Sharma GS, Gupta A, Khanna M, Prakash NB. Post-Stroke Depression and Its Effect on Functional Outcomes during Inpatient Rehabilitation. Journal of Neurosciences in Rural Practice. 2021;12:543. doi:10.1055/s-0041-1731958

    49. Mazzeo S, Pancani S, Sodero A, et al. Depressive Symptoms Moderate the Association Between Functional Level at Admission to Intensive Post-Stroke Rehabilitation and Effectiveness of the Intervention. J Geriatr Psychiatry Neurol. 2024;37(3):222–233. doi:10.1177/08919887231204543

    50. Dong L, Brown DL, Chervin RD, Case E, Morgenstern LB, Lisabeth LD. Pre-stroke sleep duration and post-stroke depression. Sleep Med. 2021;77:325–329. doi:10.1016/j.sleep.2020.04.025

    51. Suh M, Choi-Kwon S, Kim JS. Sleep disturbances after cerebral infarction: role of depression and fatigue. J Stroke Cerebrovasc Dis. 2014;23(7):1949–1955. doi:10.1016/j.jstrokecerebrovasdis.2014.01.029

    52. Geusgens CAV, van Tilburg DCH, Fleischeuer B, Bruijel J. The relation between insomnia and depression in the subacute phase after stroke. Neuropsychological Rehabilitat. 2025;35(4):757–773. doi:10.1080/09602011.2024.2370072

    53. Wang L, Tao Y, Chen Y, Wang H, Zhou H, Fu X. Association of post stroke depression with social factors, insomnia, and neurological status in Chinese elderly population. Neurol Sci. 2016;37(8):1305–1310. doi:10.1007/s10072-016-2590-1

    Continue Reading

  • Two Supply Chain Reports Clash over what Fingerprint Technology the 2026 Foldable iPhone will Adapt – patentlyapple.com

    1. Two Supply Chain Reports Clash over what Fingerprint Technology the 2026 Foldable iPhone will Adapt  patentlyapple.com
    2. Apple’s big iPhone launch is coming on September 9. What to expect  CNN
    3. Apple to Kick Off Three-Year Plan to Reinvent Its Iconic iPhone  Bloomberg.com
    4. iPhone 17 Air: Will a Slim Model Replace the iPhone 17 Plus?  CNET
    5. Kuo reiterates Touch ID in the iPhone Fold; unlikely to be in-display  9to5Mac

    Continue Reading

  • CEO who snatched boy’s hat at US Open says he made ‘huge mistake’

    CEO who snatched boy’s hat at US Open says he made ‘huge mistake’

    The man who was caught on camera snatching a hat off a young boy at the US Open has said he made a “huge mistake” after footage of the incident went viral.

    Piotr Szczerek, a Polish chief executive of a paving firm, said he was “convinced” tennis star Kamil Majchrzak had been “passing his hat in my direction”.

    “I know I did something that seemed like consciously collecting a memento from a child,” he wrote in a statement. “This wasn’t my intention, but it doesn’t change the fact that I hurt the boy and disappointed the fans.”

    The video, taken during Majchrzak’s match on Thursday, showed the tennis player offering his cap to a child, before Mr Szczerek appears to take it.

    Versions of the clip were shared widely on social media and prompted harsh criticism of Mr Szczerek’s actions.

    The CEO wrote on social media on Monday: “I would like to unequivocally apologise to the injured boy, his family, as well as all the fans and the player himself.”

    He added that he had given the hat back to the boy, and hoped that it had “at least partially repaired the damage that was done”.

    Majchrzak, who had just won his match against Russian ninth seed Karen Khachanov when the incident unfolded, told the New York Post on Saturday that “obviously it was some kind of confusion”.

    “I was pointing, giving the hat, but I had a lot going on after my match, after being super tired and super excited for the win,” he said.

    “I just missed it. I had like a dead look, if you know what I mean. I’m sure the guy was also acting in the moment of heat, in the moment of emotions.”

    The tennis star reunited with the boy over the weekend, sharing clips of him giving the young fan a cap and other merchandise on Instagram.

    “Today after warm up, I had a nice meeting,” the tennis pro wrote, adding: “Do you recognise [the cap]?”

    Majchrzak, ranked 76th in the world in men’s singles, came back from two sets down to beat Khachanov in a second-round match at Flushing Meadows, but was forced to retire injured during the first set of his third-round tie against Switzerland’s Leandro Riedi on Saturday.

    He later confirmed he had torn an intercostal muscle.

    Continue Reading

  • Association between dynamic changes in the triglyceride-glucose index and prognosis in patients with acute ST-segment elevation myocardial infarction | Cardiovascular Diabetology

    Association between dynamic changes in the triglyceride-glucose index and prognosis in patients with acute ST-segment elevation myocardial infarction | Cardiovascular Diabetology

    Since the Framingham Heart Study, traditional risk factors for coronary heart disease (CHD)—including hypertension, diabetes mellitus, dyslipidemia, smoking, and obesity—have been widely recognized [21,22,23,24,25]. However, one study evaluated whether the 256 clinical trials cited by current international guidelines (from the European Society of Cardiology [ESC] and the American College of Cardiology/American Heart Association [ACC/AHA]), drawn from a pool of 1,133 studies, reported the proportion of patients with each cardiovascular risk factor, such as hypertension, smoking, hypercholesterolemia, and diabetes. The results showed that among the 256 clinical trials, none explicitly reported the proportion of patients without traditional cardiovascular risk factors [26]. This highlights the fact that CHD patients lacking conventional risk factors have been underrepresented in both guidelines and clinical trials, posing significant challenges for their management in secondary prevention. As most traditional cardiovascular secondary prevention drugs, such as statins and ACEI/ARB, are designed to target specific risk factors, their efficacy may be limited in patients without such conditions. In real-world practice, a substantial number of patients still face residual cardiovascular risk and remain susceptible to cardiovascular events despite standard therapy [27,28,29,30].

    Insulin resistance (IR) refers to reduced sensitivity of tissues to insulin, and is considered the central mechanism underlying the development of type 2 diabetes, often existing years before clinical diagnosis [31]. Studies have shown that the severity of insulin resistance exerts a significant and independent negative effect on myocardial mechanical efficiency, even in non-diabetic individuals, highlighting the potential role of IR in cardiovascular disease, especially among populations without diabetes. Regardless of diabetic status, IR and its accompanying metabolic disturbances can promote the onset and progression of CHD [32]. Individuals with IR are more prone to hyperglycemia, dyslipidemia, and hypertension, all of which are closely associated with adverse cardiovascular outcomes [33]. Therefore, IR is not only regarded as a potential pathophysiological mechanism of cardiovascular disease but also serves as an important marker for cardiovascular risk assessment. Furthermore, IR plays a particularly critical role in metabolic syndrome, being strongly associated with obesity, hypertension, hypertriglyceridemia, and low levels of high-density lipoprotein cholesterol [34, 35]. Notably, these metabolic abnormalities have been confirmed as independent risk factors for cardiovascular disease (CVD) [36,37,38], further underscoring the pivotal role of IR in the development and progression of CVD.

    Due to the complexity and high cost of traditional IR assessment methods, researchers have proposed the triglyceride-glucose (TyG) index as a convenient and cost-effective surrogate marker [39, 40]. The TyG index does not rely on insulin measurements and is applicable to a wide range of populations, offering significant clinical value for metabolic health assessment and cardiovascular risk prediction [41].

    In individuals with normal glucose metabolism, activation of insulin receptors triggers signaling to IRS-1 (insulin receptor substrate-1), which then interacts with and activates the downstream PI3-kinase (PI3K, a p85/p110 complex). This leads to activation of Akt, promoting the translocation of GLUT4 to the cell membrane and facilitating glucose uptake into cells for metabolism. Additionally, this pathway activates nitric oxide synthase (NOS), promoting vasodilation and maintaining endothelial function. In patients with hyperglycemia, impaired IRS-1 signaling results in reduced glucose metabolism and decreased eNOS activity, thereby impairing vasodilation. Meanwhile, the MAP kinase pathway remains sensitive to insulin. The insulin resistance of the IRS-1/PI3K pathway leads to compensatory hyperinsulinemia, which in turn excessively activates the MAPK pathway, promoting inflammation, cellular proliferation, and the progression of atherosclerosis. Dyslipidemia contributes to this process by triggering inflammatory storms mediated by NF-κB and inhibiting IRS-1 function, thus forming a vicious cycle between insulin resistance and atherosclerosis. As an integrated marker of glucose and lipid metabolism, the TyG index reflects the metabolic burden resulting from the combined dysregulation of triglycerides and fasting glucose. Compared to individual metabolic parameters, the TyG index more sensitively captures abnormal signals arising from the interaction between glucose and lipid metabolism. In recent years, studies have suggested that dynamic monitoring of biomarkers offers substantial advantages over single time-point measurements. Considering that triglyceride and fasting glucose levels may fluctuate over time, potentially leading to regression dilution bias, this study identified three distinct TyG trajectories in acute STEMI patients following PCI, based on serial TyG values: the persistently high group (trajectory 1), the moderate-level group (trajectory 2), and the rapid decline group (trajectory 3). The incidence of major adverse cardiovascular events (MACE) differed significantly among the three groups (P < 0.05). Compared with the persistently high group, the rapid decline group exhibited significantly lower rates of ischemia-driven target vessel revascularization, heart failure rehospitalization, nonfatal acute myocardial infarction, cardiac death, and total MACE events (all P < 0.05). Survival analysis demonstrated that the prognostic endpoints significantly differed among the groups, as indicated by the survival curves (P for log-rank < 0.001). To further explore the prognostic impact of different metabolic trajectories within various clinical subgroups, a subgroup analysis was performed. The persistently high group exhibited a consistently higher risk of major adverse cardiovascular events (MACE) across most subgroups, with statistically significant differences compared to the rapid decline group. This association was particularly pronounced among patients with multivessel disease and those classified as Killip class III–IV, suggesting that metabolic trajectories hold important prognostic value, especially for risk stratification in high-risk patients. Furthermore, multivariate Cox regression analysis, after adjusting for confounding variables including sex, age, low-density lipoprotein cholesterol, uric acid, body mass index (BMI), hypertension, diabetes mellitus, family history of coronary heart disease, smoking history, multivessel disease, ejection fraction, Killip classification, as well as baseline and final TyG values, confirmed that the trajectory grouping remained an independent prognostic factor (P < 0.05). These findings indicate that the predictive value of TyG trajectories for long-term prognosis in STEMI patients remains robust across different adjustment models, highlighting its independent prognostic significance. In addition to STEMI, the TyG index has also been reported as a valuable risk predictor in various cardiovascular diseases, including hypertension, heart failure, and atherosclerosis. Existing evidence preliminarily supports the potential of the TyG index as a metabolic biomarker for clinical risk assessment. In the following section, we will systematically summarize the current research progress on the TyG index across different cardiovascular diseases.

    TyG index and hypertension: from risk prediction to target organ damage and clinical outcomes

    In a longitudinal study of hypertension involving 4,600 Chinese adults, analysis showed that for each 1.0-unit increase in the TyG index, the risk of developing new-onset hypertension increased by 17%. Long-term follow-up further identified three distinct TyG index trajectories: the “low-level rising group,” the “moderate-level rising group,” and the “high-level stable group.” Compared with the “low-level rising group,” the risk of hypertension was significantly higher in the other two groups (HR = 1.38, 95% CI: 1.23–1.54; HR = 1.69, 95% CI: 1.40–2.02), with the risk being particularly pronounced in the female subgroup (HR = 2.63 and 4.66) [42]. Moreover, coronary heart disease patients with concomitant hypertension and elevated TyG index showed a 47% increased risk of MACE within one year [43, 44]. The TyG index is also significantly associated with the incidence of albuminuria, vascular injury, and cardiac remodeling in hypertensive patients. Patients with higher TyG index levels were found to have significantly increased left atrial volume index and left ventricular mass index, along with reduced E′ velocity and E/A ratio (P < 0.05). Multivariate regression analysis further confirmed that this association was independent of other risk factors (P < 0.001) [45, 46]. A retrospective study involving 4,710 patients demonstrated that individuals with persistently elevated and poorly controlled TyG index levels had a significantly increased risk of stroke (OR = 2.161, 95% CI: 1.446–3.228) [47]. The TyG index has been identified as an independent risk factor for stroke [48], and in hypertensive patients, the risk of first-ever stroke increases with higher TyG index levels. This association was especially prominent among elderly individuals (HR = 2.15, 95% CI: 1.50–3.07, P < 0.001) [49]. Further supporting evidence comes from a multicenter analysis conducted across 22 hospitals in Suzhou, which included 3,216 patients with acute ischemic stroke. The study revealed that a higher TyG index was closely associated with an increased risk of poor functional outcomes at discharge and higher in-hospital mortality, highlighting its potential value in short-term prognostic assessment [50]. Additionally, a study involving 356 hospitalized patients showed that the TyG index was significantly higher in the atrial fibrillation group compared to the non-atrial fibrillation group, and the TyG index was identified as an independent risk factor for atrial fibrillation. Subgroup analysis revealed that the association between the TyG index and atrial fibrillation was more evident in non-diabetic patients, whereas this correlation was not observed in diabetic individuals [51].

    The relationship between TyG index and heart failure risk, and prognosis

    Beyond hypertension, the TyG index has also been shown to be closely associated with the incidence of heart failure. Analyses from the Kailuan prospective cohort, which included 95,996 participants, and a retrospective Hong Kong cohort involving 19,345 participants — with 2,726 and 1,709 heart failure cases identified respectively — demonstrated that the TyG index is an independent risk factor for heart failure [52, 53]. Other studies have reported that a higher TyG index is significantly associated with increased risks of mortality and rehospitalization among patients with heart failure with preserved ejection fraction (HFpEF), suggesting that the TyG index could serve as a promising prognostic marker for this patient population [54]. In patients with acute decompensated heart failure, an elevated TyG index was significantly linked to an increased risk of all-cause mortality, cardiovascular death, and major adverse cardiovascular and cerebrovascular events (MACCEs). Particularly, patients with a TyG index ≥ 9.32 were at markedly higher risk for both mortality and MACCEs [55]. Among patients with chronic heart failure, those with a higher TyG index also exhibited significantly increased risks of all-cause and cardiovascular mortality. Specifically, patients with elevated TyG index had a 1.84-fold higher risk of all-cause death and a 1.94-fold higher risk of cardiovascular death compared to those with lower values. Moreover, incorporating the TyG index into existing risk prediction models significantly improved their predictive performance, further underscoring its critical role as a prognostic indicator for mortality in chronic heart failure patients [56,57,58].

    The association between TyG index and arteriosclerosis

    Arteriosclerosis, an early manifestation of vascular aging, is characterized primarily by reduced arterial elasticity and elevated pulse pressure, and it is closely associated with an increased risk of cardiovascular disease. A growing body of evidence has demonstrated a positive correlation between the TyG index and arteriosclerosis, as measured by parameters such as pulse wave velocity (PWV), and has suggested that the TyG index may outperform traditional insulin resistance measures like the HOMA-IR index in predicting arteriosclerosis. Lambrinoudaki et al. reported a positive association between the TyG index and brachial-ankle pulse wave velocity (baPWV), although their study population was limited to postmenopausal women [59]. Similarly, a Korean study confirmed an independent positive correlation between the TyG index and baPWV and showed that the TyG index was superior to HOMA-IR for predicting arteriosclerosis [60]. Li et al. further demonstrated that this association was particularly pronounced in men [61], whereas Nakagomi et al. found the correlation to be stronger in women [62], suggesting that sex and age differences may influence this relationship. Additionally, Wu et al. confirmed that the TyG index was correlated with changes in baPWV among hypertensive patients, indicating a possible synergistic effect of insulin resistance and hypertension in promoting the development of arteriosclerosis [63, 64]. Wang et al. reported, in a cohort of 3,185 patients with type 2 diabetes, that the TyG index was positively correlated with arteriosclerosis and again showed superior predictive value compared to HOMA-IR [65]. Guo et al. further demonstrated that the TyG index was significantly and positively associated with the 10-year cardiovascular disease risk in patients with arteriosclerosis [66]. These findings collectively highlight the potential of the TyG index as a predictive marker for arteriosclerosis and provide valuable guidance for clinical management.

    The clinical significance of the TyG index in predicting cardiovascular disease and All-Cause mortality

    The TyG index has shown significant value in predicting the development, severity, and prognosis of coronary artery disease (CAD). A meta-analysis including 12 cohort studies with 6,354,990 participants confirmed that higher TyG levels are associated with increased risks of CAD (moderate certainty), myocardial infarction, and cardiovascular disease (very low certainty). A linear relationship was observed between the TyG index and the risk of CAD and composite cardiovascular events, though further prospective studies are needed, especially in non-Asian populations [67]. Additionally, an elevated TyG index is closely related to coronary lesion complexity (SYNTAX score > 22) in ACS patients, independent of diabetes status [68, 69]. In a 10-year cohort of 6,095 non-diabetic subjects, higher TyG quartiles were linked to a significantly increased risk of CVD, CHD, and stroke, and the inclusion of the TyG index improved the predictive performance of traditional risk models [70]. NHANES-based studies also revealed that the TyG index is positively associated with chest pain and nonlinearly linked to all-cause mortality, independent of chest pain status [71]. Moreover, the TyG index outperformed other markers in predicting long-term mortality in non-diabetic STEMI patients, particularly when TyG ≥ 9.83 [72]. The TyG index also correlates with coronary lesion severity, abdominal aortic calcification [73, 74]. Furthermore, a large NHANES-based study (20,194 participants) demonstrated that the TyG index is nonlinearly associated with all-cause and cardiovascular mortality, particularly in individuals under 65 years. Compared to HOMA-IR, the TyG index showed superior predictive power for mortality outcomes [75].

    The clinical value of the TyG index in predicting complications after PCI in coronary artery disease patients

    Studies based on the MIMIC-III database have suggested that the TyG index holds potential value for predicting complications following percutaneous coronary intervention (PCI) ([76]. Several studies have demonstrated that the TyG index is associated with an increased risk of in-stent restenosis and repeated revascularization, and has been identified as an independent predictor of major adverse cardiovascular events (MACE) in patients with premature coronary artery disease. A study [77, 80] conducted by Fuwai Hospital in 2017 investigated the relationship between the TyG index, repeated revascularization, and in-stent restenosis in patients with chronic coronary syndrome who underwent drug-eluting stent implantation. A total of 1,414 patients were consecutively enrolled and followed for a median of 60 months. During follow-up, 548 patients (38.76%) experienced at least one major endpoint event. The incidence of major endpoints increased progressively with higher TyG index levels. After adjusting for potential confounders, the TyG index remained independently associated with major adverse outcomes in patients with chronic coronary syndrome (HR: 1.191, 95% CI: 1.038–1.367, P = 0.013) ([77,78,79], These findings suggest that an elevated TyG index is associated with a higher risk of post-PCI complications, including repeated revascularization and in-stent restenosis. However, its incremental predictive value for major adverse events remains limited, highlighting the need for further multicenter and large-scale clinical studies to clarify the role of the TyG index in long-term risk stratification and prognostic management after PCI ([81, 82].

    As a surrogate marker of insulin resistance (IR), the triglyceride-glucose (TyG) index is closely associated with the onset and progression of the aforementioned diseases. Accumulating evidence has shown that the TyG index serves as a reliable predictor for cardiovascular outcomes in various populations. Notably, several nomogram-based models incorporating the TyG index have demonstrated good predictive performance for MACEs in patients with HFpEF post-CABG, new-onset hypertension, STEMI undergoing PCI, and chronic coronary disease. These models consistently reported satisfactory C-index values (ranging from 0.73 to 0.82), improved net reclassification, and clinical utility via DCA, indicating the wide applicability of TyG in cardiovascular risk prediction I [83,84,85,86].

    From a systemic pathological perspective, metabolic dysregulation caused by insulin resistance involves multiple tissues and signaling pathways. The underlying mechanisms mainly involve endothelial dysfunction, chronic inflammatory responses, and oxidative stress. Under physiological conditions, insulin regulates blood flow and glucose metabolism by promoting nitric oxide (NO) production in vascular endothelial cells. However, in the IR state, NO synthesis is reduced, impairing vascular relaxation function [33]. In addition, IR can induce the sustained release of pro-inflammatory cytokines, such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and C-reactive protein (CRP), as well as reactive oxygen species (ROS). These processes mutually reinforce each other, forming a vicious cycle of “inflammation-oxidative stress,” which further aggravates endothelial injury [87, 88]. Meanwhile, a persistently elevated TyG index may also disrupt insulin secretion, promote myocardial fibrosis and left ventricular hypertrophy, ultimately leading to cardiac dysfunction [89]. These pathological changes not only impair myocardial structure and function but may also destabilize coronary plaques and increase the risk of thrombosis. Furthermore, studies have shown that IR and hyperinsulinemia can activate serum/glucocorticoid-regulated kinase 1 (SGK1) through multiple signaling pathways and stimulate aldosterone secretion, which in turn further activates SGK1. As a shared node in insulin and aldosterone signaling, SGK1 increases intracellular sodium ion concentration in vascular smooth muscle cells by activating sodium channels, while inhibiting nitric oxide synthase activity and reducing NO production. This cascade ultimately leads to increased vascular tone, endothelial dysfunction, and vascular stiffening [33]. In addition, the TyG index may contribute to the development of coronary artery disease (CAD) through its roles in dyslipidemia, diabetes, and hypertension, highlighting its central role in metabolic syndrome [90, 91]. From the perspective of molecular signaling pathways, hyperglycemia impairs the IRS-1/PI3K signaling pathway, reducing glucose metabolism and endothelial nitric oxide synthase (eNOS) activity, thereby contributing to endothelial dysfunction. Concurrently, hyperinsulinemia abnormally activates the MAPK pathway, promoting inflammation and vascular smooth muscle cell proliferation, which accelerates atherosclerosis. Lipid dysregulation further aggravates insulin resistance via NF-κB–mediated inflammatory cascades and inhibition of IRS-1 signaling, creating a vicious cycle in the progression of atherosclerosis. As a composite indicator reflecting the metabolic burden of both glucose and lipid dysregulation, the TyG index may more sensitively capture these interactive abnormalities and underlying metabolic stress. Although no significant differences in FBG and TG levels were observed among groups in this study, the observed differences in TyG index suggest that it may better represent the overall metabolic risk and possess independent prognostic value.

    Based on the above mechanistic analysis, this study found that the group with a rapidly decreasing TyG index exhibited the lowest incidence of major adverse cardiovascular events (MACE). This finding may be attributed to improved insulin sensitivity, reduced inflammatory levels, and restored coronary circulation function. Future research could further explore whether lifestyle interventions (such as low-carbohydrate diets and exercise training) or pharmacological therapies (such as sodium-glucose co-transporter 2 [SGLT2] inhibitors and glucagon-like peptide-1 [GLP-1] receptor agonists) can improve the TyG index and thereby reduce the long-term cardiovascular risk in patients with ST-segment elevation myocardial infarction (STEMI).

    The results of this study suggest that the TyG index trajectory holds significant clinical value in risk assessment for patients with ST-segment elevation myocardial infarction (STEMI). Trajectory-based monitoring of the TyG index can provide a more accurate prediction of major adverse cardiovascular events (MACE) and assist in formulating more proactive metabolic management and cardiovascular protection strategies. These findings indicate that a single measurement of the TyG index is insufficient for comprehensive risk assessment, and regular monitoring of its trajectory may improve the accuracy of prognosis prediction. Future studies could explore the incorporation of the TyG trajectory into existing STEMI risk assessment models and evaluate the effectiveness of targeted interventions. Overall, as a simple and effective metabolic biomarker, the TyG index demonstrates considerable value for the early identification and personalized management of cardiovascular disease.

    Continue Reading

  • Effects of Brown Rice, Meal Replacements, and Anti-Obesity Drugs on Mi

    Effects of Brown Rice, Meal Replacements, and Anti-Obesity Drugs on Mi

    Introduction

    The global burden of obesity has reached unprecedented levels, emerging as a leading public health concern across both high-income and developing countries. Defined as a body mass index (BMI) of 30 kg/m² or more, obesity has witnessed a dramatic rise in prevalence, increasing from 4.6% in 1980 to approximately 14.0% by 2019.1,2 Data from the World Health Organization (WHO) indicate that, as of 2016, over 1.9 billion adults were overweight, with more than 650 million classified as obese.3 This alarming trend spans all age groups and demographics, implicating a constellation of sociocultural, behavioral, and nutritional determinants. Central to this phenomenon is a global dietary transition marked by increased intake of energy-dense, highly processed foods high in fats and sugars.4,5 The expansion of global food supply chains and urbanization has led to greater availability of such diets, contributing to poor dietary quality and the replacement of traditional food practices.6–8 This nutrition transition, especially in low- and middle-income countries, further accelerates the rise of obesity.

    In parallel, rising levels of sedentary behavior—driven by urban infrastructure, occupational patterns, and digital media consumption—have exacerbated this nutritional shift. Prolonged screen time, reduced physical activity, and limited engagement in outdoor recreational activities are increasingly prevalent among youth and adults, thereby promoting positive energy balance and fat accumulation.4,9 The COVID-19 pandemic has intensified these patterns, with lockdowns and school closures contributing to increased obesity rates among children and adolescents.10,11 Additionally, cultural shifts, including the Westernization of diets and the decline of home-cooked meals, have weakened protective dietary traditions and normalized fast-food consumption, particularly among younger populations.5,12 Together, these converging factors underscore the need for integrative public health strategies that address both nutritional and behavioral determinants of obesity.

    Beyond the epidemiological concerns, the biological mechanisms underlying obesity have gained increasing research attention. One critical dimension of obesity pathogenesis is the expansion of white adipose tissue (WAT) and its intricate link with mitochondrial dysfunction. WAT is the primary energy storage tissue, and its pathological expansion under chronic positive energy balance disrupts cellular homeostasis and metabolic regulation. Excessive WAT accumulation impairs adipose tissue function through hypoxia, inflammation, and lipotoxicity, resulting in systemic insulin resistance and metabolic complications, such as type 2 diabetes and cardiovascular diseases.13,14 WAT expansion leads to the hypertrophy of adipocytes, which compromises mitochondrial bioenergetics, reduces oxidative capacity, and impairs fatty acid oxidation, culminating in the generation of reactive lipid species and the deterioration of metabolic health.15–17

    Furthermore, local hypoxia due to adipocyte enlargement contributes to inflammatory cascades. As adipocytes outgrow their vascular supply, hypoxic conditions upregulate pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), which in turn exacerbate mitochondrial dysfunction and fuel a state of chronic low-grade inflammation.18–21 These effects are further magnified by impaired vascular function and endothelial stress, which compromise nutrient and oxygen delivery to adipose tissue.22 The adipokine profile also undergoes significant changes in obesity, with reduced adiponectin and dysregulated leptin secretion contributing to impaired mitochondrial biogenesis and energy imbalance.14,23 Downregulation of energy-sensing pathways such as AMP-activated protein kinase (AMPK) is frequently observed in obesity, further compromising mitochondrial integrity and promoting metabolic inflexibility.15

    The transformation of WAT into beige adipose tissue (BeAT)—a process known as browning—represents a promising adaptive mechanism for enhancing mitochondrial function and increasing thermogenic capacity. However, obesity-related inflammation and cellular dysfunction can suppress this adaptive response, leading to compromised energy dissipation and exacerbated fat accumulation.24,25 These mechanistic insights emphasize the centrality of mitochondrial health in WAT to overall metabolic homeostasis and underscore the need for interventions targeting both adipose remodeling and mitochondrial integrity.

    A growing body of evidence implicates the gut microbiota in the pathogenesis of obesity, particularly via alterations in microbial diversity and the Firmicutes/Bacteroidetes (F/B) ratio. The gut microbiome is intricately linked with nutrient metabolism, energy extraction, immune modulation, and endocrine signaling. Numerous studies have shown that individuals with obesity tend to have a higher F/B ratio compared to lean individuals, suggesting that a Firmicutes-dominant microbiota enhances the capacity for energy extraction from the diet.26–29 Firmicutes ferment dietary fibers into short-chain fatty acids (SCFAs), which can serve as additional energy substrates, thereby increasing caloric absorption and promoting adiposity.30,31 In contrast, Bacteroidetes are associated with more efficient carbohydrate metabolism and a leaner phenotype.32,33 Thus, a skewed F/B ratio contributes to the positive energy balance characteristic of obesity.34

    In addition to energy metabolism, the gut microbiota modulates systemic inflammation and immune responses. The overrepresentation of Firmicutes has been associated with increased inflammatory cytokine expression and compromised intestinal barrier integrity, which further exacerbate insulin resistance and metabolic syndrome.35–37 The microbiota’s influence extends to hormonal regulation of appetite and satiety, affecting leptin and ghrelin pathways.38,39 Notably, interventions that modulate the gut microbiota—including prebiotic and probiotic supplementation—have demonstrated potential in improving the F/B ratio and metabolic outcomes.40–43 These findings highlight the gut microbiota as both a contributor to and a potential target for obesity treatment.

    In response to these multifactorial contributors to obesity, several dietary and pharmacological interventions have been explored for their potential to restore metabolic balance. Among them, brown rice, commercial meal replacements, and thiazolidinediones (TZDs) have garnered significant attention. Brown rice retains the bran and germ layers, offering a higher content of fiber, antioxidants, and essential nutrients compared to refined white rice. Its fiber content improves gastrointestinal motility, supports the proliferation of beneficial gut bacteria, and enhances satiety, contributing to reduced caloric intake and improved glycemic control.44–46 Brown rice consumption has also been linked to a decreased risk of type 2 diabetes, supporting its inclusion in metabolic disease prevention strategies.

    Meal replacements represent another strategic intervention, offering controlled caloric intake with optimized macronutrient composition. Clinical trials have demonstrated that meal replacements lead to greater weight loss and improved adherence compared to conventional diets.47,48 High-protein, low-glycemic formulations have been shown to enhance insulin sensitivity and reduce fat mass while preserving lean body mass.49,50 Furthermore, their standardized composition facilitates long-term dietary compliance and supports structured nutritional interventions.51,52

    Pharmacologically, TZDs such as pioglitazone exert beneficial effects on insulin sensitivity and adipose tissue function by activating peroxisome proliferator-activated receptor gamma (PPARγ). This activation promotes adipocyte differentiation, enhances glucose uptake, and redistributes fat from visceral to subcutaneous depots, reducing obesity-related metabolic risks.53–55 Notably, TZDs have also been implicated in modulating gut microbiota composition, potentially contributing to their metabolic effects.47,56

    In light of these findings, the present study aims to comparatively evaluate the effects of brown rice, meal replacements, and TZDs on mitochondrial function in white adipose tissue and gut microbiota composition, particularly the Firmicutes/Bacteroidetes ratio, within a high-fat, high-fructose (HFHF) diet-induced obesity model in rats. By integrating assessments of dietary impact, mitochondrial dynamics, and microbial profiles, this study seeks to illuminate potential mechanisms through which these interventions mitigate obesity-related metabolic disturbances. This integrative approach may provide a foundation for targeted therapeutic strategies addressing both systemic metabolism and gut-adipose axis interactions in the context of obesity.

    Materials and Methods

    Study Design

    This study employed a post-test only controlled group design within a laboratory in vivo experimental framework. The protocol was conducted at the Laboratory for Animal Model Development, Faculty of Medicine, Universitas Brawijaya. Ethical clearance was obtained from the Ethics Committee of the Faculty of Health Sciences, Universitas Brawijaya (Reference No: 2020/UN10.F17.10.4/TU/2023), and all procedures were carried out in accordance with internationally accepted standards for animal care and use approved by the Animal Lab Medicine Faculty – Universitas Brawijaya follows the 3Rs, the Five Freedoms, and Indonesian Government Regulation No. 95/2012 on veterinary public health and animal welfare.

    The selected design was appropriate for investigating the comparative effects of dietary and pharmacological interventions in an obesity model, as it enables outcome analysis after the intervention period without prior baseline measurements, reducing potential handling-induced stress on the animals. The use of Sprague Dawley rats is well-established in metabolic studies due to their susceptibility to diet-induced obesity and their physiological similarity to human metabolic responses.13,15

    Animal and Grouping

    Twenty male Sprague Dawley rats aged 8–14 weeks, with initial body weights ranging between 150 and 250 g, were selected. All animals were confirmed to be in healthy condition prior to allocation. Randomization was performed using simple random sampling to assign animals into five groups (n = 4 per group), based on dietary regimen as follows:

    • Group 1, control: Standard AIN-93M diet;
    • Group 2, HFHF: high-fat, high-fructose;
    • Group 3, HFHF with BR: HFHF diet supplemented with brown rice;
    • Group 4, HFHF with TZD: HFHF diet with thiazolidinedione (TZD) treatment;
    • Group 5, HFHF with MR: HFHF diet supplemented with commercial meal replacement.

    Rats exhibiting signs of illness—such as dull fur, hair loss, abnormal behavior, or unusual discharge from bodily orifices—were excluded from the study. Animals that died before completion of the intervention period were also excluded from analysis.

    Interventions

    Obesity was induced in Groups 2 to 5 over a 14-week period by administering a high-fat, high-fructose (HFHF) diet formulated to mimic human dietary patterns associated with obesity and metabolic syndrome in humans.26,40 The specific nutrient compositions of the standard, HFHF, and intervention diets administered to each group are detailed in Table 1. The control group received a standard AIN-93M diet.

    Table 1 The Compositions of the Diet per 100 g

    At the end of the 14th week, animals from Groups 1 (Control) and 2 (HFHF) were euthanized to serve as pre-intervention benchmarks. White adipose tissue (WAT) was collected for mitochondrial analysis, and fecal samples were obtained for gut microbiota profiling.

    During the subsequent 12 weeks (weeks 15–26), animals in Groups 3 to 5 received dietary or pharmacological interventions:

    • Brown Rice (BR): Substitution of HFHF carbohydrate sources with whole grain brown rice.
    • TZD Treatment: Daily administration of TZD at an appropriate dosage determined from prior literature.54
    • Meal Replacement (MR): Inclusion of a high-fiber, balanced macronutrient meal replacement product.

    Throughout the study, daily food and fluid intakes were recorded by measuring feed and fructose solution leftovers. Body weight was measured weekly. The Lee Index (g/cm³), a recognized obesity indicator in rodent models, was calculated using the formula: Lee Index = [Body weight^(1/3) / Nasal-anal length (cm)] × 1000. This index was employed to confirm obesity status (>300 g/cm³).17

    Mitochondrial Analysis

    Mitochondrial content and distribution in WAT were assessed using Bio Tracker™ 488 Green Mitochondria Dye, which selectively stains active mitochondria based on membrane potential. Tissue samples were fixed and stained following manufacturer protocols, then visualized using fluorescence microscopy.

    Images were captured at magnifications optimized to distinguish mitochondrial morphology and density. Quantification was performed via image analysis software to evaluate fluorescence intensity, a proxy for mitochondrial membrane potential and activity.18 This analysis was conducted at the Central Biomedical Laboratory, Faculty of Medicine, Universitas Brawijaya.

    Gut Microbiota Analysis

    At week 26, fecal samples (1–2 g) were collected from each rat and stored at −40°C until analysis. Microbial DNA was extracted, and quantitative Real-Time Polymerase Chain Reaction (RT-PCR) was conducted targeting 16S rRNA genes specific to Firmicutes and Bacteroidetes. Total DNA was extracted from fecal samples using QIAamp Fast DNA Stool Mini Kit (QIAGEN, Germany) according to the manufacturer’s protocol. Quantification of the Firmicutes and Bacteroidetes phyla was performed using quantitative PCR (qPCR) targeting specific 16S rRNA gene regions.

    The following primer sequences were used:

    Forward: 5’-TGAAACTYAAAGGAATTGACG-3’

    Reverse: 5’-ACCATGCACCACCTGTC-3’

    Forward: 5’-GGARCATGTGGTTTAATTCGATGAT-3’

    Reverse: 5’-AGCTGACGACAACCATGCAG-3’

    The qPCR reactions were carried out in a total volume of 20 µL, consisting of 10 µL SYBR Green Master Mix (Applied Biosystems), 0.5 µL of each primer (10 µM), 2 µL of DNA template, and nuclease-free water. Amplification was conducted using a StepOnePlus™ Real-Time PCR System (Applied Biosystems) with the following thermal cycling conditions:Initial denaturation at 95°C for 10 minutes, followed by 40 cycles of denaturation at 95°C for 15 seconds, annealing at 60°C for 30 seconds, and extension at 72°C for 30 seconds.

    Standard curves were generated using serial dilutions of cloned 16S rRNA gene fragments to ensure accurate quantification. All reactions were performed in triplicate. Negative controls (no template) were included to monitor contamination, and melting curve analysis was performed to confirm the specificity of amplification.

    The Firmicutes/Bacteroidetes (F/B) ratio was calculated using the relative quantification method: 2^−ΔCt, where ΔCt represents the difference in threshold cycles between the two bacterial groups. This ratio served as a proxy for gut microbiota composition, given its established association with obesity phenotypes.27,37

    Statistical Analysis

    Data were expressed as mean ± standard deviation (SD). Continuous variables such as body weight, energy intake, fiber intake, and mitochondrial counts were tested for normality and homogeneity using Shapiro–Wilk and Levene’s tests, respectively. Where necessary, variables were log-transformed to meet assumptions for parametric testing.

    One-way Analysis of Variance (ANOVA) followed by Tukey’s post-hoc test was used to assess group differences. Linear regression was employed to examine associations between fiber intake, F/B ratio, and mitochondrial count. Statistical significance was set at p < 0.05. All analyses were performed using IBM SPSS Statistics for Windows, Version 26.

    This methodology enables robust exploration of the interconnections between diet, gut microbiota, and mitochondrial dynamics in a controlled obesity model. By integrating morphological, microbial, and biochemical assessments, the study provides a comprehensive framework for evaluating the mechanistic effects of nutritional and pharmacological interventions on metabolic health.

    Results

    Body Weight Trends Across Experimental Groups

    Longitudinal assessment of body weight demonstrated significant variations among the five experimental groups (Figure 1A). At baseline (week 1), no statistically significant differences in body weight were observed among the groups (p > 0.05). However, by week 14, after administration of the high-fat high-fructose (HFHF) diet in Groups 2–5, there was a pronounced increase in body weight, most notably in the HFHF group, indicating successful obesity induction.

    Figure 1 Impact of different treatments on anthropometry over time in obese rat models. (A) Body weight (g); (B) Lee index (g/cm³). Data are expressed as mean ± SD for body weight and Lee index at weeks 1, 14, and 26 (n = 4 per group). Error bars represent standard deviation (SD). Data points labeled with different symbols (*, #, &) indicate significant differences between groups (p < 0.05) according to one-way ANOVA followed by Tukey’s post hoc test.

    The HFHF group exhibited the steepest weight gain throughout the study, culminating in the highest body weight at week 26 (p < 0.05). Conversely, the control group displayed a gradual and modest increase in body weight over the same period, reflecting a typical growth pattern in the absence of dietary perturbation. The brown rice (BR) group experienced a notable deceleration in weight gain between weeks 14 and 26, suggesting a mitigating effect of BR on HFHF-induced weight gain. The TZD group followed a similar trajectory, with weight gain plateauing after week 14, indicating that TZD might attenuate further weight accrual post-obesity induction. Meanwhile, the meal replacement (MR) group showed intermediate results: although body weight remained elevated at week 26 compared to the control group, it was significantly lower than the HFHF group, suggesting a partial protective effect.

    LEE Index Variation as an Obesity Indicator

    The LEE index, a morphological obesity marker equivalent to the BMI in rodents, also varied significantly across groups (p < 0.05) (Figure 1B). At week 14, both the HFHF and BR groups exceeded the obesity threshold (>300 g/cm³), confirming successful model induction. However, by week 26, the BR group exhibited a substantial reduction in LEE index, falling below the obesity threshold, which underscores its potential in reversing diet-induced obesity. Similarly, the TZD group experienced a notable drop in LEE index post-intervention, indicating its effectiveness in modulating adiposity.

    In contrast, the HFHF group continued to show an upward trajectory in LEE index, reaching its peak at week 26, thereby confirming that the HFHF diet perpetuates obesity. Notably, its LEE index was significantly higher than that of the control group, further supporting the obesogenic effect of the HFHF diet. The MR group maintained a relatively stable LEE index around the obesity threshold, suggesting moderate efficacy in obesity prevention. The control group consistently maintained LEE index values below the threshold across all time points, as expected.

    Post hoc comparisons indicated that the BR group had a significantly higher LEE index than both the TZD and MR groups, as denoted by distinct annotation letters in the figure.

    Dietary Intake Characteristics

    Dietary intake parameters are summarized in Table 2. Energy intake was highest in the HFHF group (126.01 ± 23.71 kcal/day), followed by BR (87.85 ± 10.37 kcal/day), whereas the MR and control groups had the lowest energy intake (p < 0.001). These findings reflect the high caloric density of the HFHF diet and the moderate intake regulation associated with fiber-enriched interventions such as BR and MR. The TZD group also demonstrated relatively low energy intake, possibly reflecting pharmacologically induced appetite suppression.

    Table 2 Dietary Intake Characteristics Across Treatment Groups During Intervention (Mean ± SD, n = 4)

    Protein intake was significantly elevated in the BR group due to its dietary formulation, whereas the MR group exhibited a slightly lower protein intake. The HFHF group had moderate protein intake, while the TZD and control groups displayed the lowest values.

    Regarding fat intake, the HFHF group again showed the highest values, consistent with the high lipid content of the diet. The BR group also had substantial fat intake, while the TZD and MR groups showed significantly reduced fat consumption. Carbohydrate intake mirrored energy intake trends, with HFHF rats consuming the highest levels, while MR rats showed minimal carbohydrate intake. Fiber intake differed significantly among groups (p < 0.001), with BR (6.36 ± 1.01 g/day) and MR (5.74 ± 0.22 g/day) exhibiting the highest values (Figure 2). The TZD group had the lowest fiber intake, potentially limiting its microbiota-modulating capacity.

    Figure 2 Fiber across different groups. Data are expressed as mean fiber intake ± SD (n = 4 per group). Bars labeled with different symbols (*, #) indicate significant differences between groups (p < 0.05) according to one-way ANOVA followed by Tukey’s post hoc test.

    Firmicutes/Bacteroidetes (F/B) Ratio

    Gut microbiota composition, assessed through the Firmicutes/Bacteroidetes (F/B) ratio, revealed significant group differences (Figure 3). The HFHF group exhibited the highest F/B ratio (~1.9), indicative of microbial dysbiosis commonly associated with obesity.26,27 The TZD group also showed a moderately elevated ratio (~1.5), whereas the BR and MR groups had ratios approximating 1.3 and 1.0, respectively, suggesting a more balanced microbiota and healthier metabolic state.

    Figure 3 Firmicutes/Bacteroidetes (F/B) ratio across treatment groups. Higher ratios indicate increased Firmicutes dominance, typically associated with greater energy extraction efficiency. F/B ratio is a unitless value derived from the relative abundance of each phylum.

    The control group maintained a near-equal balance (F/B ≈ 1.0), consistent with a normobiotic gut profile. These findings support previous reports that a higher F/B ratio is associated with increased energy harvest and fat accumulation, while a lower ratio correlates with improved metabolic outcomes.32,35

    Mitochondrial Abundance in Adipose Tissue

    Quantitative analysis of mitochondrial abundance in white adipose tissue (WAT) showed significant differences among groups (Figure 4). The MR group displayed the highest mitochondrial count (~60 units), followed by the TZD group (~50 units). These results suggest a marked enhancement of mitochondrial biogenesis or activity in these intervention groups.

    Figure 4 The Number of mitochondria representative Mitotracker staining of WAT in the (A) control group; (B) HFHF group; (C) brown rice group; (D) TZD group; (E) MR group; (F) The number of mitochondria across different groups. Data are expressed as mean number of mitochondria ± SD (n = 4 per group).

    The BR and control groups had intermediate values (~30–40 units), while the HFHF group had the lowest mitochondrial count (~30 units), consistent with diet-induced mitochondrial impairment. These findings reinforce existing literature on the deleterious effects of high-fat diets on mitochondrial function and the potential for dietary and pharmacological strategies to restore mitochondrial health.13,18 Despite these observable differences, statistical analysis showed that the variations were not statistically significant (p > 0.05), indicating that the differences in mean mitochondrial counts did not reach the threshold for significance.

    Correlation Between Fiber Intake and F/B Ratio

    Correlation analysis (Figure 5) revealed a positive association between dietary fiber intake and a more balanced F/B ratio across groups. The BR and MR groups, which had the highest fiber intake, also exhibited the most favorable microbiota compositions. In contrast, the HFHF and TZD groups, with lower fiber intakes, maintained elevated F/B ratios, indicating an association between dietary fiber and microbial homeostasis.33,37

    Figure 5 Linear regression analysis of the relationship between fiber intake and Firmicutes/Bacteroidetes ratio across all groups. values demonstrate the strength of association.

    Combined Effects of Diet on Mitochondria and Microbiota

    A 3D bubble plot (Figure 6) integrated fiber intake, F/B ratio, and mitochondrial count. The BR and MR groups clustered in the quadrant representing high fiber intake, balanced microbiota, and elevated mitochondrial abundance. The TZD group showed relatively high mitochondrial activity despite lower fiber intake, suggesting that TZD may exert direct pharmacological effects on mitochondrial biogenesis and metabolic signaling pathways.47,53 In contrast, the HFHF group was isolated in the quadrant indicating low fiber intake, high F/B ratio, and diminished mitochondrial content, reinforcing the detrimental effect of this dietary pattern on metabolic health.

    Figure 6 3D bubble plot illustrating the relationship among fiber intake (x-axis), F/B ratio (y-axis), and mitochondrial count (bubble size) across all groups. Bubble size reflects mean mitochondrial abundance.

    Together, these findings demonstrate that interventions with BR and MR exert beneficial metabolic effects by modulating gut microbiota composition and enhancing mitochondrial activity. TZD, while effective in improving mitochondrial outcomes, was less impactful on microbiota composition, likely due to its lower fiber content. This multidimensional analysis highlights the interdependent roles of diet, gut microbiota, and mitochondrial function in the context of obesity and provides insight into the mechanistic efficacy of these interventions.

    Discussion

    The present study evaluated the comparative efficacy of brown rice (BR), thiazolidinediones (TZDs), and commercial meal replacements (MRs) in modulating obesity-related outcomes in an HFHF-induced rat model. The discussion herein integrates experimental findings with relevant literature to interpret observed physiological, biochemical, and microbiological changes, while highlighting the implications and limitations of the study.

    The HFHF diet successfully induced obesity, evidenced by significant weight gain, elevated Lee Index values, and adverse metabolic alterations, corroborating earlier studies where excessive fructose intake disrupted hypothalamic satiety signaling by reducing leptin sensitivity and decreasing expression of anorexigenic peptides60. As expected, this diet promoted hyperphagia, triglyceride accumulation, and adipocyte hypertrophy, supporting previous findings linking high-fructose diets to WAT expansion, insulin resistance, and hepatic steatosis.13,17

    The Lee Index served as a reliable indicator of obesity severity, with values >300 g/cm^3 observed in HFHF-fed rats by week 14, aligning with thresholds established for rodent obesity classification15. In contrast, interventions involving BR and TZD effectively reduced the Lee Index below this critical threshold, demonstrating their potential to reverse obesity phenotypes despite initial weight gains. MRs also showed a stabilizing effect, albeit less pronounced.

    Analysis of nutrient intake revealed the highest caloric and fat consumption in the HFHF group, while BR and MR groups, rich in fiber, demonstrated moderated energy intake and improved metabolic indices. The high fiber content in BR (43.6 g/100 g) and MR (49.7 g/100 g) likely contributed to satiety, reduced energy intake, and modulated lipid metabolism, consistent with prior evidence on dietary fiber’s role in weight management.57

    Dietary fiber’s fermentation into short-chain fatty acids (SCFAs) plays a crucial role in host metabolism. SCFAs such as acetate and butyrate not only serve as energy sources but also regulate gut barrier integrity and immune function.35 In the current study, the BR and MR groups exhibited improved gut microbial profiles and mitochondrial abundance, suggesting SCFA-mediated metabolic benefits. Notably, these groups had lower Firmicutes/Bacteroidetes (F/B) ratios and higher mitochondrial counts, indicating gut eubiosis and enhanced energy metabolism.

    The F/B ratio, often used as a marker of gut dysbiosis, was significantly elevated in the HFHF group (~1.9), consistent with findings that associate higher F/B ratios with obesity and increased energy harvest.26,27 BR and MR interventions normalized this ratio (~1.3 and 1.0, respectively), demonstrating dietary fiber’s capacity to restore microbial balance and reduce obesity risk. These results align with prior studies where high-fiber diets were linked to increased Bacteroidetes abundance and reduced adiposity.58

    Mitochondrial quantification further elucidated intervention effects. The HFHF group exhibited the lowest mitochondrial count in WAT, reflecting impaired mitochondrial biogenesis likely due to lipotoxicity, inflammation, and hypoxia.14,19 Conversely, the MR group showed the highest mitochondrial abundance (~60 units), followed by TZD and BR groups, implicating these interventions in mitochondrial restoration. This observation echoes the findings of Pollicino (2023), who highlighted the Mediterranean diet’s capacity to reduce mitochondrial ROS and improve respiration.59 Furthermore, although Mitotracker-based staining reflects mitochondrial abundance and membrane potential, it does not directly assess mitochondrial bioenergetics. Future studies should incorporate functional assays such as ATP production, mtDNA copy number, and oxidative phosphorylation to enhance mechanistic insight.

    TZDs, specifically PPAR-γ agonists like rosiglitazone, have been shown to induce browning of WAT and enhance mitochondrial function by upregulating UCP1 and PRDM16 expression.53 However, their limited effect on the F/B ratio suggests that while TZDs improve cellular metabolism, they lack direct influence on microbial ecology. Additionally, the dose-dependent risks associated with TZDs, including fluid retention, heart failure, and mitochondrial toxicity, underscore the importance of cautious administration.60,61

    In contrast, BR and MR exhibited a dual mechanism—modulating both gut microbiota and mitochondrial biogenesis—without the adverse effects observed in pharmacological approaches. Gamma-oryzanol, a bioactive component of BR, has been implicated in metabolic regulation through hypothalamic and hepatic pathways, although not primarily via microbiota modulation.62,63

    The 3D bubble plot visualization integrated dietary fiber, microbiota composition, and mitochondrial count, demonstrating that high-fiber groups (BR and MR) clustered within the quadrant representing favorable metabolic outcomes. This multi-parametric representation reinforces the synergistic role of diet in regulating host-microbiota-mitochondria interactions, a concept supported by Colangeli (2023), who demonstrated that gut microbiota regulates mitochondrial function and energy expenditure.64

    Moreover, findings in this study align with previous reports suggesting that Firmicutes abundance is enhanced by high-fat intake, leading to increased LPS production and inflammatory signaling.65 The association between microbiota-driven inflammation and metabolic dysfunction has been extensively reviewed, with cytokine-mediated mitochondrial inhibition and apoptosis contributing to obesity pathology.20,21

    Despite its relevance, this study has several limitations that warrant consideration. The small sample size (n = 4/group), although aligned with exploratory preclinical models, may limit statistical power and increase the risk of Type I and II errors. Future research should incorporate power calculations and larger cohorts. Mechanistic interpretations regarding SCFA-mediated benefits, mitochondrial biogenesis, and hypothalamic regulation have been revised as hypotheses, given the absence of direct markers such as SCFA levels, UCP1 expression, or AMPK/PGC-1α signaling. Additionally, reliance on the Firmicutes/Bacteroidetes (F/B) ratio as the sole microbiota marker is acknowledged as a limitation. Broader microbiome analyses, including diversity indices and taxonomic resolution, are recommended in future studies. We also recognize that the intervention diets were not isocaloric, which may confound interpretations of mitochondrial and microbial changes. However, ad libitum feeding was intentionally applied to simulate real-world overnutrition. Lastly, while the findings are promising, recommendations for clinical trials have been reframed to emphasize the need for further mechanistic and preclinical validation prior to translation into human studies.

    In conclusion, this research underscores the potential of dietary strategies—particularly those high in fiber—to ameliorate obesity by restoring gut microbiota balance and mitochondrial function. These findings support the integration of nutritional interventions into obesity management protocols and emphasize the importance of holistic approaches targeting both host metabolism and microbial ecology. Further studies exploring the mechanistic underpinnings and clinical translation of these findings are warranted to enhance therapeutic precision in metabolic disease management.

    Conclusion

    This study provides compelling evidence that dietary interventions using brown rice (BR) and meal replacements (MR), as well as pharmacological treatment with thiazolidinediones (TZDs), exert distinct yet overlapping effects in ameliorating diet-induced obesity in a rodent model. The high-fat, high-fructose (HFHF) diet effectively induced obesity, as indicated by increased body weight, elevated Lee Index, disrupted gut microbiota composition, and reduced mitochondrial abundance in white adipose tissue (WAT). Each intervention produced differential impacts on these metabolic parameters.

    Among the key findings, BR and MR significantly improved gut microbiota balance, as reflected by more favorable Firmicutes/Bacteroidetes (F/B) ratios, and promoted higher mitochondrial counts in WAT, suggesting enhanced cellular energy metabolism. These effects are likely attributable to the high dietary fiber content and associated short-chain fatty acid production, which modulate both microbial ecology and host metabolic signaling pathways. BR also demonstrated unique benefits, potentially mediated by bioactive compounds such as γ-oryzanol.

    TZD treatment, while effective in improving mitochondrial abundance and reversing obesity markers, exhibited a more limited impact on gut microbiota composition, indicating that its metabolic benefits may be primarily mediated through PPARγ activation and mitochondrial biogenesis rather than modulation of gut flora. Nonetheless, its role in promoting the browning of WAT and enhancing oxidative capacity is notable.

    These findings underscore the interrelated roles of diet, gut microbiota, and mitochondrial function in the development and management of obesity. The integration of microbiota and mitochondrial metrics into obesity research enhances our understanding of the mechanistic pathways underlying metabolic health and supports the development of targeted, non-pharmacological interventions.

    Importantly, this study contributes to the growing literature advocating for dietary strategies—particularly those rich in whole grains and fiber—as sustainable and safe approaches to managing obesity and its complications. It also highlights the potential complementary role of pharmacotherapy when dietary modifications alone are insufficient.

    Future research should expand on these findings by employing larger sample sizes, comprehensive microbial and metabolomic profiling, and mechanistic studies exploring host-microbe-mitochondria interactions. Clinical trials translating these preclinical insights into human populations will also be essential for validating the practical applicability of these interventions.

    In sum, this study advances the understanding of how specific dietary and pharmacological interventions influence obesity-related metabolic pathways and reinforces the critical role of diet quality in promoting long-term metabolic health.

    Abbreviations

    ATP, adenosine triphosphate; BAT, brown adipose tissue; BeAT, beige adipose tissue; BR, brown rice; F/B ratio, the Firmicutes-Bacteroidetes ratio; HFHF, high-fat, high-fructose; LPL, lipoprotein lipase; LPS, lipopolysaccharides; MedDiet, Mediterranean diet; MR, meal replacement; mtROS, mitochondrial reactive oxygen species; Myf5−, myogenic factor 5 negative cells; Myf5+, myogenic factor 5 positive cells; PGE2, prostaglandin E2; PPARy, peroxisome proliferator-activated receptor gamma; PRDM16, protein PR-domain containing 16; RT-PCR, real-time polymerase chain reaction; SCFA, short-chain fatty acids; SD, standard deviation; T2DM, type 2 diabetes mellitus; TG, triglycerides; TZD, thiazolidinediones; UCP1, uncoupling protein 1; VLDL, very low-density lipoprotein; WAT, white adipose tissue.

    Data Sharing Statement

    The raw data including mitochondria raw data, analytical codes, and other collected data that support the findings of this study are available from the corresponding author upon request.

    Ethics Approval and Consent to Participate

    Ethical approval was granted by the Ethics Committee, Faculty of Health Sciences, Universitas Brawijaya (2020/UN10.F17.10.4/TU/2023).

    Acknowledgments

    The authors would like to thank the Faculty of Health Sciences, Universitas Brawijaya, for their support in this study.

    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.

    Funding

    This research was funded by BPPM funding under contract number 2/UN10.F17.01/PT.01.03.2/2023.

    Disclosure

    The authors declare that the study was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

    References

    1. Boutari C, Mantzoros C. 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

    2. Jeong S, Jung J, Yang Y, et al. 2023 obesity fact sheet: prevalence of obesity and abdominal obesity in adults, adolescents, and children in Korea from 2012 to 2021. J Obes Metab Syndr. 2024;33(1):27–35. doi:10.7570/jomes24012

    3. Kim K, Shin Y. Males with obesity and overweight. J Obes Metab Syndr. 2020;29(1):18–25. doi:10.7570/jomes20008

    4. Chooi Y, Ding C, Magkos F. The epidemiology of obesity. Metabolism. 2019;92:6–10. doi:10.1016/j.metabol.2018.09.005

    5. Fan H, Zhang X. Prevalence of and trends in the co-existence of obesogenic behaviors in adolescents from 15 countries. Front Pediatr. 2021;9:664828. doi:10.3389/fped.2021.664828

    6. Fox A, Feng W, Asal V. What is driving global obesity trends? Globalization or “modernization”? Glob Health. 2019;15(1):32. doi:10.1186/s12992-019-0457-y

    7. Chong B, Jayabaskaran J, Kong G, et al. Trends and predictions of malnutrition and obesity in 204 countries and territories: an analysis of the global burden of disease study 2019. EClinicalMedicine. 2023;57:101850. doi:10.1016/j.eclinm.2023.101850

    8. Fontes F, Damasceno A, Jessen N, et al. Prevalence of overweight and obesity in Mozambique in 2005 and 2015. Public Health Nutr. 2019;22(17):3118–3126. doi:10.1017/s1368980019002325

    9. Sun X, Du T. Trends in weight change patterns across life course among US adults, 1988–2018: population-based study. BMC Public Health. 2023;23(1):17137. doi:10.1186/s12889-023-17137-x

    10. Park H, Seo J, Jung H, Lim J. Prevalence and trends in obesity and severe obesity in Korean children and adolescents, 2007–2020: a population-based study. Pediatr Int. 2023;65(1):e15472. doi:10.1111/ped.15472

    11. Kang S, Seo M, Kim S, Park M. Changes in lifestyle and obesity during the COVID-19 pandemic in Korean adolescents: based on the Korea youth risk behavior survey 2019 and 2020. Ann Pediatr Endocrinol Metab. 2022;27(4):281–288. doi:10.6065/apem.2142228.114

    12. Gilla S, Abida H. Prevalence of overweight and obesity among women older than 20 years. Int J Community Med Public Health. 2019;6(5):2229. doi:10.18203/2394-6040.ijcmph20191849

    13. Heinonen S, Jokinen R, Rissanen A, Pietiläinen K. White adipose tissue mitochondrial metabolism in health and in obesity. Obes Rev. 2019;21(2):e12958. doi:10.1111/obr.12958

    14. Chait A, Hartigh L. Adipose tissue distribution, inflammation and its metabolic consequences, including diabetes and cardiovascular disease. Front Cardiovasc Med. 2020;7:22. doi:10.3389/fcvm.2020.00022

    15. White U. Adipose tissue expansion in obesity, health, and disease. Front Cell Dev Biol. 2023;11:1188844. doi:10.3389/fcell.2023.1188844

    16. Wang J, Yang L, Yang L, et al. Adipose dysfunction in adulthood insulin resistance of low-birth weight mice: a proteomics study. Diabetes Metab Syndr Obes. 2022;15:849–862. doi:10.2147/dmso.s353095

    17. Longo M, Zatterale F, Naderi J, et al. Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int J Mol Sci. 2019;20(9):2358. doi:10.3390/ijms20092358

    18. Morigny P, Boucher J, Arner P, Langin D. Lipid and glucose metabolism in white adipocytes: pathways, dysfunction and therapeutics. Nat Rev Endocrinol. 2021;17(5):276–295. doi:10.1038/s41574-021-00471-8

    19. Karlina R, Lutter D, Miok V, et al. Identification and characterization of distinct brown adipocyte subtypes in C57BL/6J mice. Life Sci Alliance. 2020;4(1):e202000924. doi:10.26508/lsa.202000924

    20. Haroun N, Bennour I, Seipelt E, et al. Maternal vitamin D deficiency in mice increases white adipose tissue inflammation in offspring. Cells. 2022;11(13):2024. doi:10.3390/cells11132024

    21. Corrêa L, Heyn G, Magalhães K. The impact of the adipose organ plasticity on inflammation and cancer progression. Cells. 2019;8(7):662. doi:10.3390/cells8070662

    22. Phuong T, Walker A, Henson G, et al. Deletion of ROBO4 prevents high‐fat diet‐induced adipose artery and systemic metabolic dysfunction. Microcirculation. 2019;26(5):e12540. doi:10.1111/micc.12540

    23. Song A, Dai W, Jang M, et al. Low- and high-thermogenic brown adipocyte subpopulations coexist in murine adipose tissue. J Clin Invest. 2019;130(1):247–257. doi:10.1172/jci129167

    24. Ma P, Xiao J, Hou B, et al. Carbonic anhydrase 2 mediates anti-obesity effects of black tea as thermogenic activator. Food Sci Hum Wellness. 2024;13(5):2917–2936. doi:10.26599/fshw.2022.9250236

    25. Im H, Park J, Im S, Han J, Kim K, Lee Y. Regulatory roles of g-protein coupled receptors in adipose tissue metabolism and their therapeutic potential. Arch Pharm Res. 2021;44(2):133–145. doi:10.1007/s12272-021-01314-w

    26. Kim J, Choi Y, Kwon H, et al. Effect of gut microbiome on minor complications after a colonoscopy. Intest Res. 2021;19(3):341–348. doi:10.5217/ir.2020.00057

    27. Duan M, Wang Y, Zhang Q, Zou R, Guo M, Zheng H. Characteristics of gut microbiota in people with obesity. PLoS One. 2021;16(8):e0255446. doi:10.1371/journal.pone.0255446

    28. Hassan N, El-Masry S, Shebini S, et al. Gut dysbiosis is linked to metabolic syndrome in obese Egyptian women: potential treatment by probiotics and high fiber diets regimen. Sci Rep. 2024;14(1):xx. doi:10.1038/s41598-024-54285-5

    29. Liu R, Shu Y, Qi W, et al. Protective effects of almond oil on streptozotocin-induced diabetic rats via regulating Nrf2/HO-1 pathway and gut microbiota. J Food Qual. 2021;2021:1–14. doi:10.1155/2021/5599219

    30. Bao M, Hou K, Xin C, et al. Portulaca oleracea L. extract alleviated type 2 diabetes via modulating the gut microbiota and serum branched‐chain amino acid metabolism. Mol Nutr Food Res. 2022;66(11):e202101030. doi:10.1002/mnfr.202101030

    31. Kim J, Kim Y, Oh G, Jung W, Park S. How is gut microbiome of patients with familial adenomatous polyposis different from healthy people? Medicine. 2022;101(49):e32194. doi:10.1097/md.0000000000032194

    32. Zeng T, Cao Y, Yin J, et al. Unraveling the gut microbiota of Tibetan chickens: insights into highland adaptation and ecological advantages. Microbiol Spectr. 2024;12(11). doi:10.1128/spectrum.00519-24

    33. Sutoyo D, Atmaka D, Sidabutar L. Dietary factors affecting firmicutes and bacteroidetes ratio in solving obesity problem: a literature review. Media Gizi Indones. 2020;15(2):94–109. doi:10.20473/mgi.v15i2.94-109

    34. Cui W, Chen F, Sun Z, et al. Catabolism of phenolics from grape peel and its effects on gut microbiota during in vitro colonic fermentation. J Sci Food Agric. 2024;104(12):7182–7193. doi:10.1002/jsfa.13540

    35. Pan H, Li Z, Xie J, et al. Berberine influences blood glucose via modulating the gut microbiome in grass carp. Front Microbiol. 2019;10:1066. doi:10.3389/fmicb.2019.01066

    36. Wu N, Zhou J, Mo H, et al. The gut microbial signature of gestational diabetes mellitus and the association with diet intervention. Front Cell Infect Microbiol. 2022;11:800865. doi:10.3389/fcimb.2021.800865

    37. Cheng Z, Zhang L, Yang L, Chu H. The critical role of gut microbiota in obesity. Front Endocrinol. 2022;13:1025706. doi:10.3389/fendo.2022.1025706

    38. Wei X, Zong W, Gao Y, Peng S, Liu K, Zheng Y. Effects of the traditional Chinese medicine Tang Luo Ning on intestinal flora and oxidative stress in diabetic rats. Evid Based Complement Alternat Med. 2020;2020(1):3452625. doi:10.1155/2020/3452625

    39. Nehmi‐Filho V, Freitas J, Franco L, et al. Modulation of the gut microbiome and firmicutes phylum reduction by a nutraceutical blend in the obesity mouse model and overweight humans: a double‐blind clinical trial. Food Sci Nutr. 2024;12(4):2436–2454. doi:10.1002/fsn3.3927

    40. Mamun M, Rakib A, Mandal M, Singh UP. Impact of a high-fat diet on the gut microbiome: a comprehensive study of microbial and metabolite shifts during obesity. Cells. 2025;14(6):463. doi:10.3390/cells14060463

    41. Ali M. Gender dependent gut microbiome in obese Egyptian individuals. Rec Pharm Biomed Sci. 2019;39–42. doi:10.21608/rpbs.2019.10975.1024

    42. Caudet J, Trelis M, Cifre S, Soriano J, Rico H, Merino-Torres J. Interplay between intestinal bacterial communities and unicellular parasites in a morbidly obese population: a neglected trinomial. Nutrients. 2022;14(15):3211. doi:10.3390/nu14153211

    43. Hankel J, El‐Wahab A, Grone R, et al. Faecal microbiota of dogs offered a vegetarian diet with or without the supplementation of feather meal and either cornmeal, rye or fermented rye: a preliminary study. Microorganisms. 2020;8(9):1363. doi:10.3390/microorganisms8091363

    44. Annunziata G, Verde L, Falco A, Colao A, Muscogiuri G, Barrea L. Very low energy ketogenic therapy (vlekt), not only a price matter: a comparison study on average costs of vlekt with fresh foods and replacement meals. Nutrients. 2025;17(3):422. doi:10.3390/nu17030422

    45. Chen B, Hong S, Wang Y, Hu Q, Ma D. Efficacy of meal replacement products on weight and glycolipid metabolism management: a 90-day randomized controlled trial in adults with obesity. Nutrients. 2024;16(19):3284. doi:10.3390/nu16193284

    46. Klein P, Tyrlíková I, Pirolli N, Barber A, Tyrlík M, Goldman L. Pilot study of “classic” 3:1 ketogenic diet complete meal replacement treatment of obesity-related type 2 diabetes. J Obes Chronic Dis. 2019;3(1):xx. doi:10.17756/jocd.2019-024

    47. Kempf K, Röhling M, Banzer W, et al. High-protein, low-glycaemic meal replacement improves physical health-related quality of life in high-risk persons for metabolic syndrome—a subanalysis of the randomised-controlled acoorh trial. Nutrients. 2022;14(15):3161. doi:10.3390/nu14153161

    48. Astbury N, Piernas C, Hartmann‐Boyce J, Lapworth S, Aveyard P, Jebb S. A systematic review and meta‐analysis of the effectiveness of meal replacements for weight loss. Obes Rev. 2019;20(4):569–587. doi:10.1111/obr.12816

    49. Barrea L, Caprio M, Camajani E, et al. Ketogenic nutritional therapy (kenut)—a multi-step dietary model with meal replacements for the management of obesity and its related metabolic disorders: a consensus statement from the working group of the club of the Italian society of endocrinology (sie)—diet therapies in endocrinology and metabolism. J Endocrinol Invest. 2024;47(3):487–500. doi:10.1007/s40618-023-02258-2

    50. Kempf K, Röhling M, Banzer W, et al. High-protein, low-glycaemic meal replacement decreases fasting insulin and inflammation markers—a 12-month subanalysis of the acoorh trial. Nutrients. 2021;13(5):1433. doi:10.3390/nu13051433

    51. Chen L, Ludin A, Shahar S, Manaf Z, Tohit N. Meal replacement in dietary management of type-2 diabetes mellitus: a scoping review protocol. Syst Rev. 2020;9(1):xx. doi:10.1186/s13643-020-01517-0

    52. Skeldon M, Harris D, Dent R, Shiau J. A comparison of virtual and in person delivery of a full meal replacement program for obesity. Obes Sci Pract. 2023;10(1):e718. doi:10.1002/osp4.718

    53. Gómez J, Izaola-Jáuregui O, Primo D, et al. Effect of two meal replacement strategies on cardiovascular risk parameters in advanced age patients with obesity and osteoarthritis. Nutrients. 2020;12(4):976. doi:10.3390/nu12040976

    54. Maston G, Franklin J, Gibson A, et al. Attitudes and approaches to use of meal replacement products among healthcare professionals in management of excess weight. Behav Sci. 2020;10(9):136. doi:10.3390/bs10090136

    55. Andela S, Burrows T, Baur L, Coyle D, Collins C, Gow M. Efficacy of very low‐energy diet programs for weight loss: a systematic review with meta‐analysis of intervention studies in children and adolescents with obesity. Obes Rev. 2019;20(6):871–882. doi:10.1111/obr.12830

    56. Lin T, Guo Y, Gordon C, et al. Association between sugar-sweetened beverage consumption as meal substitutes, workload, and obesity in nurses: a cross-sectional study. Int J Environ Res Public Health. 2019;16(24):4984. doi:10.3390/ijerph16244984

    57. Kundi Z, Lee J, Pihlajamäki J, et al. Dietary fiber from oat and rye brans ameliorate western diet–induced body weight gain and hepatic inflammation by modulating short‐chain fatty acids, bile acids, and tryptophan metabolism. Mol Nutr Food Res. 2020;65(1):1900580. doi:10.1002/mnfr.201900580

    58. de Wit N, Derrien M, Bosch-Vermeulen H, et al. Saturated fat stimulates obesity and hepatic steatosis and affects gut microbiota composition by an enhanced overflow of dietary fat to the distal intestine. Am J Physiol Liver Physiol. 2012;303:G589–G599. doi:10.1152/ajpgi.00488.2011

    59. Pollicino F, Veronese N, Dominguez LJ, Barbagallo M. Mediterranean diet and mitochondria: new findings. Exp Gerontol. 2023;176:112165. doi:10.1016/j.exger.2023.112165

    60. He Y, Zhang R, Yu L, et al. Pparγ acetylation in adipocytes exacerbates bat whitening and worsens age-associated metabolic dysfunction. Cells. 2023;12(10):1424. doi:10.3390/cells12101424

    61. Riess ML, Elorbany R, Weihrauch D, Stowe DF, Camara AKS. PPARγ-independent side effects of thiazolidinediones on mitochondrial redox state in rat isolated hearts. Cells. 2020;9(1):252. doi:10.3390/cells9010252

    62. Kozuka C, Yabiku K, Sunagawa S, et al. Brown rice and its component, gamma-oryzanol, attenuate the preference for high-fat diet by decreasing hypothalamic endoplasmic reticulum stress in mice. Diabetes. 2012;61(12):3084–3093. doi:10.2337/db11-1767

    63. Fujiwara H, Hiraoka R, Kawashima Y. The absorption, elimination, distribution and metabolism of triterpene alcohol ferulate (y-Oryzanol). Yakubutsu Ryoho. 1972;5:123–130.

    64. Colangeli LE, Escobar Marcillo DI, Simonelli V, et al. The crosstalk between gut microbiota and white adipose tissue mitochondria in obesity. Nutrients. 2023;15(7):1–19.

    65. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology. Human gut microbes associated with obesity. Nature. 2006;444(7122):1022–1023. doi:10.1038/4441022a

    Continue Reading

  • ‘Sustainable ambition’ is the key to success, two-time founder says

    ‘Sustainable ambition’ is the key to success, two-time founder says

    Earlier in her career, Amanda Goetz viewed her unrelenting determination as a “badge of honor.”

    “I always took it as a sign of pride that I could push myself,” she says.

    Goetz, 39, is a longtime marketing executive and two-time startup founder, as well as the creator of “Life’s A Game,” a popular newsletter for aspiring multi-hyphenates.

    During the pandemic, Goetz kicked her career ambitions into high gear: she left her corporate job and built her second startup, CBD-based product company House of Wise, all while navigating a difficult divorce and solo-parenting her three small children.

    From the outside, Goetz was thriving, but juggling all of her responsibilities with no breaks caused severe burnout, she says.

    While raising capital for House of Wise, Goetz ended up in the hospital twice in one week from what she later learned were panic attacks.

    “You can handle a lot, until your body just says ‘no,’” she says.

    That was a pivotal moment for Goetz: “I realized that you can have it all, just not all at once, and I needed to shift things around and reprioritize my life.”

    Finding time for “proactive rest”

    Goetz describes her previous attitude toward work as “toxic grit,” which she defines as “hustle without intention.”

    “So many of us, especially as ambitious people, are running towards these goals with no checkpoints to understand if that’s even still something we want,” she says.

    In order to get a handle on her priorities, Goetz developed a new strategy for “sustainable ambition”: instead of working until she crashes, Goetz intentionally schedules periods of hard work and rest.

    “I realized that I needed to alleviate the intensity that was happening in my life before my body would force me to do it,” she says.

    According to Goetz, finding balance isn’t necessarily the goal: it’s natural, and often necessary, to adjust our priorities around big life events.

    For example, Goetz is currently “hustling” ahead of publishing her first book, “Toxic Grit: How to have it all and (actually) love what you have,” which debuts in October. Afterwards, she plans to put work on the back burner for a couple months to relax and spend time with family.

    “It’s about building a rhythm into your life of proactive rest instead of waiting until your body demands it,” she says.

    Every few weeks, Goetz reevaluates which areas of her life she needs to invest more energy into. She’s found success in scheduling her work and rest cycles in a 2:1 cadence.

    “It can mean two weeks of really pushing at work, and then one week where you kind of take the foot off the pedal and say, OK, I’m going to leave every day at 5 o’clock on the dot, I’m shutting my computer, and I’m going to go see friends,” she says.

    Those “bare minimum” periods allow her to create space for the other important aspects of her life, she says.

    “If I just let the ambitious side of myself call all the shots, I would work all the time,” she says. “But that’s not what I want for my full life.”

    Last Chance: Want to stand out, grow your network, and get more job opportunities? Sign up for Smarter by CNBC Make It’s new online course, How to Build a Standout Personal Brand: Online, In Person, and At Work. Learn how to showcase your skills, build a stellar reputation, and create a digital presence that AI can’t replicate. Sign up today with coupon code EARLYBIRD for an introductory discount of 30% off the regular course price of $67 (plus tax). Offer valid July 22, 2025, through September 2, 2025.

    Plus, sign up for CNBC Make It’s newsletter to get tips and tricks for success at work, with money and in life, and request to join our exclusive community on LinkedIn to connect with experts and peers.

    Continue Reading

  • Ministry of Health marks Public Health Week with 7 key themes

    Ministry of Health marks Public Health Week with 7 key themes

    The Ministry of Health will celebrate Public Health Week from Sept. 3-9 across Türkiye with seven days of activities, each focusing on a different theme.

    Associate professor Dr. Muhammed Emin Demirkol, director general of Public Health at the Ministry of Health, told Anadolu Agency (AA) that this year’s Public Health Week will be celebrated with great enthusiasm.

    Emphasizing the importance of preventive health services, Dr. Demirkol stated that they aim to provide active services under the “Healthy Türkiye Century” program, utilizing a health model that protects, promotes and fosters health.

    Dr. Demirkol noted that the Public Health Directorate is heavily involved in protecting citizens’ health and said: “We aim to reach citizens this week by organizing intensive activities on topics such as health literacy, family medicine and the fight against addiction, both to raise awareness of health protection and to promote our services.”

    He added that each day of Public Health Week will feature a different theme. “We want to proceed with seven themes in seven days. On Sept. 3, with the slogan ‘May your breath be smoke-free, your life be healthy,’ we focus on our vision of a smoke-free Türkiye. We also want to remind citizens about our hotlines 171, 191 and 184, as well as our green detector program.”

    On Sept. 4, under the motto “Early Diagnosis in Cancer Saves Lives,” services provided in breast, colorectal and cervical cancer screenings at Healthy Life Centers, KETEM units and family medicine practices will be highlighted. He also stated that SMS reminders sent to citizens for cancer screenings have received very positive responses.

    Dr. Demirkol noted that Healthy Life Centers will be promoted on Sept. 5 and reported that the number of such centers has reached 320.

    He added, “These centers provide effective services with physiotherapists, dietitians, psychologists, social workers, child development specialists, and smoking cessation clinics, and there is at least one in each province working alongside family medicine to structure healthy living. We also have academies. So far, nearly 100,000 graduates have come through our Healthy Life Academies.”

    “We aim to increase health literacy, explain why antibiotics should only be used under doctor supervision, raise awareness about the harms of smoking, emphasize the importance of early cancer diagnosis, promote physical activity and combat obesity,” he added.

    Dr. Demirkol emphasized that Healthy Life Centers are among the most important places where health is fundamentally protected and added, “With digital integration, our referrals to Healthy Life Centers have increased significantly, resulting in a 50% rise in applications. We want citizens to be aware of these centers and visit them as easily as they would a shopping mall. We also encourage including Healthy Life Centers in weekend activities. We aim to place these centers at the heart of everyday life. Soon, we will also establish youth, baby and children’s academies within these centers.”

    On Sept. 6, under the theme “Manage your digital world, don’t miss life,” the focus will be on combating digital addiction. Demirkol emphasized that, as with tobacco, they aim to take strong measures against digital addiction. He noted that children’s excessive use of phones and social media negatively affects not only their academic success but also their social relationships.

    On Sept. 7, under the theme “Move, eat right, live healthy,” activities will focus on promoting an active lifestyle and combating excess weight, both key components of a healthy lifestyle.

    On Sept. 8, under the theme “Your nearest family doctor,” family medicine will be promoted. “We aim to introduce family medicine as the first point of contact in life and explain the services provided through this system,” he stated.

    Finally, on Sept. 9, in collaboration with the Directorate General for Health Promotion, the “Healthy Child, Healthy Future” program will be conducted in partnership with the Ministry of National Education to support children’s journey toward becoming health ambassadors.

    “In all 81 provinces, coordinated with our provincial health directorates and districts, we will work with governors, mayors and leading community figures to implement seven days of intensive activities that introduce all our services in the most effective way. Our goal is to instill the motto ‘Healthy living is very important’ in citizens,” Dr. Demirkol concluded.

    The Daily Sabah Newsletter

    Keep up to date with what’s happening in Turkey,
    it’s region and the world.


    You can unsubscribe at any time. By signing up you are agreeing to our Terms of Use and Privacy Policy.
    This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.

    Continue Reading

  • The International Series confirms addition of Jakarta International Championship to 2025 season

    The International Series confirms addition of Jakarta International Championship to 2025 season

    JAKARTA, Indonesia – The inaugural Jakarta International Championship has been confirmed by the Asian Tour – adding an exciting new event to its schedule that will contribute further to a gripping end to the season.

    The tournament will boast prize money of $2 million (U.S.), making it Indonesia’s most lucrative golf tournament and enhancing the nation’s reputation for being a strong supporter of professional golf in the region.

    The Jakarta International Championship will be played at one of the country’s most-renowned venues, Damai Indah Golf – PIK course, from Oct. 2-5, and will be the 13th event of the season on the Asian Tour.

    In addition, it will be part of The International Series – 10 upper-tier events on the Asian Tour that offer a direct pathway to the LIV Golf League. It is the fifth stop of the year on the Series.

    Importantly, the Government of Jakarta has lent its support to the tournament, which will mark the Asian Tour’s second visit of the season to Indonesia.

    The government is confident the collaboration will help promote Jakarta as a global city while providing an incredible opportunity for local players to compete at the highest level.

    Cho Minn Thant, Commissioner & CEO, Asian Tour, said: “Jakarta has been a popular and regular destination for the Asian Tour for decades, so staging the Jakarta International Championship has great meaning to us.

    “It is going to be an incredible addition to our schedule, not only bringing something new to our line-up but also adding importance in terms of a lucrative purse, outstanding golf course and place on The International Series.”

    Rahul Singh, Head of The International Series, said: “Jakarta is the perfect launch pad as we prepare for a thrilling conclusion to the season.

    “This tournament, which kickstarts the second half of the campaign, will go a long way to deciding who wins The International Series Rankings race and earns a spot on the LIV Golf League next season.

    “The Jakarta International Championship is another milestone moment in a successful season which is taking us to new markets and established destinations, once more showcasing the strength of The International Series brand.”

    The Asian Tour now has 13 events remaining this season – with over $20 million (U.S.) in prize money to play for. Action resumes next week at the Shinhan Donghae Open in Korea.

    (Photo courtesy of The International Series)

    Continue Reading

  • Most Gulf markets dip on weak oil prices – Reuters

    1. Most Gulf markets dip on weak oil prices  Reuters
    2. UAE: Loans, mortgages set to become cheaper soon amid US rate cut forecast  Khaleej Times
    3. Mideast Stocks: Most Gulf markets dip on weak oil prices  ZAWYA
    4. Major Gulf markets ease ahead of US economic data  Business Recorder
    5. A noticeable variance in the performance of Gulf financial markets at the end of today’s trading  المتداول العربي

    Continue Reading