Blog

  • New mechanism explains life-threatening pre-eclampsia at the molecular level

    New mechanism explains life-threatening pre-eclampsia at the molecular level

    Pre-eclampsia is one of the most serious complications in pregnancy, affecting millions of women and newborns worldwide and potentially life-threatening. Despite intensive research, it has remained unclear what processes in the…

    Continue Reading

  • Tong Liya makes stage return as she embarks on national tour

    Tong Liya makes stage return as she embarks on national tour

    Tong Liya at the launch ceremony of a national tour of Here, and Beyond. (PROVIDED TO CHINA DAILY)

    At 42, Tong Liya has returned to a stage that once defined her dreams — this time, as a dancer.

    After years of focusing on her acting career, she…

    Continue Reading

  • How Companies Are Reframing Climate Communication in 2025 – FTI Consulting

    1. How Companies Are Reframing Climate Communication in 2025  FTI Consulting
    2. McKinsey at Climate Week: Scaling Innovation, Accelerating Opportunity  McKinsey & Company
    3. What Has Climate Week NYC Taught Us In The Lead Up To COP30?  Forbes
    4. What You Need To Know From Climate Week NYC and the Path to COP30  Morningstar
    5. 5 Indigenous Takeaways From NYC Climate Week  NDN Collective

    Continue Reading

  • Timing of Estrogen Therapy Crucial for Women

    Timing of Estrogen Therapy Crucial for Women

    CLEVELAND, Ohio (Oct 21, 2025)—Menopause may take a toll on women physically and emotionally due to declining estrogen levels. For some, the use of hormone therapy has proven valuable in managing bothersome menopause symptoms. A new…

    Continue Reading

  • Ninja Gaiden 4 revives series with kinetic carnage, clumsy platforming

    Ninja Gaiden 4 revives series with kinetic carnage, clumsy platforming

    Hot on the heels of the rebirth of Joe Musashi and Sega’s Shinobi series, yet another famed ninja is back from the dead: Ryu Hayabusa returns with an all-new adventure in Ninja Gaiden 4, the first mainline…

    Continue Reading

  • Robotron. Code and Utopia – Announcements

    Robotron. Code and Utopia – Announcements

    With works by Karl-Heinz Adler, Tina Bara, Horst Bartnig, Nadja Buttendorf, Carlfriedrich Claus, Karl Clauss Dietel, Georg Eckelt, Antye Guenther, Su Yu Hsin, Margret Hoppe, Francis Hunger, knowbotiq, Irma Markulin, Helga Paris, A. R. Penck,…

    Continue Reading

  • Yin-Hua Li-Shi Decoction alleviates atopic dermatitis through regulati

    Yin-Hua Li-Shi Decoction alleviates atopic dermatitis through regulati

    Introduction

    Atopic dermatitis (AD) is the most prevalent chronic and pruritic inflammatory skin disease,1 characterized by epidermal desquamation, intense pruritus, and lichenified lesions.2 This disease affects 20% of children and 4% of adults globally, causing significant quality-of-life impairment due to chronic pruritus, sleep disturbance, and psychosocial burden.3,4 Common antigens triggering AD include house dust mites, Staphylococcus aureus enterotoxins, food allergens (eg, egg, milk), and environmental pollens.5 AD is a continuous process from acute to chronic phase, accompanied by three interconnected pathological mechanisms, skin barrier dysfunction, immune dysregulation, and abnormal neural signaling.6–8 Skin barrier dysfunction manifests as downregulated expression of barrier genes, such as loricrin (LOR), filaggrin (FLG), and elongation of very long-chain fatty acid (ELOVL).9–11 Immune dysregulation mainly results from sustained inflammation mediated by type I, II, and III adaptive immune responses driven by differentiated CD4+ T cells (Th1/ Th2/ Th17).12 Abnormal neural signaling manifests as sensory nerve hyperinnervation, upregulated pruritogens, such as interleukin (IL) −31 and thymic stromal lymphopoietin (TSLP), and neuroimmune crosstalk amplifying itch-scratch cycles.13 These three components reinforce each other.14 Environmental antigens first damage the skin barrier, which activates dendritic cells (DCs) and recruits Th cells. Activated Th cells release cytokines like IL-4, IL-13, and IL-31. These cytokines further break down barrier proteins and sensitize sensory nerves. Finally, neurogenic inflammation causes tissue damage which makes the immune system overactive, establishing a harmful cycle that worsens AD.15

    For decades, the cornerstone of AD treatment has relied on topical corticosteroids and topical calcineurin inhibitors,16 whose adverse effects include skin atrophy, pigmentation changes, anaphylaxis, and potential complications (eg, folliculitis or tinea infection) after drug discontinuation.17 Recent advances in biological agents and small molecule inhibitors have introduced novel treatment options for AD, such as dupilumab (targeting IL-4Rα), tralokinumab/ lebrikizumab (targeting IL-13), CIM331/ nemolizumab (targeting IL-31R), crisaborole (targeting Phosphodiesterase-4), and tofacitinib (targeting Janus kinase 1/ 3).18 At the same time, because of potential safety problems, high recurrence rate and high economic burden of the AD, the use of these drugs is limited. In this context, traditional Chinese medicine (TCM) has attracted the attention of clinicians and AD patients as a complementary treatment for AD, especially in and around China, due to its abundance of natural anti-inflammatory compounds.19

    Yin-Hua Li-Shi Decoction (YLD) is a TCM approved by medical institutions, which is composed of six herbs aimed at removing dampness. It has a long history of being used for the treatment of AD. The chlorogenic acid and luteoloside derived from honeysuckle in YLD has been shown to inhibit the secretion of pro-inflammatory cytokines such as IL-6 and TSLP that acts on multiple cell lineages, including macrophages, mast cells, neutrophils, DCs, and T cells, ultimately suppressing moderate to severe immune responses.20,21 However, the specific mechanisms underlying YLD in the treatment of AD were still unclear and lacked systematic validation.

    The present study was aimed at exploring the therapeutic effects and mechanisms of YLD in AD. We used MC903-induced AD-like mouse model to evaluate YLD therapeutic benefits for AD and to clarify the mechanisms by which it regulated immunity and restored the skin barrier.

    Material and Methods

    Drugs and Reagents

    All herbs were purchased from WanShiCheng Pharmaceutical Co., Ltd. (Shanghai, China), and authenticated according to the Pharmacopoeia of the People’s Republic of China 2020 Edition by Professor Huijun Pan from Shanghai Skin Disease Hospital, School of Medicine, Tongji University. Voucher specimens of these herbal materials were deposited at the Shanghai Skin Disease Hospital with reference numbers YL1-6. Chlorogenic acid and specnuezhenide used as standard compounds were from Meilunbio (Shanghai, China). MC903 was purchased from Macklin Biochemical Co., Ltd. (#C833062, Shanghai, China). Antibodies used in the study were obtained from the following sources: anti-FLG (#GTX23137, GeneTex, Beijing, China), anti-LOR (#A21039, ABclonal, Wuhan, China), anti-ELOVL6 (#A21094, ABclonal, Wuhan, China), anti-TSLP (#ab188766, Abcam, Cambridge, U.K)., anti-β-Actin (#AC026, ABclonal, Wuhan, China), HRP-conjugated Goat anti-Rabbit IgG (H+L) (#AS014, ABclonal, Wuhan, China), anti-IL-4 (#25-7042-42, Invitrogen, California, USA), anti-IL-17A (#506904, Biolegend, California, USA), anti-IFN-γ (#563376, BD Biosciences, New Jersey, USA), anti-rabbit IgG (H+L) Ab HRP Affinity purified polyclonal (#95058–730, KPL, Maryland, USA), anti-CD4 (#ab183685, Abcam, Cambridge, U.K)., anti-CD4 (#555349, BD Biosciences, New Jersey, USA), anti-CD8 (#100733, Biolegend, California, USA), anti-CD86 and anti-CD80 (#561962 and #561955, BD Biosciences, New Jersey, USA). ELISA kits used for analysis were IL-4 (#EM3199M, WellBio, Shanghai, China), IL-13 (#EM3167M, WellBio, Shanghai, China), TNF-α (#RK00027, ABclonal, Wuhan, China), and IgE (#RK00170, ABclonal, Wuhan, China). Annexin V-FITC/PI Apoptosis Detection Kit was purchased from Vazyme Biotechnology Co., Ltd (#A211-02, Nanjing, China). All other chemicals used in the experiments were of analytical grade.

    Preparation of YLD

    The YLD consists of six Chinese herbal medicines, including Jinyinhua (Honeysuckle), Shanyao (Yam, Siberian), Huangjing (Solomonseal rhizome), Digupi (Cortex lycii radices), Nvzhenzi (Fructus ligustri lucidi), Yiyiren (Coix seed), and the daily dose of adult clinical YLD is 54 g of crude drugs (Table 1). YLD extraction is the first step to adding 8 times amount of water in crude drugs (w/w), decocting 2 h after filtering, the rest of the student to join six times the amount of water decoction 1 h again. The above water extract was concentrated to 40 mL using a EYELA N-1300D-WB rotary evaporator, and the final YLD concentration had a crude drug equivalent of 6.00 g/mL. The concentration was appropriately diluted to crude drug equivalents of 3.00 g/mL and 1.50 g/mL to form medium and low doses, respectively. All prepared YLD samples were stored at 4 °C until subsequent use.

    Table 1 The Herbal Composition of YLD

    Quality Control of YLD

    Fingerprint Identification

    High-performance liquid chromatography (HPLC) was employed to construct a fingerprint profile and evaluate the quality of YLD and for quality control. Chlorogenic acid and specnuezhenide were dissolved in methanol to prepare a stock solution. The YLD reference solution (R) (200 μg/mL) was prepared by diluting the original 1.50 g/mL YLD solution and then filtering through a 0.45 μm membrane filter. The concentration of YLD extract (g crude herb/mL) represented a drug extract ratio, which was calculated by the quotient of total dry herb mass over final decoction volume.22 The YLD HPLC test solution (YLD sample) (150 μg/mL) was prepared by diluting the original 1.50 g/mL YLD solution and then filtering through a 0.45 μm membrane filter. The quality consistency validation and methodological parameters are detailed in Supplementary Information 1 Table S1. The content of the reference compounds in YLD was calculated based on the pre-constructed standard curves of chlorogenic acid and specnuezhenide. The fingerprint profile of YLD was identified by comparing the relative retention time and ultraviolet characteristics of the internal reference with the YLD test sample. The quality of YLD was controlled by comparing the similarity of the fingerprint profiles among 10 batches of YLD (S1-S10). To ensure batch consistency, all 10 tested batches were derived from the same cultivation batch of raw plants.

    The liquid chromatography system used was an Waters Alliance HPLC (Waters, Massachusetts, USA), consisting of an E2695 separation module and a 2998 photodiode array detector with an autosampler. HPLC was performed on an ZORBAX Eclipse Plus C18 column (4.6 mm × 250 mm, 5 μm) (Agilent, California, USA). The mobile phase consisted of solvent A (acetonitrile) and solvent B (0.1% phosphoric acid water), and the gradient elution conditions were shown in Table 2. The UV absorption wavelength was set at 230 nm, column temperature at 25 °C, injection volume at 10 μL, and flow rate at 1.0 mL/min.

    Table 2 HPLC Gradient Evaluation Conditions of YLD

    Ingredient Identification

    The components of YLD water extract were identified by liquid chromatography-tandem mass spectrometry (LC-MS). The YLD LC-MS test solution (600 μg/mL) was prepared by diluting the original 1.50 g/mL YLD solution and then filtering through a 0.45 μm membrane filter. The YLD water extract was analyzed using a LC-MS system composed of an ACQUITY UPLC I-Class HF ultra-high performance liquid chromatography coupled with a QE high-resolution mass spectrometer. The mobile phase consisted of solvent A (0.1% formic acid aqueous solution) and solvent B (acetonitrile). The sample was separated at a flow rate of 0.35 mL/min, and the gradient elution conditions are shown in Table 3.

    Table 3 LC-MS Gradient Elution Conditions of YLD

    LC-MS detection was performed using an ACQUITY UPLC HSS T3 column (100 mm × 2.1 mm, 1.8 μm) (Waters, Massachusetts, USA). Mass spectrometry data acquisition was carried out in electrospray ionization (ESI) positive and negative modes, and the data-dependent acquisition (DDA) mode was used, with a mass range of m/z 90 to 1300. The capillary temperature was set to 320 °C, and the probe heating temperature was set to 350 °C.

    Cell Culture

    Murine macrophage cell line (RAW264.7) and human keratinocyte cell line (HaCaT) were obtained from the National Collection of Authenticated Cell Cultures (Shanghai, China). RAW264.7 and HaCaT cells were cultured in DMEM (Gibco, USA) supplemented with 10% FBS (Gibco, USA) and 1% penicillin-streptomycin (Beyotime, China). All cultures were maintained in a humidified atmosphere with 5% CO2 at 37 °C.

    Animals

    Male BALB/c mice aged 8–9 weeks (weighing 22–25 g) were obtained from the SiPeiFu Biotechnology Co., Ltd. (Shanghai, China) and housed under SPF conditions. The mice were maintained at a room temperature of 26 °C with a relative humidity of 40% and a 12:12-hour light/dark cycle. They were provided with standard mouse maintenance feed and water ad libitum.

    Apoptosis Assay

    HaCaT cells were cultured in 6-well plates (30×104 cells per well) for 24 h. The 6-well plate was then subjected to a 24-hour incubation in the following groups: the PBS group (0 µg/mL YLD) and the PBS+YLD group (150 µg/mL YLD). Apoptosis of HaCaT cells was assessed using propidium iodide (PI) and fluorescein isothiocyanate (FITC)-labeled Annexin V staining, followed by detection and analysis with Navios 6 COLORS/2 LASER flow cytometer and FlowJo Software.

    Analysis of Macrophage Phenotype

    To induce the inflammatory phenotype of macrophages, RAW264.7 cells were treated with 0.5 µg/mL LPS and 2 ng/mL IFN-γ. 1 h prior to LPS and IFN-γ stimulation, RAW264.7 cells were treated with YLD (150 µg/mL) to assess the impact of YLD on macrophages. After 18 h, cells were incubated with CD86 and CD80 antibodies for 30 minutes. The Navios 6 COLORS/2 LASER flow cytometer and FlowJo software were used to evaluate the differentiation of YLD in suppressing the inflammatory phenotype of macrophages.

    Induction of AD-Like Mice Model and YLD Administration

    Fifty male BALB/c mice were induced with AD by repeated topical application of MC903 on their ears for 14 days. The mice were randomly divided into five groups (n = 10/group): control mice treated with ethanol (Ethanol), control mice treated with MC903 alone (MC903), experimental mice treated with both MC903 and low-dose YLD (YLD-L), experimental mice treated with both MC903 and medium-dose YLD (YLD-M), and experimental mice treated with both MC903 and high-dose YLD (YLD-H). In brief, Ethanol group was topically applied with 20 μL of ethanol on the ears daily, while other groups were sensitized with 2 nmol of MC903 on the ears daily for 15 days (Day 0 to Day 14). From Day 1 to Day 14, Ethanol group received daily treatment with ethanol, and YLD-treated groups received oral administration after diluting YLD extract. Three different dosage levels were used: low, medium, and high, corresponding to final concentrations of 1.50 g/mL, 3.00 g/mL, and 6.00 g/mL of the YLD extract, respectively. The lose dose was calculated based on the clinical dose converted to mouse dose referring to the FDA human dose conversion table for animal doses. And each 20 g mouse received 0.15 mL decoction orally daily. The efficacy of the treatment was evaluated 12 h after the last administration (Day 15). Blood samples were collected by retro-orbital bleeding to obtain serum samples, and tissue samples were collected after cervical dislocation for subsequent experimental analysis.

    AD-Related Evaluation

    During the study, daily monitoring of mice included recording body weight using an electronic balance, measuring transepidermal water loss (TEWL) using an intelligent skin analyzer (#W-2100, Yizi-moqi, Guangzhou, China), assessing ear thickness using vernier calipers, and scoring AD (SCORAD). The SCORAD consisted of the following items: (i) pruritus/itching, (ii) erythema/hemorrhage, (iii) edema, (iv) excoriation/erosion, and (v) desquamation/dryness. Each symptom was graded as follows: 0 (no symptoms), 1 (mild), 2 (moderate), or 3 (severe). The total score for AD ranged from 0 to 15. Additionally, spleen index was measured on Day 15. The spleen index was calculated using the formula:

    Spleen Index (mg/g) = Spleen weight (mg) /Mouse weight (g).

    Histopathological Analysis

    Tissue specimens from the inflamed area of the ear were fixed in formalin and embedded in paraffin. For hematoxylin and eosin (HE) staining, paraffin sections were stained with hematoxylin and eosin. Samples were examined and captured using an optical microscope (QT50GS, Yuehe, Shanghai, China), and the epidermal thickness was counted at five randomly selected sites under 40× and 100× magnification. For toluidine blue (TB) staining, paraffin sections were stained with TB. Samples were examined and captured using an optical microscope, and the number of mast cells was counted at five randomly selected sites under 40× magnification.

    Enzyme-Linked Immunosorbent Assay (ELISA)

    Mouse blood samples were centrifuged at 12,000 g and 4 °C for 20 minutes to separate the upper serum. ELISA was performed to measure the concentrations of IL-4, IL-13, TNF-α, and IgE in the serum. The measurements were carried out using commercially available ELISA kits following the manufacturer’s instructions.

    Immunohistochemical Analysis

    Paraffin-embedded tissue specimens were incubated overnight with anti-FLG (1:500), anti-TSLP (1:500), and anti-CD4 (1:500). After washing with PBS, Anti-Rabbit IgG (H+L) Ab HRP Affinity purified polyclonal (1:200) were added and incubated for 1 h, followed by DAB staining. The sections were counterstained with hematoxylin. Immunohistochemically stained slides were examined and captured using an optical microscope, and records were taken at five randomly selected sites under 40× magnification.

    Western Blot Analysis

    Tissue protein lysates were obtained in RIPA buffer (Beyotime, Shanghai, China) containing PMSF (Beyotime, Shanghai, China). Protein concentration was quantified using a BCA assay kit (Beyotime, Shanghai, China). Proteins were separated by 10% SDS-PAGE (Servicebio, Wuhan, China) and then transferred onto PVDF membranes. The membranes were incubated overnight at 4 °C with corresponding primary antibodies: anti-FLG, anti-LOR, anti-ELOVL6, anti-TSLP (1:1000), and anti-β-Actin (1:2000). Subsequently, the membranes were incubated with secondary HRP-conjugated (1:1000) for 1 h to detect antibody binding. β-Actin was used as an internal reference. The target protein signals were analyzed using Image J application.

    Analysis of Spleen T Cells

    Spleen tissue was dissociated and filtered through a 70 μm cell strainer. The cell suspension was centrifuged, and resuspended in PBS to prepare a single-cell suspension. Red blood cells in the splenocytes were removed using ACK lysis buffer (#A1049201, Thermo Fisher Scientific, Massachusetts, USA) and washed before staining. Cells were incubated at 4 °C in the dark for 30 minutes with 2.5 μL of CD4 antibody and CD8 antibody. Subsequently, a permeabilization wash buffer was added. Then, 2.5 μL of IL-4, IFN-γ, and IL-17A antibodies were added, and the cells were incubated at 4 °C for 30 minutes. The analysis was performed using a Beckman moflo Astrios EQ flow cytometer with FlowJo Software, and the proportions of Th1, Th2, and Th17 cells in CD4+ T cells were recorded.

    Statistical Analysis

    Data analysis and graph plotting were performed using GraphPad software (version 8.0). Normality of data distribution was verified using the Shapiro–Wilk test (α=0.05). Normally distributed data were analyzed by one-way ANOVA with Tukey’s post hoc test. Data were expressed as mean ± standard deviation (SD), and one-way ANOVA was used for comparisons among multiple groups. A P-value < 0.05 was considered statistically significant.

    Results

    Fingerprint and Component Identification of YLD

    To ensure the stability of YLD, chlorogenic acid and specnuezhenide were selected as reference compounds and studied as specific indicators of YLD (Figure 1A). HPLC analysis was performed on the reference compounds, with a retention time of 12.7 minutes for chlorogenic acid and 26.3 minutes for specnuezhenide. The peak shape of both chlorogenic acid and specnuezhenide exhibited Gaussian distribution, with sharp and symmetrical peaks.

    Figure 1 HPLC fingerprint of YLD. Chromatogram at 230 nm showing the reference compounds and the chromatogram (A), and fingerprints of ten different batches (B) of YLD. (C) Distribution chart of the top 10 components in terms of content in YLD. (D) BPC chromatograms of YLD in positive and negative ion modes.

    The standard curves of chlorogenic acid and specnuezhenide reference compounds were provided in the Supplementary Information 1 Tables S2 and S3, Figure S1a and b). Based on the standard curves of these two reference compounds, the content of chlorogenic acid in YLD (clinical) was determined to be 2.697 mg/mL and the content of specnuezhenide was determined to be 8.405 mg/mL. We conducted fingerprint analysis of YLD from 10 batches (Figure 1B), and the highest similarity exceeded 0.9006 (Supplementary Information 1 Table S4). The above analysis data indicated that YLD was stable and of controllable quality.

    The chemical components of the YLD extract included a total of 705 compound molecules, which were identified using LC-MS. These 705 compounds were classified chemically based on their quantity and content, as shown in Figure 1C. The Base Peak Chromatogram (BPC) in both positive and negative ion modes is presented in Figure 1D. The top 15 most abundant compounds among the 705 metabolites identified are Secologanic acid, Citric acid, Cryptochlorogenic acid, GL3, Specnuezhenide, Secoxyloganin, Mannoheptulose, Sucrose, Turanose, Swertiamarin, D-Galactose, Verbenalol, Dambose, 5-Hydroxymethylfurfural, 6α-dihydrocornic acid, and Salidroside (Table 4). The quantitative and qualitative identification results of the 705 metabolites in the YLD extract are provided in the Supplementary Information 2.

    Table 4 Top 15 Most Abundant Compounds Among YLD Metabolites

    YLD Did Not Induce Apoptosis in HaCaT

    Flow cytometry, utilizing PI and membrane-associated protein V-FITC staining, was employed to assess the impact of YLD on apoptosis in HaCaT. After treatment with 150 μg/mL of YLD for 24 h, there was an increase in the percentage of apoptotic HaCaT but was no significant difference (Figure 2A). However, the viability of HaCaT remained above 80%. Generally, a cell survival rate greater than 80% post-drug treatment is considered indicative that the drug does not induce apoptosis.

    Figure 2 In vitro pharmacological activity of YLD. (A) Apoptotic HaCaT treated with 150 μg/mL YLD for 24 h were detected by flow cytometry. (B) M1 macrophages were treated with YLD (150 μg/mL) for 24 h and then detected the M1-phenotype surface marker (CD86 and CD80) by flow cytometry. The data was collected from three independent experiments and was presented as a mean ± SD.

    YLD Inhibited Macrophage to M1 Differentiation

    Macrophages, essential phagocytic cells of the immune system, play a crucial role in coordinating innate immune responses.23 It is noteworthy that macrophages also possess antigen-presenting capabilities, facilitating the presentation of antigen peptides to T cells, thereby initiating adaptive immune responses.24 M1 macrophages represent classically activated macrophages that exhibit a pro-inflammatory phenotype, characterized by the production of high levels of cytokines such as IL-1β, IL-6, and TNF-α.25

    Stimulation of RAW264.7 with 0.5 µg/mL LPS and 2 ng/mL IFN-γ for 24 h induced the differentiation of macrophages into the M1 subtype. CD86 and CD80 are widely used as markers for M1 polarization, with their upregulation considered indicative of activated macrophages polarizing towards the M1 phenotype. Examination of M1 macrophages stimulated by LPS and IFN-γ revealed higher expression of the specific functional markers CD86 and CD80, suggesting the induction of M0 differentiation into M1. In comparison to the LPS + IFN-γ group, YLD treatment for 24 h significantly downregulated the positivity rates of CD86 and CD80, which indicated that YLD possessed the capability to inhibit the differentiation of macrophages into the inflammatory phenotype (Figure 2B).

    YLD Alleviated Clinical Symptoms of MC903-Induced AD in Mice

    The experimental design for the model construction in this study was shown in Figure 3A. MC903 is a vitamin D3 analog that has been widely used as an experimental drug for establishing AD animal models.26 Under the stimulation of MC903, keratinocytes in mouse skin express and secrete TSLP, which induces the development of immature DCs to a mature phenotype by binding to TSLP receptors on DCs.27,28 The activated DCs further initiate the differentiation of naive Th0 cells into Th2 subsets, thereby inducing Th2-mediated inflammatory response, down-regulating the expression of skin barrier related proteins, and promoting the production of allergen-specific IgE by B cells.29 Thus, the mechanism by which MC903 induces AD-like skin lesions is similar to the pathogenesis of human AD.30

    Figure 3 Improvement of AD symptoms in mice by YLD. (A) Schematic representation of the construction of the AD animal model and treatment regimen: BALB/c mice aged 9 weeks were administered MC903 at a dose of 2 nmol/ear for 15 consecutive days, followed by treatment with 0.2 mL of YLD-L, YLD-M, or YLD-H for 14 days. (B) Visual images/representative phenotypic manifestations of the ears of mice from the Ethanol group, MC903 group, YLD-L group, YLD-M group, and YLD-H group on day 15 of AD induction. (C) Daily SCORAD during YLD treatment. (D) Daily percentage change in body weight of mice during YLD treatment. (E) The TEWL of AD-like mice during YLD treatment. (F) Percentage change in ear thickness of mice during YLD treatment measured using a micrometer. Data were expressed as mean ± SD (n = 10 for each group). ***P < 0.001.

    Compared to the ethanol group, continuous stimulation with MC903 resulted in typical AD symptoms, significant ear epidermal swelling, erythema, and crust formation in mice (Figure 3B). The SCORAD, used to evaluate the severity of skin lesions in the MC903 group, reached 8.50 (Figure 3C). However, the YLD-M and YLD-H groups shown significantly reduced severity of skin damage compared to the MC903 group, with SCORAD of 6.08 and 6.62, respectively. Additionally, AD caused a decrease in body weight in the MC903 group, which was improved after oral administration of YLD. The YLD-M group exhibited the best control of body weight (Figure 3D). Moreover, while both the YLD-M and YLD-H groups shown therapeutic effects, it was observed that the YLD-H group caused adverse reactions in the gastrointestinal tract of AD-like mice.

    Comparing the dynamic changes in TEWL in the lesional skin of mice in each group during YLD treatment, the results showed that the TEWL of mice in the MC903 group continued to increase under the action of MC903. There were significant differences between the MC903 group and other groups. After YLD treatment, TEWL in AD mice was significantly down-regulated, but there was no significant difference in TEWL between gradient YLD treatment groups (Figure 3E). AD-like mice’s ear thickness changes due to ear swelling during YLD treatment were statistically analyzed. Prolonged stimulation with MC903 resulted in significant ear swelling in mice, with an average change rate in ear thickness of 40.61% in the MC903 group at the end of AD induction. However, the average change rate in ear thickness was 25.24% in the YLD-L group, 23.17% in the YLD-M group, and 17.49% in the YLD-H group (Figure 3F), which was weaker than the counterpart in the MC903 group.

    YLD Ameliorated Skin Lesions in AD Mice

    HE staining was used to compare the epidermal lesions in the ears of AD-like mice and the YLD-treated groups. Compared to healthy ears, MC903-induced AD-like ears exhibited apparent hyperkeratosis (with a thickness of up to 95.36), incomplete keratinization, thickened spinous layers, increased granular layer, and infiltration of numerous inflammatory cells and eosinophils in the dermis (Figure 4A). The epidermal thickness in healthy mice was 36.31. In the YLD-L group, the epidermal thickness in mice could be reduced to 71.01. However, results shown that the mice in the YLD-M and YLD-H groups gradually reduced these skin lesion symptoms and decreased epidermal thickness (69.7 in the YLD-M group and 62.52 in the YLD-H group). From the perspective of epidermal thickness, the therapeutic effects of YLD-L, YLD-M, and YLD-H in AD-like mice demonstrated a dose-dependent pattern.

    TB staining was used to process ear specimens for counting classical immune sentinel mast cells to compare the number of inflammatory cells in the ears of AD-like mice and the YLD-treated groups. Mast cells are considered classical immune cells implicated in itch sensation, and their excessive infiltration can directly contribute to itching behavior in AD-like mice. By comparing sections from the blank group and AD-like skin, it was observed that the infiltration of mast cells in the dermis and subcutaneous tissue of AD-like mice significantly increased, surpassing the levels in healthy mice. However, in AD-like mice administered YLD orally, the infiltration of mast cells in the dermis and subcutaneous tissue decreased, and this reduction exhibited a correlation with the dosage (Figure 4B).

    Figure 4 Improvement of skin lesions in AD-like mice by YLD. (A) HE staining of longitudinal cross-sections of ears and quantification histogram of stratum corneum thickness within the field of view. A solid black line represented the stratum corneum. Scale bar represented 100 μm. (B) TB staining of longitudinal cross-sections of ears and quantification histogram of mast cell infiltration within the field of view. Red arrows indicated mast cells. Scale bar represented 100 μm. Data were expressed as mean ± SD (n = 10 for each group). ***P < 0.001.

    YLD Suppressed Pro-Inflammatory Cytokines in Serum

    To evaluate the inflammatory status of mice, the concentrations of pro-inflammatory cytokines and an antibody in collected serum samples were measured using ELISA. Treatment with MC903 increased levels of IL-4, IL-13, TNF-α cytokines, and IgE antibodies in the serum (Figure 5A). However, YLD reduced the serum levels of these pro-inflammatory factors in AD-like mice. Mainly, in the YLD-H group, the mice shown a decrease of 49% in IL-4, 38% in IL-13, 38% in TNF-α, and 35% in IgE.

    Figure 5 YLD alleviated AD by maintaining barrier protein expression and inhibiting pro-inflammatory factors. (A) Serum ELISA results shown that YLD downregulated the levels of typical pro-inflammatory factors IL-4/13, TNF-α, and IgE antibodies in AD-like mice, exhibiting a dose-dependent relationship. (B) Western blot results of AD-like skin lesions demonstrated that compared to MC903, YLD maintained the expression of typical barrier proteins FLG, LOR, and ELOVL6 in the skin lesions of mice with AD-like symptoms, with a dose-dependent effect observed for LOR and ELOVL6. Furthermore, compared to MC903, YLD downregulated the expression of pro-inflammatory factor TSLP in the skin lesions of AD-like mice. (C) Immunohistochemistry results of AD-like skin lesions shown that YLD maintained the expression of barrier protein FLG and inhibited the expression of pro-inflammatory factor TSLP, with a dose-dependent effect observed for the inhibition of TSLP. Scale bar represented 100 μm. Data were expressed as mean ± SD (n = 10 for each group). **P < 0.01, ***P < 0.001.

    YLD Upregulated Barrier Proteins and Downregulated Pro-Inflammatory Factors in Lesional Skin

    Previous studies indicated that AD lesional skin exhibits defects or downregulation of FLG, LOR, and ELOVL barrier proteins. To investigate the role of YLD in improving the downregulation of barrier proteins in AD-affected skin, western blot analysis was performed to examine the expression levels of barrier proteins. As shown in Figure 5B, the expression of FLG, LOR, and ELOVL was downregulated in the skin of mice treated with MC903, consistent with impaired barrier function in AD skin. However, in mice treated with YLD, the expression of FLG, LOR, and ELOVL increased, with the most significant upregulation observed in the YLD-H group. YLD also exhibited a typical downregulation effect on the pro-inflammatory cytokine TSLP. These protein expression assessments suggested that the anti-AD results of YLD were achieved by upregulating barrier protein expression and downregulating inflammatory factors.

    Furthermore, the expression of FLG and TSLP in the skin was visually observed using immunohistochemistry (Figure 5C). Under continuous stimulation by MC903, the expression level of FLG in mice significantly decreased while TSLP expression increased. After YLD treatment, the expression level of FLG in AD lesional tissues recovered, and the highly expressed level of TSLP was controlled. These results were consistent with the findings from the western blot. However, in immunohistochemistry, dose-dependency was primarily reflected in FLG, and no obvious dose-dependency was observed for TSLP.

    YLD Inhibited Splenic Atopic Immune Responses

    To elucidate the mechanisms by which YLD regulated immune responses, immunohistochemistry analysis was used to analyze the infiltration of CD4+ T cells in lesional tissues and flow cytometry was used to analyze the quantity and proportion of Th1/ Th2/ Th17 immune cells in the spleens of mice from different treatment groups, aiming to characterize the regulation of YLD on CD4+ T cell subsets in AD-like mice. Compared to the skin tissue of healthy mice, CD4+ T cell infiltration in the epidermis of AD lesional skin significantly increased, indicating severe adaptive immune responses induced by MC903 (Figure 6A). After YLD-L, YLD-M, and YLD-H treatments, a gradient reduction in infiltrating CD4+ T cells in the epidermis and dermis was observed, showing a correlation with the dosage. In the YLD-H treatment, the infiltration of CD4+ T cells in lesional areas approached levels seen in normal skin. As a typical immune organ, the spleen exhibited morphological changes indicative of AD-like immune responses, such as volume reduction. The number of immune cells in the spleen can represent the degree of immune responses. Morphologically, the spleen shown different degrees of shrinkage after treatment with MC903. However, YLD treatment attenuated the degree of spleen shrinkage (Figure 6B). Significant differences were observed in the spleens of the YLD-L and YLD-H groups compared to the MC903 group, which was also reflected in the spleen index.

    Figure 6 YLD exerted therapeutic effects in AD by inhibiting the activation of immune cells in the lesional skin. (A) Immunohistochemistry results shown a significant reduction in CD4+ T cell infiltrates in mice epidermis and dermis of the lesional skin after YLD treatment, compared to MC903. A slight dose-dependent relationship was observed among the three doses of YLD treatment. Red arrows indicated positive signal CD4+ T cells. (B) YLD inhibited splenic atopic immune responses, improved the typical atopic symptom of spleen shrinkage in AD-like mice, and restored the spleen index. Among the different doses of YLD treatment, YLD-H exhibited the most optimal inhibition of splenic atopic immune responses. Data were expressed as mean ± SD (n = 10 for each group). ***P < 0.001.

    Flow cytometry results shown that the proportions of CD4+ T, CD8+ T cells, and Th1, Th2, and Th17 cells in the spleens of AD-like mice significantly increased (Figure 7). Compared to healthy mice, the proportions of Th1, Th2, and Th17 cells in AD-like mice approximately doubled, representing the occurrence of type I, II, and III adaptive immune responses. Oral administration of YLD at all three dosages reduced the proportions of CD4+ T and CD8+ T cells in the spleen, indicating that YLD slowed down excessive immune responses. Moreover, YLD exerted different degrees of regulatory effects on type I, II, and III adaptive immune responses, as evidenced by the downregulation of the proportions of Th1, Th2, and Th17 cells.

    Figure 7 YLD exerted therapeutic effects in AD by modulating the balance of Th1/ Th2/ Th17 cells. YLD downregulated the CD4+/ CD8+ ratio and effectively controls the differentiation of Th1/ Th2/ Th17 cells induced by MC903 in CD4+ T cells after YLD treatment, relieving type I, type II, and type III immune responses. Furthermore, the inhibitory effect on Th cells differentiation correlated with the dose of YLD, with YLD-H exhibiting the best inhibitory effect on Th cells differentiation.

    Discussion

    The predominant mechanisms underlying AD pathogenesis were the adaptive immune response mediated by Th cells and the downregulation of barrier genes such as LOR, FLG, and ELOVL6, leading to epidermal barrier impairment.31 In non-lesional phase of AD, allergen stimulation of the skin leaded to excessive scratching, initially compromising the skin barrier. This response activated epidermal langerhans cells (LCs) and dermal DCs,32 resulting in infiltration and low-level activation of various Th cell subsets (Th1/ Th2/ Th17/ Th22).33,34 During the acute phase of AD, Th2/ Th22 cells significantly increased and released multiple inflammatory mediators,35 such as Th2-associated cytokines IL-4, IL-5, IL-13, IL-31, C-C motif chemokine ligand 18 (CCL18), and Th22-associated cytokines IL-22, S100A proteins, leading to acute skin inflammation.36 The immunomodulatory cytokines IL-4 and IL-13 released by Th2 cells induce significantly reduced expression of FLG,37 LOR,38 and ELOVLs39 in differentiated keratinocytes, thereby inhibiting the production of antimicrobial peptides and promoting colonization by Staphylococcus aureus.40 These consequences further worsened skin barrier impairment. In the chronic phase of AD, apart from Th2/ Th22 cells, Th1/ Th17 cells contributed to epidermal remodeling and hyperplasia.41

    The safety and efficacy of TCM in alleviating AD have been well established, and they were commonly used as an adjunctive therapy in clinical AD treatment.42 This type of TCM formula contains plants with anti-inflammatory effects, which can exert synergistic effects through different mechanisms and then produce stable therapeutic effects.43 This analysis revealed abundant contents of chlorogenic acid, luteoloside, and specnuezhenide in YLD. Chlorogenic acid accounted for 13.06% and specnuezhenide accounted for 8.83% of YLD, respectively. In previous studies, these components have demonstrated anti-inflammatory activity through mechanisms including inhibition of the MAPK/ERK/JNK pathway, the NF-κB pathway, and the JAK2/STAT3 pathway.44–46 From the compositional point of view, YLD seems to have a therapeutic effect on AD. However, the pharmacological actions of YLD in the treatment of AD remain unclear and lack systematic validation.

    Flow cytometry analysis showed that YLD treatment did not cause keratinocyte apoptosis, suggesting that it was not cytotoxic at commonly used doses. After specifying the concentration at which YLD had no effect on cell growth, we examined the effect of YLD on the regulation of antigen-presenting cells (APCs). T cell activation is mediated by APCs such as dendritic cells and M1-type macrophages.47 Costimulatory molecules such as CD80 and CD86 expressed by APCs activate specific immunity by interacting with CD28 on the surface of T cells.48,49 We confirmed in vitro that YLD inhibited the differentiation of M0-type macrophages into M1, suggesting that YLD may reduce T cell activation through this process.

    In vivo study, we used the vitamin D3 analog MC903 to induce AD-like skin lesions in mice. Following the continuous application of MC903 to mice, AD-like symptoms such as ear swelling, redness, and dryness were observed. Still, these symptoms were alleviated to varying degrees by oral administration of YLD. YLD was found to reduce the thickness of the stratum corneum, and alleviated epidermal edema, thereby reducing SCORAD in mice in a dose-dependent manner, with higher doses of YLD being more effective. YLD at all doses reduced mast cell infiltration in the epidermis and dermis and attenuated MC903-induced weight loss. The above results validate the therapeutic effect of YLD on AD.

    We found by western blot and immunohistochemistry that YLD reduced the elevated levels of the pro-inflammatory cytokines IL-4, IL-13, TNF-α, and TSLP, and IgE antibodies in AD-like mice, which were generally elevated in patients with moderate to severe AD. Notably, YLD may suppress pathogenic IgE production by downregulating IL-4 and IL-13, thereby inhibiting STAT6-mediated IgE class-switching in B cells.50 Additionally, YLD promoted the expression of essential proteins involved in maintaining epidermal barrier integrity, including FLG, LOR, and ELOVL, whose expression was down-regulated in MC903-treated mouse skin. As expected, we found that YLD reduced the proportion of CD4+T cells and CD8+ T cells, and downregulated the proportion of Th1/ Th2/ Th17 cells in splenic lymphocytes. These results suggest that the mechanisms of YLD in treating AD include regulating T cell differentiation and regulating type I, II and III immune responses. The ability of YLD to inhibit the differentiation of CD4+ T cells was dose-dependent. Among the three doses of YLD, YLD-H exhibited a potent inhibitory effect on immune responses, while YLD-M had a more suitable immunomodulatory effect with the number and proportion of Th cells approaching those of normal mice. YLD-M is the equivalent dose for human clinical use, and its appropriate immunosuppressive effect meets the clinical safety requirements.

    Although this work confirmed YLD efficacy in modulating Th responses and barrier repair, it remained unclear which component contributed to its anti-AD upstream signaling mechanisms. Future research will integrate existing LC-MS phytochemical data with network pharmacology approaches to identify its critical pharmacodynamic material basis and molecular targets, and clarify the synergistic mechanisms of its multicomponent system.

    Conclusions

    This study demonstrated that YLD can alleviate AD skin lesions, improve the histopathological characteristics of skin tissue, downregulate the levels of pro-inflammatory cytokines in serum and tissues, upregulate the expression of barrier genes, and inhibit T cell differentiation. These findings supported the therapeutic potential of YLD in AD by maintaining skin barrier function and suppressing adaptive immune responses, while also suggesting its potential for treating other inflammatory diseases. In summary, this study provided systematic validation of the therapeutic efficacy of YLD in AD, elucidated the mechanisms underlying its action in AD treatment, and provided a basis for applying YLD in AD.

    Data Sharing Statement

    The datasets generated and analyzed during this study are available from the primary corresponding author, Professor Zhongjian Chen, upon reasonable request.

    Ethics Approval

    The animal study was carried out in strict accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals and approved by the Ethics Committee of Shanghai Skin Disease Hospital (Tongji University, Shanghai, China) (grant number: 2021-107).

    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 work was supported by the National Natural Science Foundation of China [grant numbers 82305231] and the Science and Technology Commission of Shanghai Municipality [grant numbers 21S21900900 and 22S21902700].

    Disclosure

    The authors report no conflicts of interest in this work.

    References

    1. Stander S. Atopic Dermatitis. N Engl J Med. 2021;384(12):1136–1143. doi:10.1056/NEJMra2023911. PubMed PMID: 33761208.

    2. Tokura Y, Hayano S. Subtypes of atopic dermatitis: from phenotype to endotype. Allergol Int. 2022;71(1):14–24. doi:10.1016/j.alit.2021.07.003. Epub 20210731. PubMed PMID: 34344611.

    3. Langan SM, Irvine AD, Weidinger S. Atopic dermatitis. Lancet. 2020;396(10247):345–360. doi:10.1016/S0140-6736(20)31286-1. PubMed PMID: 32738956.

    4. Silverberg JI, Gelfand JM, Margolis DJ, et al. Patient burden and quality of life in atopic dermatitis in US adults: a population-based cross-sectional study. Ann Allergy Asthma Immunol. 2018;121(3):340–347. doi:10.1016/j.anai.2018.07.006. Epub 20180716. PubMed PMID: 30025911.

    5. Zhang J, Li G, Guo Q, et al. Allergens in Atopic Dermatitis. Clin Rev Allergy Immunol. 2025;68(1):11. doi:10.1007/s12016-025-09024-7. Epub 20250210. PubMed PMID: 39924626.

    6. Gittler JK, Krueger JG, Guttman-Yassky E. Atopic dermatitis results in intrinsic barrier and immune abnormalities: implications for contact dermatitis. J Allergy Clin Immunol. 2013;131(2):300–313. doi:10.1016/j.jaci.2012.06.048. Epub 20250210. PubMed PMID: 22939651; PubMed Central PMCID: PMCPMC4281264.

    7. Li H, Zhang Z, Zhang H, Guo Y, Yao Z. Update on the Pathogenesis and Therapy of Atopic Dermatitis. Clin Rev Allergy Immunol. 2021;61(3):324–338. doi:10.1007/s12016-021-08880-3. Epub 20210802. PubMed PMID: 34338977.

    8. Liu AW, Gillis JE, Sumpter TL, Kaplan DH. Neuroimmune interactions in atopic and allergic contact dermatitis. J Allergy Clin Immunol. 2023;151(5):1169–1177. doi:10.1016/j.jaci.2023.03.013. PubMed PMID: 37149370; PubMed Central PMCID: PMCPMC10167546.

    9. Furue M. Regulation of Filaggrin, Loricrin, and Involucrin by IL-4, IL-13, IL-17A, IL-22, AHR, and NRF2: pathogenic Implications in Atopic Dermatitis. Int J Mol Sci. 2020;21(15). doi:10.3390/ijms21155382. Epub 20200729. PubMed PMID: 32751111; PubMed Central PMCID: PMCPMC7432778

    10. van den Bogaard EH, Elias PM, Goleva E, et al. Targeting Skin Barrier Function in Atopic Dermatitis. J Allergy Clin Immunol Pract. 2023;11(5):1335–1346. doi:10.1016/j.jaip.2023.02.005. Epub 20230219. PubMed PMID: 36805053; PubMed Central PMCID: PMCPMC11346348.

    11. Wu J, Li L, Zhang T, et al. The epidermal lipid-microbiome loop and immunity: important players in atopic dermatitis. J Adv Res. 2025;68:359–374. doi:10.1016/j.jare.2024.03.001. Epub 20240307. PubMed PMID: 38460775; PubMed Central PMCID: PMCPMC11785582.

    12. Biedermann T, Skabytska Y, Kaesler S, Volz T. Regulation of T Cell Immunity in Atopic Dermatitis by Microbes: the Yin and Yang of Cutaneous Inflammation. Front Immunol. 2015;6:353. doi:10.3389/fimmu.2015.00353. Epub 20150713. PubMed PMID: 26217343; PubMed Central PMCID: PMCPMC4500098.

    13. Jha MK, Han Y, Liu Z, et al. Type 2 cytokines pleiotropically modulate sensory nerve architecture and neuroimmune interactions to mediate itch. J Allergy Clin Immunol. 2025. doi:10.1016/j.jaci.2025.05.011. Epub 20250526. PubMed PMID: 40436117.

    14. Weidinger S, Beck LA, Bieber T, Kabashima K, Irvine AD. Atopic dermatitis. Nat Rev Dis Primers. 2018;4(1):1. doi:10.1038/s41572-018-0001-z. Epub 20180621. PubMed PMID: 29930242.

    15. Dainichi T, Kitoh A, Otsuka A, et al. The epithelial immune microenvironment (EIME) in atopic dermatitis and psoriasis. Nat Immunol. 2018;19(12):1286–1298. doi:10.1038/s41590-018-0256-2. Epub 20181116. PubMed PMID: 30446754.

    16. Pena J, Zameza PA, Pixley JN, Remitz A, Feldman SR. A Comparison of Topical Corticosteroids and Topical Calcineurin Inhibitors for the Treatment of Atopic Dermatitis. J Allergy Clin Immunol Pract. 2023;11(5):1347–1359. doi:10.1016/j.jaip.2023.03.022. Epub 20230329. PubMed PMID: 36997119.

    17. Freitas E, Gooderham M, Torres T. New Topical Therapies in Development for Atopic Dermatitis. Drugs. 2022;82(8):843–853. doi:10.1007/s40265-022-01722-2. Epub 20220521. PubMed PMID: 35596877.

    18. Yang X, Kambe N, Takimoto-Ito R, Kabashima K. Advances in the pathophysiology of atopic dermatitis revealed by novel therapeutics and clinical trials. Pharmacol Ther. 2021;224:107830. doi:10.1016/j.pharmthera.2021.107830. Epub 20210302. PubMed PMID: 33662453.

    19. Chew YL, Khor MA, Xu Z, et al. Cassia alata, Coriandrum sativum, Curcuma longa and Azadirachta indica: food Ingredients as Complementary and Alternative Therapies for Atopic Dermatitis-A Comprehensive Review. Molecules. 2022;27(17). doi:10.3390/molecules27175475. Epub 20220826. PubMed PMID: 36080243; PubMed Central PMCID: PMCPMC9457827.

    20. Dong F, Tan J, Zheng Y. Chlorogenic Acid Alleviates Allergic Inflammatory Responses Through Regulating Th1/Th2 Balance in Ovalbumin-Induced Allergic Rhinitis Mice. Med Sci Monit. 2020;26:e923358. doi:10.12659/MSM.923358. Epub 20200901. PubMed PMID: 32868754; PubMed Central PMCID: PMCPMC7485287.

    21. Gendrisch F, Esser PR, Schempp CM, Wolfle U. Luteolin as a modulator of skin aging and inflammation. Biofactors. 2021;47(2):170–180. doi:10.1002/biof.1699. Epub 20201225. PubMed PMID: 33368702.

    22. Monagas M, Brendler T, Brinckmann J, et al. Understanding plant to extract ratios in botanical extracts. Front Pharmacol. 2022;13:981978. doi:10.3389/fphar.2022.981978. Epub 20220930. PubMed PMID: 36249773; PubMed Central PMCID: PMCPMC9561911.

    23. Chen S, Saeed A, Liu Q, et al. Macrophages in immunoregulation and therapeutics. Signal Transduct Target Ther. 2023;8(1):207. doi:10.1038/s41392-023-01452-1. Epub 20230522. PubMed PMID: 37211559; PubMed Central PMCID: PMCPMC10200802.

    24. Guerriero JL. Macrophages: their Untold Story in T Cell Activation and Function. Int Rev Cell Mol Biol. 2019;342:73–93. doi:10.1016/bs.ircmb.2018.07.001. Epub 20180801. PubMed PMID: 30635094.

    25. Gao J, Liang Y, Wang L. Shaping Polarization Of Tumor-Associated Macrophages In Cancer Immunotherapy. Front Immunol. 2022;13:888713. doi:10.3389/fimmu.2022.888713. Epub 20220630. PubMed PMID: 35844605; PubMed Central PMCID: PMCPMC9280632.

    26. Alam MJ, Xie L, Yap YA, Robert R. A Mouse Model of MC903-Induced Atopic Dermatitis. Curr Protoc. 2023;3(3):e695. doi:10.1002/cpz1.695. PubMed PMID: 36913546.

    27. Leyva-Castillo JM, Hener P, Jiang H, Li M. TSLP produced by keratinocytes promotes allergen sensitization through skin and thereby triggers atopic march in mice. J Invest Dermatol. 2013;133(1):154–163. doi:10.1038/jid.2012.239. Epub 20120726. PubMed PMID: 22832486.

    28. Tanaka Y, Yokoyama Y, Kambayashi T. Skin-derived TSLP stimulates skin migratory dendritic cells to promote the expansion of regulatory T cells. Eur J Immunol. 2023;53(10):e2350390. doi:10.1002/eji.202350390. Epub 20230816. PubMed PMID: 37525585; PubMed Central PMCID: PMCPMC10592182.

    29. Maddur MS, Sharma M, Hegde P, et al. Human B cells induce dendritic cell maturation and favour Th2 polarization by inducing OX-40 ligand. Nat Commun. 2014;5:4092. doi:10.1038/ncomms5092. Epub 20140609. PubMed PMID: 24910129; PubMed Central PMCID: PMCPMC4388556.

    30. Hoshino Y, Kirima K, Arichika N, et al. Long-term application of MC903 in mice prolongs the characteristic symptoms of atopic dermatitis, such as inflammation, skin barrier dysfunction, and itching. Exp Anim. 2025;74(2):276–285. doi:10.1538/expanim.24-0088. Epub 20241226. PubMed PMID: 39721714; PubMed Central PMCID: PMCPMC12044355.

    31. Wang F, Trier AM, Li F, et al. A basophil-neuronal axis promotes itch. Cell. 2021;184(2):422–440. doi:10.1016/j.cell.2020.12.033. Epub 20210114. PubMed PMID: 33450207; PubMed Central PMCID: PMCPMC7878015.

    32. Xiao C, Zhu Z, Zhang C, et al. A population of dermal Langerin(+) dendritic cells promote the inflammation in mouse model of atopic dermatitis. Front Immunol. 2022;13:981819. doi:10.3389/fimmu.2022.981819. Epub 20221003. PubMed PMID: 36304463; PubMed Central PMCID: PMCPMC9592551.

    33. Iwamoto K, Numm TJ, Koch S, Herrmann N, Leib N, Bieber T. Langerhans and inflammatory dendritic epidermal cells in atopic dermatitis are tolerized toward TLR2 activation. Allergy. 2018;73(11):2205–2213. doi:10.1111/all.13460. Epub 20181030. PubMed PMID: 29672867.

    34. De Bruyn Carlier T, Badloe FMS, Ring J, Gutermuth J, Kortekaas Krohn I. Autoreactive T cells and their role in atopic dermatitis. J Autoimmun. 2021;120:102634. doi:10.1016/j.jaut.2021.102634. Epub 20210420. PubMed PMID: 33892348.

    35. Brunner PM, Guttman-Yassky E, Leung DY. The immunology of atopic dermatitis and its reversibility with broad-spectrum and targeted therapies. J Allergy Clin Immunol. 2017;139(4S):S65–S76. doi:10.1016/j.jaci.2017.01.011. PubMed PMID: 28390479; PubMed Central PMCID: PMCPMC5405702.

    36. Gittler JK, Shemer A, Suarez-Farinas M, et al. Progressive activation of T(H)2/T(H)22 cytokines and selective epidermal proteins characterizes acute and chronic atopic dermatitis. J Allergy Clin Immunol. 2012;130(6):1344–1354. doi:10.1016/j.jaci.2012.07.012. Epub 20120827. PubMed PMID: 22951056; PubMed Central PMCID: PMCPMC3991245.

    37. Honzke S, Wallmeyer L, Ostrowski A, et al. Influence of Th2 Cytokines on the Cornified Envelope, Tight Junction Proteins, and ss-Defensins in Filaggrin-Deficient Skin Equivalents. J Invest Dermatol. 2016;136(3):631–639. doi:10.1016/j.jid.2015.11.007. Epub 20151119. PubMed PMID: 27015451.

    38. Kim BE, Leung DY, Boguniewicz M, Howell MD. Loricrin and involucrin expression is down-regulated by Th2 cytokines through STAT-6. Clin Immunol. 2008;126(3):332–337. doi:10.1016/j.clim.2007.11.006. Epub 20071231. PubMed PMID: 18166499; PubMed Central PMCID: PMCPMC2275206.

    39. Berdyshev E, Goleva E, Bronova I, et al. Lipid abnormalities in atopic skin are driven by type 2 cytokines. JCI Insight. 2018;3(4). doi:10.1172/jci.insight.98006. Epub 20180222. PubMed PMID: 29467325; PubMed Central PMCID: PMCPMC5916244.

    40. Geoghegan JA, Irvine AD, Foster TJ. Staphylococcus aureus and Atopic Dermatitis: a Complex and Evolving Relationship. Trends Microbiol. 2018;26(6):484–497. doi:10.1016/j.tim.2017.11.008. Epub 20171209. PubMed PMID: 29233606.

    41. Orciani M, Campanati A, Caffarini M, et al. T helper (Th)1, Th17 and Th2 imbalance in mesenchymal stem cells of adult patients with atopic dermatitis: at the origin of the problem. Br J Dermatol. 2017;176(6):1569–1576. doi:10.1111/bjd.15078. Epub 20170427. PubMed PMID: 27639070.

    42. Cai X, Sun X, Liu L, et al. Efficacy and safety of Chinese herbal medicine for atopic dermatitis: evidence from eight high-quality randomized placebo-controlled trials. Front Pharmacol. 2022;13:927304. doi:10.3389/fphar.2022.927304. Epub 20220927. PubMed PMID: 36238577; PubMed Central PMCID: PMCPMC9551201.

    43. Yang Y, Zhang Z, Li S, Ye X, Li X, He K. Synergy effects of herb extracts: pharmacokinetics and pharmacodynamic basis. Fitoterapia. 2014;92:133–147. doi:10.1016/j.fitote.2013.10.010. Epub 20131028. PubMed PMID: 24177191.

    44. Wang L, Pan X, Jiang L, et al. The Biological Activity Mechanism of Chlorogenic Acid and Its Applications in Food Industry: a Review. Front Nutr. 2022;9:943911. doi:10.3389/fnut.2022.943911. Epub 20220629. PubMed PMID: 35845802; PubMed Central PMCID: PMCPMC9278960.

    45. Caporali S, De Stefano A, Calabrese C, et al. Anti-Inflammatory and Active Biological Properties of the Plant-Derived Bioactive Compounds Luteolin and Luteolin 7-Glucoside. Nutrients. 2022;14(6):1155. doi:10.3390/nu14061155. Epub 20220309. PubMed PMID: 35334812; PubMed Central PMCID: PMCPMC8949538.

    46. Wang QQ, Han S, Li XX, et al. Nuezhenide Exerts Anti-Inflammatory Activity through the NF-kappaB Pathway. Curr Mol Pharmacol. 2021;14(1):101–111. doi:10.2174/1874467213666200611141337. PubMed PMID: 32525787; PubMed Central PMCID: PMCPMC8778660.

    47. Soskic B, Jeffery LE, Kennedy A, et al. CD80 on Human T Cells Is Associated With FoxP3 Expression and Supports Treg Homeostasis. Front Immunol. 2020;11:577655. doi:10.3389/fimmu.2020.577655. Epub 20210108. PubMed PMID: 33488578; PubMed Central PMCID: PMCPMC7820758.

    48. Buchbinder E, Hodi FS. Cytotoxic T lymphocyte antigen-4 and immune checkpoint blockade. J Clin Invest. 2015;125(9):3377–3383. doi:10.1172/JCI80012. Epub 20150901. PubMed PMID: 26325034; PubMed Central PMCID: PMCPMC4588295.

    49. Ahmed I, Ismail N. M1 and M2 Macrophages Polarization via mTORC1 Influences Innate Immunity and Outcome of Ehrlichia Infection. J Cell Immunol. 2020;2(3):108–115. doi:10.33696/immunology.2.029. PubMed PMID: 32719831; PubMed Central PMCID: PMCPMC7384756.

    50. Geha RS, Jabara HH, Brodeur SR. The regulation of immunoglobulin E class-switch recombination. Nat Rev Immunol. 2003;3(9):721–732. doi:10.1038/nri1181. PubMed PMID: 12949496.

    Continue Reading

  • Multiple roles of palmitic acid in cardiovascular diseases

    Multiple roles of palmitic acid in cardiovascular diseases

    Introduction

    Palmitic acid (PA) is a 16-carbon long-chain saturated fatty acid (SFA),1,2 which is widely found in animals and plants.3 It is an essential constituent acid of adipose tissue and the most abundant SFAs in the body,4 accounting for approximately 44–52% of the body’s total fat content5 and 28–32% of the total serum fatty acid (FA).6

    Cardiovascular diseases (CVD) is one of the deadliest diseases worldwide.7 CVD mainly includes coronary heart disease, cerebrovascular disease, peripheral artery disease, rheumatic heart disease, congenital heart disease, deep vein thrombosis, and pulmonary embolism.8 In addition, atrial fibrillation is very closely linked to atherosclerosis (AS) and has largely the same pathophysiological basis as other CVD: endothelial dysfunction and inflammation, coronary artery disease is an important and clinically relevant risk factor of atrial fibrillation.9 According to the World Health Organization, 17.3 million people died from CVD in 2016, accounting for 31.5% of all deaths. This number is expected to increase to 23.6 million by 2030.8 The mortality rate of CVD has exceeded that of cancer, infectious diseases, maternal diseases, and neonatal diseases.10 Hyperlipidaemia (elevated total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and reduced high-density lipoprotein cholesterol (HDL-C)), Systemic inflammation, and oxidative stress play a crucial role in the development of CVD.11–14

    Emerging evidence indicates that elevated circulating FA levels correlate with CVD incidence, and free fatty acids show diagnostic potential as early biomarkers for AS.15–20 In vivo and in vitro experiments evidence suggests potential mechanisms linking PA intake with CVD pathogenesis.21,22 Epidemiological studies also indicate that high dietary PA exposure associates with increased CVD risk across diverse populations.23,24 Elevated serum PA concentrations have been proposed to heighten atrial fibrillation risk primarily through PA’s impact on endothelial dysfunction and inflammation.25 However, critical gaps persist in current research: the analysis of PA’s biosynthetic pathways remains incomplete, with insufficient mechanistic delineation specific to individual pathologies, particularly AS, ischemic heart disease (IHD), and ischemic stroke (IS); in addition, lack of translational research frameworks connecting PA-related molecular mechanisms to therapeutic strategies; finally, while substantial evidence supports PA’s detrimental cardiovascular effects, several studies report context-dependent outcomes (Table 1). To address these gaps, this review: Systematically synthesizes PA’s anabolic pathways and pathological mechanisms in AS, IHD, and IS; Identifies novel targetable nodes for CVD prevention/treatment by pinpointing therapeutically exploitable sites within key biological pathways.

    Table 1 Palmitic Acid Associations with Traditional Cardiovascular Risk Factors

    Methods

    A systematic search was performed across four electronic databases (PubMed, Scopus, Web of Science, and Google Scholar) to comprehensively identify literature examining the association between palmitic acid and specific cardiovascular diseases, namely atherosclerosis, ischemic heart disease, and ischemic stroke. Search results were merged and deduplicated. Initial study inclusion/exclusion was determined by screening titles and abstracts. The review encompassed literature published through December 2024.

    The Anabolic Pathways of Palmitic Acid

    Endogenous Synthesis and Exogenous Uptake of Palmitic Acid

    Palmitic acid is mainly synthesized in the liver. In the de novo synthesis, glucose and glutamine produce pyruvate by glycolysis, which undergoes the tricarboxylic acid cycle in the mitochondria to produce citrate. And then the citrate is cleaved in the cytoplasm by ATP-citrate lyase (ACLY) to Acetyl-CoA and oxaloacetate. Acetyl-CoA is then carboxylated to malonyl-CoA by acetyl-CoA carboxylase (ACC) and condensed by fatty acid synthase (FASN) in a repeated reactions to generate PA.30,31 The endogenous synthesis of PA is controlled precisely under normal circumstances. However, when carbohydrate intake is excessive, the carbohydrate response element-binding protein (ChREBP) is activated, upregulation of the transcription factor sterol regulatory element binding protein-1c (SREBP-1c) and resulting in insulin production, which subsequently increases PA production32–34 (Figure 1).

    Figure 1 Endogenous synthesis and exogenous uptake of palmitic acid. In the de novo synthesis, glucose and glutamine are enzymatically catalyzed to produce citrate, which is cleaved to acetyl-CoA and oxaloacetate. Acetyl-CoA is carboxylated to malonyl-CoA, which is condensed by the repeated actions of FASN to produce PA. In the process of exogenous uptake, dietary fat is digested into free fatty acids and monoglycerides through the emulsification of bile acids in the duodenum and upper jejunum, which are then absorbed and converted into TG by intestinal epithelial cells, and then combined with apolipoproteins to form chylous particles, which enter the lymphatic system and the ultimately the bloodstream.

    Exogenous Uptake Pathway of Palmitic Acid

    Palmitic acid is found in plant oils including palm oil, peanut oil, and coconut oil, as well as in animal fats like butter and cream. Therefore, the human body can also obtain PA through exogenous dietary intake.35,36 The primary sites for digestion and absorption of fats in the human body are the duodenum and the upper jejunum. When the body consumes fats containing PA from the diet, they are emulsified by bile acids to form hydrophobic fat globules, which are then further broken down into smaller droplets. These droplets are subsequently hydrolyzed by pancreatic lipase into free fatty acids and monoacylglycerol, which are absorbed by the intestinal epithelial cells. In the endoplasmic reticulum (ER) of the epithelial cells, free fatty acids are converted into TG, which then combine with apolipoproteins. These TG, together with apolipoproteins, are transported through chylomicrons to the lymphatic system and eventually enter the bloodstream37,38 (Figure 1).

    Metabolism of Palmitic Acid

    The distribution and metabolism of PA in tissues is strictly controlled by the organism, which normally regulates the de novo synthesis pathway according to the amount of exogenous PA consumed.39,40 First, PA as a kind of FA, have the capability of providing the body with energy through the process of oxidative catabolism.41,42 PA combines with carnitine to produce an acylcarnitine molecular, and then the acylcarnitine molecular is transported across the mitochondrial membrane to the mitochondrial matrix to generate a molecule of nicotinamide adenine dinucleotide (NADH), a molecule of flavin adenine dinucleotide, reduced (FADH2), and an acetyl-CoA, which is eventually consumed as energy for the body.43 Secondly, PA is elongated or desaturated for conversion to other FA or compounds,44 which are produced in the presence of FA elongases (elongation of very long-chain fatty acids 1–7 (ELOVL1-7)) to produce longer chain FA (eg, stearic acid (SA) and arachidonic acid).45 Moreover, PA synthesized endogenously in adipocytes is converted to other FA or compounds through elongation and desaturation in preference to exogenous PA, thus ensuring that the concentration of PA in tissues is within the normal range to maintain cell membrane fluidity and insulin sensitivity.46,47 Finally, PA itself can be transformed into an important component of biofilms (phospholipids), which plays an important role in biological processes (eg, cellular proliferation, reproductive processes, and intracellular transport). PA was found to generate phosphatidylcholine and phosphatidylethanolamine (PE) by deacylation in rat hepatocyte, the final synthesis of membrane phospholipids.48 This process is regulated by membrane-binding transcription factors and can further regulate lipid synthesis.49

    In obese subjects, the activity of stearoyl coenzyme a desaturase 1 (SCD1) was increased, and SCD1 was associated with insulin sensitivity.46 However, under pathological conditions including insulin resistance and chronic nutritional imbalance, this regulatory mechanism can be disrupted, leading to excessive PA deposition in the liver and eventually to a series of CVD.50,51 Several studies have measured plasma PA concentrations in healthy subjects, indicating a range of 100~409 µM. Nevertheless, patients with diabetes, hypertriglyceridemia, and CVD have elevated plasma PA levels (Table 2).

    Table 2 Plasma Palmitic Acid Levels

    Palmitic Acid and Cardiovascular Diseases

    Palmitic Acid and Atherosclerosis

    Atherosclerosis is the basis of most CVD and causes of death, for example, coronary heart disease and stroke.58 It is characterized by the endothelial dysfunction and inflammation, form cells formation from macrophage, atherosclerotic plaque formation in the intima of arteries and apoptosis,59–61 which may result in acute cardiovascular events due to plaque rupture and thrombosis.62 Studies have demonstrated that the high concentrations of PA in blood are involved in the formation of AS through a variety of biological processes, including hyperlipidaemia,63,64 inflammation,65 vascular endothelial damage,66 form cells formation,67 and downregulation of apolipoprotein M (APOM).68

    Palmitic Acid Induces Hyperlipidemia

    There is an increased risk of CVD associated with high levels of TC, LDL-C, and lower levels of HDL-C.69,70 PA can induce AS by altering blood cholesterol levels, particularly through elevating LDL-C levels.63,64 PA inhibits the expression of low density lipoprotein (LDL) receptors and accelerates the secretion of very low-density lipoprotein (VLDL) from the liver.51 Genes related to lipid transport, adipogenesis, lipid droplet formation, and glucose and FA metabolism were found to be upregulated after incubation with PA in human hepatocytes cultured in vitro, similar effects were observed in primary cultures of human pancreatic islets.71,72 Specifically, PA promoted lipid accumulation by upregulating the CCN1/integrin α5β1 pathway.73 Lipid accumulation and apoptosis were also observed in PA-treated human kidney-2 (HK2).74 Increased dietary levels of 18:2(n-6) FA lead to lower total and LDL-C levels, while at low dietary levels of 18:2(n-6) FA, increased PA content leads to a significant increase in total and LDL-C levels.75 Meanwhile, in a controlled metabolic feeding study, PA intake promotes elevated blood cholesterol levels, consistent with previous studies.76–78 In addition, PA also induces insulin resistance, leading to impaired lipid metabolism. Prolonged exposure of cultured human, rat or mouse islets to PA leads to reduced insulin transcription, impairment of glucose-induced insulin secretion, and finally to β-cell apoptosis.79–81 PA promotes β-cell apoptosis via mTOR-mediated downregulation of protein kinase B (AKT).82 In human umbilical cord endothelial cells, PA induces insulin resistance by upregulating human regulator of G protein signaling 2 (RGS2) expression, which inhibit insulin-mediated AKT phosphorylation83,84 (Figure 2).

    Figure 2 Overview of the mechanisms by which palmitic acid promotes atherosclerosis. PA promotes the progression of by inducing hyperlipidemia, vascular endothelial cell injury, foam cell formation, downregulation of APOM, and proinflammatory effects. Its proinflammatory effect is by activating TLR2 and TLR4, enhanced LPS production and synergistic interactions with LPS, promoting FABP4 expression, amplification of proinflammatory T-cell responses, and induction of ER stress and oxidative stress (↑: increase/activation; ↓: decrease/inhibition).

    Palmitic Acid Mediates Inflammation

    Palmitic Acid Promotes the Production of Inflammatory Factors

    PA has been shown to directly increase levels of interleukin-6 (IL-6) in vivo and in vitro.85–87 PA upregulates the expression of C-reactive protein (CRP), tumor necrosis factor-α (TNF-α), and inducible nitric oxide synthase (iNOS) in vascular smooth muscle cells (VSMCs), thereby triggering an inflammatory response in cardiac fibroblasts and inducing apoptosis in VSMCs.88 PA increases the level of the cysteine-rich angiogenic inducer 61 (CYR61) in endothelial cells, thereby stimulating the production of pro-inflammatory cytokines and pro-apoptotic factors.89 PA also induces the secretion of interleukin-1β (IL-1β), monocyte chemoattractant protein-1 (MCP-1), and TNF-α by peritoneal macrophages, which activated the inflammatory process in LDLr KO mice and ultimately induced AS formation.90 In microvascular endothelial cells (EOMA lineage), palmitate stimulates the activation of NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome.88 Further studies showed that PA treatment of mouse primary macrophages induced the formation of crystals within the macrophages, which activated the NLRP3 inflammasome, resulting in lysosomal dysfunction and increased IL-1β release91 (Figure 2).

    Palmitic Acid Activates Toll-Like Receptor 4 (TLR4) to Promote Inflammation

    During the inflammatory response, toll-like receptors (TLR) serve as receptors for lipopolysaccharide (LPS).92–94 Several studies have demonstrated that PA is a TLR agonist that activates TLR4 and TLR2, and induces dimerization among TLR2 and TLR1, TLR2 and TLR6, or TLR4 and TLR6.95,96 TLR4 translocates into lipid rafts after activation and recruits its downstream adapter molecules (MyD88 and TRIF) to the rafts. After dimerizing with MyD88 or TRIF, initiates pro-inflammatory cytokine and type I interferon production.84 In addition, activated TLR4 forms a complex with myeloid differentiation protein 2 (MD2), which triggers downstream signaling. However, it is uncertain whether PA is a direct agonist of TLR4-MD2.97 During the activation of TLR4, atypical protein kinase Czeta (PKCζ) is triggered by RhoA, next PKCζ activates transforming growth factor β-activated kinase 1 (TAK1), which then participates in the activation of NF-κB,98 which results in the production of inflammatory cytokines (eg, TNF-α and IL-6).90 PA promoted the TLR4/phosphorylated-NF-κB signaling pathway by inhibiting Krüppel-like factor 4 (KLF4), upregulated Galectin-3 expression, and improved insulin resistance in macrophage99 (Figure 2).

    Palmitic Acid Activates the Proinflammatory Function of T Cells

    T cells are an instrumental component of adaptive immunity and account for 10% of all cells in atherosclerotic plaques.100,101 Using single-cell sequencing techniques, T cells were found to account for approximately 30–65% of white blood cells in atherosclerotic plaques in humans and mice.102–104 CD4+ T cells are the predominant T cell subtype in AS and exacerbate atherogenesis in immunodeficient Apoe-/- mice.105 Researchers found that both CD4+ T cells and CD8+ T cells were increased at atherosclerotic lesion sites associated with acute coronary syndrome.106 PA activates the proinflammatory function of T cells in four ways: metabolism, activation, proliferation, and polarization.107 There is evidence that PA increases insulin receptors (IR), insulin-like growth factors 1 (IGF-1), glucose transporter type 4 (GLUT4), and insulin receptor substrate 1 (IRS1) on the surface of T cells, resulting in T cell activation. PA also stimulates the proliferation of T cells and induces the polarization of T cells into proinflammatory subpopulations (Th1 cells and Th17 cells), which then induce an inflammatory response.107 The addition of 1 mM PA to peripheral blood mononuclear cells activated with anti-CD3 and anti-CD28 increased the proportion of Th1 and Th17 cells, while decreasing that of TH 2 and Treg cells. After in vitro exposure to PA, CD4+ T cells or CD8+ T cells isolated from five healthy, non-diabetic, and glucose-tolerant individuals were found to be activated in a time and concentration-dependent manner108 (Figure 2).

    Palmitic Acid Promotes Inflammation in Synergy with LPS

    A high-fat diet increases the levels of short-chain FA by altering the gut microbiome, which leads to elevated levels of LPS and enhanced activation of TLR4.109 PA also increases ceramide production through de initio synthesis and sphingolipid hydrolysis, thereby enhancing IL-6 expression and TNF-α stimulation induced by LPS.110 Researchers fed mice both LPS and a high-fat diet rich in PA, which accelerated thoracic aortic atherosclerosis.111 In human aortic endothelial cells (HAECs) and cardiac microvascular endothelial cells (MICECs), co-treatment with LPS and PA increased IL-6 expression at 36 hours111 (Figure 2).

    Palmitic Acid Promotes the Expression of Fatty Acid‑binding Protein 4 (FABP4)

    As a cytoplasmic FA carrier protein, FABP4 regulates lipid transport and responses in cells, and is associated with metabolic and inflammatory pathways.112–115 FABP4 bind a long-chain FA, including PA, SA, oleic acid (OA), linoleic acid (LA), and facilitates the translocation of FAs to specific organelles in the cell (eg mitochondria, peroxisomes, ER, and nucleus), regulates enzymatic activity, and stores excess FAs as lipid droplets.116 The FABP4 protein has a high affinity for free monounsaturated fatty acids (MUFAs) and polyunsaturated fatty acids (PUFAs) in cells under normal conditions, however, under oxidative stress conditions, the conformation of FABP4 changes, losing its affinity for most FA (except PA), and triggers an inflammatory response.116 PA increases FABP4 protein expression in macrophages via ER stress.117,118 The genetic ablation of FABP4 in macrophages showed inhibition of inflammatory signaling, reduced NF-κB pathway activation, and reduced ER stress, protecting mice from AS and dyslipidemia.119,120 In C2C12 skeletal muscle cells, overexpression of FABP4 protein decreases expression the expression of Sirtuin 3, uncoupling protein 2 (UCP2), and Peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), ultimately leading to increased ROS production in mitochondria and inflammation121–123 (Figure 2).

    Palmitic Acid Activates ER Stress

    The ER is involved in the biosynthesis of cholesterol, steroids, and other lipids. A high concentration of free fatty acids (eg PA) may disrupt lipid metabolism, which triggers stress in the ER. When PA is transformed into phospholipids and diacylglycerol (DAG), it accumulates in the ER, causing disruptions in the structure of the ER and activation of the stress sensors.124–126 The extracellular signal-regulated kinase (ERK) pathway mediates translation of CCAAT/enhancer binding protein (C/EBP) homologous proteins and genes involved in autophagy that are dependent on activating transcription factor 4 (ATF4). Inositol-requiring enzyme 1α (IRE1-α) mediates the expression of tumor necrosis factor receptor-associated factor 2 (TRAF2) and apoptosis signal-regulated kinase 1 (ASK1)/C-jun N-terminal kinase (JNK). They contribute to the ability of stress cells to maintain autophagy, which ultimately triggers ER oxidative and inflammatory signaling pathways leading to apoptosis.127–131 Phosphorylated ERK, IRE1α, and JNK activation are elevated in both adipose tissue and liver of high fat diet fed mice, which triggers ER stress, eventually leads to apoptosis.132–135 By upregulating ATF4 and C/EBP homologous protein (CHOP) expression, decreasing cytoplasmic NAD+/NADH, and reducing Sirt1 activity, PA induced ER stress in H9c2 myogblasts.136 Heart-specific sirt1 knockout mice fed a high palmitate diet were found to express higher levels of CHOP and ATF4.136 In obese individuals and type 2 diabetes mellitus (T2DM) patients, chronic exposure of β-cells to FA results in ER stress and lipotoxicity137 (Figure 2).

    Palmitic Acid Induces Oxidative Stress

    Increased reactive oxygen species (ROS) are the primary cause of palmitate-induced oxidative stress. PA enhances ROS production by promoting lipid uptake in podocytes, and the activity calcium/protein kinase Cα/NADH oxidase 4 (NOX4) pathway in endothelial cells, inhibited mitochondrial respiratory chain complex I and complex III. And the activity of adenine nucleotide carrier protein (ADP/ATP carrier protein).138–141 Normal mouse hepatocytes AML12 treated with PA. Lipid accumulation, expression of total ROS, mitochondrial ROS, NOX4, inflammasomes, and IL-1β were detected in hepatocytes after 24 h142 (Figure 2).

    Palmitic Acid Induces Vascular Endothelial Injury

    Vascular endothelial injury is an important pathological process in the process of AS. Endothelial dysfunction, characterized by impaired vasodilation, inflammation, and thrombosis, triggers future CVD.143 Reduced endothelial progenitor cells are independent predictors of CVD morbidity and mortality.144 Lipotoxicity of PA decreases immune surveillance protein DDX58/Rig-1 expression and activity, leading to impaired autophagy and apoptosis;145 apoptosis in vascular endothelial cells induces endothelial injury and promotes AS progression.146,147 A member of the angiopoietin-like protein family involved in lipid metabolism promotes endothelial cell proliferation and inhibits PA-induced endothelial cell injury by increasing autophagy, which may inhibit AS.66 Also, activation of the interferon regulator 3 (IRF3) pathway causes endothelial inflammation.148 Nitric oxide (NO) from enzymatic NO synthases (NOS) system importantly contributes to vascular homeostasis, in addition to the classical NOS system, NO can also be generated via the nitrate-nitrite-NO pathway.149 The addition of PA to HAECs resulted in decreased cell viability, reduced intracellular NO production, increased migratory capacity of HAECs, and cellular oxidative stress, ultimately leading to endothelial-to-mesenchymal transition.150 In endothelial cells, PA upregulated the expression of phosphorylated p38, JNK, and caspase-3, thereby increasing endothelial apoptosis dose- and time-dependently.151,152 Patients with coronary artery disease showed significantly higher levels of phosphorylation of p38 and mitogen-activated protein kinase (MAPK) in endothelial progenitor cells than healthy individuals.153 Inhibition or knockout of p38 and MAPK significantly increases the number of circulating endothelial progenitor cells154 (Figure 2).

    Palmitic Acid Promotes Foam Cells Formation

    Form cells is one of the major causes of AS, which is due to the accumulation of oxidized LDL (oxLDL) in the arterial intima. Macrophages absorb accumulated oxLDL and form cells. The presence of high levels of PA in the blood enhances the ability of macrophages to take up oxLDL and produce more form cells. OxLDL is a dysfunctional lipid metabolite that is a major promoter of the prothrombotic state in both animal models and human patients.67,155 In macrophages, PA enhances lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) expression, promotes oxLDL uptake, a process mediated mainly through the ROS-p38 MAPK pathway.156 5-hydroxytryptamine (5-HT) takes part in platelet aggregation, vasoconstriction, proliferation of VSMCs, ER function, and macrophage foam cell formation, play a key role in the development of AS.157,158 In vitro experiments, exposure of macrophages and human umbilical vein endothelial Cells (HUVECs) to oxLDL or PA demonstrated that activation of 5-HT2A receptor regulates TG synthesis and oxLDL uptake by activating PKCε, resulting in the formation of lipid droplets and even foam cells.159 PA increases CD146 expression in macrophages, promoting foam cell formation and disrupting migration-related signaling,160 by activating JNK signaling and inhibiting STAT3 activation, CD146 (Gp130) promotes proinflammatory polarization of M1-like adipose tissue macrophages (ATMs)161 (Figure 2).

    Palmitic Acid Induces Apolipoprotein M Downregulation

    Palmitic acid can downregulate the expression of human APOM, promote the accumulation of cholesterol in the blood and induce the development of AS. APOM facilitates HDL metabolism and stabilization, which can reduce blood cholesterol levels, with anti-AS, anti-inflammatory and antioxidant effects.162 Generally, it is found in hepatocytes and renal tubular epithelial cells, and is weakly expressed in colorectal tissues.163,164 APOM has been shown to be a possible HDL-carrying receptor for sphingosine 1-phosphate, which enhances HDL-mediated antioxidant effects.165,166 APOM plays a role in the formation of preb-HDL,167,168 PA significantly inhibited APOM gene expression in HepG2 cells, and the peroxisome proliferator-activated receptor β/δ (PPAR β/δ) antagonist GSK3787 completely reversed PA-induced downregulation of APOM expression, indicating that PA-induced downregulation of APOM expression is mediated through the PPAR β/δ pathway.68 A key regulator of lipid metabolism, peroxisome proliferator-activated receptor (PPAR), is expressed in platelets. This receptor upregulates the transcription of lipid metabolizing enzymes, including carnitine palmitoyl coenzyme A transferase-I (GPT-I) and acyl-CoA oxidase, both of which are important to thrombosis and hemostasis169,170 (Figure 2).

    Palmitic Acid and Ischemic Heart Disease

    Ischemic heart disease is heart disease caused by narrowing/occlusion of the coronary arteries or by ischemia, hypoxia, or necrosis of the heart muscle due to spasm of the coronary arteries. Approximately 40–80% of the heart’s energy comes from FA, several cohort studies have revealed, compared with healthy young subjects, patients with chronic heart failure, myocardial ischemia, T2DM, and obese individuals elevated levels of free fatty acids (include PA) in the blood.171–177 Additionally, there are studies that indicate that PA levels in adipose tissue are related to IHD incidence. Insull et al found that SA (18:0), lauric acid (12:0), palmitoleic acid (16:1), myristic acid (14:0), and LA (18:2) acids were associated with coronary artery disease, and PA (16:0) content in adipose tissue was associated with plasma cholesterol levels.178 A study by Lee et al compared the FA composition of adipose tissue in two races with different prevalences of coronary heart disease and found significant differences in PA, palmitoleic, and OA (18:1).179 Thus, high concentrations of PA, both circulation and adipose tissue, are associated with the incidence of IHD. There was a significant increase in FA uptake and FA oxidation in the heart when the supply of free FA was increased, according to Lopaschuk GD.180 Replacing saturated FA (FA and SA) with plant-based proteins may reduce the risk of myocardial infarction.181

    Palmitic Acid Induces Apoptosis in Cardiomyocytes

    Palmitic acid induces cardiomyocyte apoptosis by promoting autophagy. Studies have shown that after treating rat cardiomyocytes with PA (0.25 and 0.5 mM) for 18 hours, the number of apoptotic cells and biochemical markers (caspase activation, DNA fragmentation), significantly increased.182 In cardiomyocytes, PA induces apoptosis by promoting the generation of ceramide and activating the mitochondrial apoptosis pathway, leading to the myofibril disintegration.183 In a cohort study involving 4249 participants, the correlation between plasma ceramide (Cer) and sphingomyelin (SM) levels and the risk of sudden heart failure was investigated. The results showed that high levels of PA were associated with a higher risk of heart failure during a median follow-up of 9.4 years.184 Ischemic events are believed to increase the flow of free fatty acids to cardiomyocytes, thereby increasing oxidative stress and causing cardiomyocyte damage.185–188 When the heart is exposed to excessive energy (eg, glucose, free fatty acids, and TG) and growth factors (eg, insulin and leptin) over a long period, it accelerates the development of cardiomyopathy, leading to cardiac hypertrophy and failure. These processes are driven by oxidative stress induced by glucolipotoxicity and become the main drivers of cell apoptosis189 (Figure 3).

    Figure 3 Overview of the mechanism by which palmitic acid promotes ischemic heart disease. PA accelerates progression of induces oxidative stress and autophagic dysregulation, and further triggers cardiomyocyte apoptosis. Additionally, PA promotes cardiomyocyte ferroptosis by reducing the protein expression of Heat Shock Factor 1 and Glutathione Peroxidase 4. These mechanisms collectively drive pathogenesis the onset and development of IHD (↑: increase/activation; ↓: decrease/inhibition).

    Palmitic Acid Promotes Cardiomyocyte Ferroptosis

    Ferroptosis is an iron-dependent form of programmed cell death.190 The primary mechanism of ferroptosis is the induction of cell death through the action of divalent iron or lipoxygenases. Additionally, the expression of the antioxidant systems glutathione and glutathione peroxidase 4 (GPX4) is also involved in the process.191 A large body of evidence has shown that ferroptosis is associated with CVD, particularly with ischemia-reperfusion injury and myocardial infarction.192 Using different ferroptosis inhibitors significantly reduced PA-induced death in both H9c2s and primary neonatal rat cardiomyocytes. Specifically, PA promotes ferroptosis by reducing the protein expression of heat shock factor 1 (HSF1) and GPX4, while overexpression of HSF1 and GPX4 effectively prevents PA-induced ferroptosis4 (Figure 3).

    Palmitic Acid and Ischemic Stroke

    Ischemic stroke has become a major cause of global disease burden due to its high incidence, prevalence, mortality, and disability rates.193 In 2013, an estimated 6.9 million new IS cases occurred globally, with only 18.25 million surviving in good health, 3.32 million deaths, and 65.54 million disabilities.194 Plasma levels of docosahexaenoic acid, LA, arachidonic acid, and PA were measured by gas chromatography in 943 participants from the Framingham Heart Study and 1406 participants from three cities of the Bordeaux Study. The results showed that PA is a risk factor for stroke.195 In a study conducted at the Minneapolis Community Atherosclerosis Risk Center, 3870 white men and women aged 45–64 years (1987–1989) were assessed for plasma cholesterol esters and phospholipid FA, revealing a significant positive correlation between plasma SFAs (particularly PA) and IS.196

    Palmitic Acid Promotes Neuroinflammation

    Palmitic acid can induce chronic inflammation in both peripheral tissues and the central nervous system, for example, hypothalamic neurons.197–200 In in vitro experiments, PA was found to induce dysfunction in human adipose tissue and soft meningeal artery endothelial cells.201 Researchers found that when Medin (a common amyloid protein) was combined with PA, there was upregulation of IL-6, IL-8, and PAI-1 gene expression in HUVECs, suggesting combined proinflammatory and prothrombotic effects in IS pathogenesis.201,202 Mechanistically, PA promotes TLR4 recruitment to lipid rafts in SH-SY5Y neuroblastoma cells, facilitating TLR4/MYD88/TIRAP complex formation a process potentiated by heme-dependent TLR4 activation.94 PA promoted the upregulation of IL-6 and TNF-α in primary hypothalamic cultures from rats.203 Further studies confirmed that mice fed a high PA diet showed increased hypothalamic cytokine levels, proinflammatory signaling, neuronal death, and impaired leptin and insulin signaling.198,204 Direct intraventricular injection of PA also led to hypothalamic inflammation and insulin resistance.203 PA induces the expression of proinflammatory cytokines in cultured hypothalamic neurons (N42) by increasing ceramide accumulation and lipotoxicity.92 Additionally, PA interacts with LPS to activate microglial cells, upregulating the expression of proinflammatory cytokines via MAPK, NF-κB, and AP-1 signaling pathways, inducing neuroinflammation in HMC3 cells205 (Figure 4).

    Figure 4 Overview of the mechanism by which palmitic acid promotes ischemic stroke. PA exacerbates IS through multi-target mechanisms: (1) Atherogenesis: Accelerates plaque formation via ceramide overproduction and proinflammatory cytokine induction. (2) Neuroinflammation: Triggers CNS inflammatory cascades through microglial TLR/NLRP3 activation and astrocytic metabolic reprogramming. (3) Neuronal Apoptosis: Induces ER stress-autophagy axis dysregulation in neurons. (4) Glial Activation: Directly stimulates microglial inflammatory signaling and astrocytic lipotoxicity. These interconnected pathways collectively drive neurovascular unit dysfunction, culminating in IS progression (↑: increase/activation).

    Palmitic Acid Promotes Apoptosis of Neuronal

    The lipotoxicity of PA triggers ER stress and autophagic impairment, leading to an increase in apoptosis and the regulation of neuronal plasticity. High concentrations of PA have been shown to induce ER stress in SH-SY5Y cells and mouse brain cells.206 In SH-SY5Y cells and human glioblastoma cells, PA-induced neurotoxicity and glial cell toxicity, as well as increased oxidative stress in neurons and astrocytes, further promoted cell apoptosis.207 Mechanistic studies reveal that PA upregulates fatty acid transport protein 1 (FATP1) expression, which enhances prefrontal cortical autophagy dysregulation and ER stress while downregulating neuroplasticity markers including synaptophysin (SYN), brain-derived neurotrophic factor (BDNF), and acetylcholine receptors (AChRs).208 High-fat diets containing PA activate the MST1/JNK/Caspase-3 signaling pathway in hippocampal HT22 cells, leading to neuronal apoptosis.209,210 In in vitro experiments, PA significantly increased the autophagic flux in hypothalamic neurons. After PA exposure, the autophagic flux in hypothalamic neurons was suppressed, leading to impaired neuronal autophagy. This autophagic dysfunction was accompanied by changes in lysosomal dynamics, increased Rab7 GTPase activity, ERK phosphorylation, elevated expression of NADPH oxidase 4, and higher levels of inflammation, oxidative stress, and apoptosis in DRG neurons211 (Figure 4).

    Palmitic Acid Activates Glial Cells

    Glial cells, primarily composed of microglia and astrocytes, PA can activate glial cells. Microglia are the principal FA sensors in the hypothalamus related to neuronal stress and inflammation and are key mediators of the inflammatory response after stroke and brain injury.212 PA promotes inflammation by activating TLR receptors distributed in microglia, and also activates NLRP3 inflammasome by increasing TLR4/MyD88/NF-κB p65 signaling, Long-term activation of hypothalamic microglia inhibits neurogenesis in the medial basal hypothalamus (MBH), and the occurrence of IS further activates microglia and exacerbates disease progression.213,214 Astrocytes are the primary cells responsible for FA oxidation in the brain and play an important role in chronic inflammatory responses associated with obesity and the development of secondary metabolic disorders.215 Although the brain’s energy is primarily provided by glucose PA accumulation in astrocytes activates mitochondrial β-oxidation pathways, generating ATP while inducing proinflammatory activation216 (Figure 4).

    Conclusion and Future Directions

    Cardiovascular impact of dietary fatty acids exhibits fundamental dichotomy: Saturated fatty acids, particularly PA, promote cardiovascular pathogenesis through pro-inflammatory, dyslipidemic, and endothelial dysfunction pathways. SA as one of the metabolic products of PA, that exhibits neutral metabolic effects. While monounsaturated (eg, oleic acid) and polyunsaturated fatty acids confer cardioprotection. As the most abundant endogenous and dietary SFA, PA serves as a pathophysiological pivot in atherosclerosis development and cerebrovascular complications. Translation of these mechanistic insights into balanced nutritional interventions represents an actionable strategy for global CVD burden reduction.

    However, current limitations must be addressed: current evidence exhibits heterogeneity in PA exposure quantification across studies; different organizations, races, and diseases should adopt specific quantitative standards, rather than simply using the same standard for measurement; moreover, most interventional data derive from preclinical models requiring human validation.

    To advance this field, future research should prioritize: establish specific quantitative standards for different organizations, races and diseases; elucidate tissue-specific signaling mechanisms (eg, endothelial vs glial PA sensing); develop targeted therapies disrupting PA-induced inflammatory cascades (eg, RGS2 inhibitors); conduct randomized trials testing precision and personalized nutrition approaches for high-risk populations; establish clinical biomarkers quantifying PA’s pathogenic contributions.

    Declaration of Generative AI and AI-Assisted Technologies in the Writing Process

    During the preparation of this work, the authors used [deep seek] in order to [improve language and readability]. After using this tool, the authors reviewed and edited the content as needed and takes full responsibility for the content of the publication.

    Abbreviations

    PA, palmitic acid; SFAs, saturated fatty acids; CVD, cardiovascular diseases; AS, atherosclerosis; IHD, ischemic heart disease; IS, ischemic stroke; FA, fatty acid; TC, total cholesterol; TG, triglyceride; LDL-C, low-density lipoprotein cholesterol; HDL-C, high-density lipoprotein cholesterol; ACLY, ATP-citrate lyase; ACC, acetyl-CoA carboxylase; FASN, fatty acid synthase; ChREBP, carbohydrate response element-binding protein; SREBP-1c, sterol regulatory element binding protein-1c; ER, endoplasmic reticulum; NADH, nicotinamide adenine dinucleotide; FADH2, flavin adenine dinucleotide, reduced; ELOVL1-7, elongation of very long-chain fatty acids 1-7; SA, stearic acid; PE, phosphatidylethanolamine; SCD1, stearoyl coenzyme a desaturase 1; APOM, apolipoprotein M; LDL, low density lipoprotein; VLDL, very low-density lipoprotein; HK2, human kidney-2; AKT, protein kinase B; RGS2, human regulator of G protein signaling 2; IL-6,interleukin-6; CRP, C-reactive protein; TNF-α, tumor necrosis factor-α; Inos, nitric oxide synthase; VSMCs, vascular smooth muscle cells; CYR61, cysteine-rich angiogenic inducer 61; IL-1β, interleukin-1β; MCP-1, monocyte chemoattractant protein-1; NLRP3, NACHT, LRR and PYD domains-containing protein 3; TLR4, Toll-like receptor 4; TLR, toll-like receptors; LPS, lipopolysaccharide; MD2, myeloid differentiation protein 2; pkcζ, atypical protein kinase Czeta; TAK1, transforming growth factor β-activated kinase 1; KLF4, Krüppel-like factor 4; IR, insulin receptors; IGF-1, insulin-like growth factors 1; GLUT4, glucose transporter type 4; IRS1, insulin receptor substrate 1; HAECs, human aortic endothelial cells; MICECs, cardiac microvascular endothelial cells; FABP4, fatty acid‑binding protein 4; OA, oleic acid; LA, linoleic acid; MUFAs, monounsaturated fatty acids; PUFAs, polyunsaturated fatty acids; PGC-1α, Peroxisome proliferator-activated receptor gamma coactivator 1α; UCP2, uncoupling protein 2; DAG, diacylglycerol; ERK, extracellular signal-regulated kinase; C/EBP, CCAAT/enhancer binding protein; ATF4, activating transcription factor 4; IRE1-α, inositol-requiring enzyme 1α; TRAF2, tumor necrosis factor receptor-associated factor 2; ASK1, apoptosis signal-regulated kinase 1; JNK, C-jun N-terminal kinase; CHOP, C/EBP homologous protein; T2DM, type 2 diabetes mellitus; ROS, reactive oxygen species; NOX4, calcium/protein kinase Cα/NADH oxidase 4; IRF3, interferon regulator 3; NO, nitric oxide; MAPK, mitogen-activated protein kinase; oxLDL, oxidized LDL; LOX-1, lectin-like oxidized low-density lipoprotein receptor-1; 5-HT, 5-hydroxytryptamine; HUVECs, human umbilical vein endothelial Cells; ATMs, M1-like adipose tissue macrophages; PPAR, eroxisome proliferator-activated receptor; GPT-I, carnitine palmitoyl coenzyme A transferase-I; Cer, ceramide; SM, sphingomyelin; GPX4, glutathione peroxidase 4; HSF1, heat shock factor 1; FATP1, fatty acid transport protein 1; SYN, synaptophysin; BDNF, brain-derived neurotrophic factor, AChRs, acetylcholine receptors; MBH, medial basal hypothalamus.

    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 work was supported by the National Natural Science Foundation of China (32460138); Priority Union Foundation of Yunnan Provincial Science and Technology Department and Kunming Medical University (202101AC070461), and Basic Research Program of Yunnan Province Science and Technology Department (202301AT070083).

    Disclosure

    The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

    References

    1. Ávalos Y, Hernández-Cáceres MP, Lagos P, et al. Palmitic acid control of ciliogenesis modulates insulin signaling in hypothalamic neurons through an autophagy-dependent mechanism. Cell Death Dis. 2022;13(7):1–13. doi:10.1038/s41419-022-05109-9

    2. Bier DM. Saturated fats and cardiovascular disease: interpretations not as simple as they once were. Crit Rev Food Sci Nutr. 2016;56(12):1943–1946. doi:10.1080/10408398.2014.998332

    3. Wu KM, Hsu YM, Ying MC, et al. High-density lipoprotein ameliorates palmitic acid-induced lipotoxicity and oxidative dysfunction in H9c2 cardiomyoblast cells via ROS suppression. Nutr Metab. 2019;16: 36. doi:10.1186/s12986-019-0356-5

    4. Wang N, Ma H, Li J, et al. HSF1 functions as a key defender against palmitic acid-induced ferroptosis in cardiomyocytes. J Mol Cell Cardiol. 2021;150:65–76. doi:10.1016/j.yjmcc.2020.10.010

    5. Mancini A, Imperlini E, Nigro E, et al. Biological and nutritional properties of palm oil and palmitic acid: effects on health. Molecules. 2015;20(9):17339–17361. doi:10.3390/molecules200917339

    6. Klein S, Wolfe RR. Carbohydrate restriction regulates the adaptive response to fasting. Am J Physiol. 1992;262(5 Pt 1):E631–636. doi:10.1152/ajpendo.1992.262.5.E631

    7. Pilz S, März W. Free fatty acids as a cardiovascular risk factor. Clin Chem Lab Med. 2008;46(4):429–434. doi:10.1515/cclm.2008.118

    8. Benjamin EJ, Muntner P, Alonso A, et al. Heart disease and stroke statistics-2019 update: a report from the American Heart Association. Circulation. 2019;139(10):e56–e528. doi:10.1161/cir.0000000000000659

    9. Batta A, Hatwal J, Sharma YP. Assessment of coronary artery disease in non-valvular atrial fibrillation: is this light at the end of the tunnel? Vasc Health Risk Manag. 2024;20:493–499. doi:10.2147/VHRM.S484638

    10. Townsend N, Wilson L, Bhatnagar P, et al. Cardiovascular disease in Europe: epidemiological update 2016. Eur Heart J. 2016;37(42):3232–3245. doi:10.1093/eurheartj/ehw334

    11. Shramko VS, Polonskaya YV, Kashtanova EV, et al. The short overview on the relevance of fatty acids for human cardiovascular disorders. Biomolecules. 2020;10(8):1127. doi:10.3390/biom10081127

    12. Gordon T, Kannel WB. Multiple risk functions for predicting coronary heart disease: the concept, accuracy, and application. Am Heart J. 1982;103(6):1031–1039. doi:10.1016/0002-8703(82)90567-1

    13. Kannel WB, Mcgee DL. Diabetes and glucose tolerance as risk factors for cardiovascular disease: the Framingham study. Diabetes Care. 1979;2(2):120–126. doi:10.2337/diacare.2.2.120

    14. Gordon T, Castelli WP, Hjortland MC, et al. Diabetes, blood lipids, and the role of obesity in coronary heart disease risk for women. The Framingham study. Ann Int Med. 1977;87(4):393–397. doi:10.7326/0003-4819-87-4-393

    15. Skeaff CM, Miller J. Dietary fat and coronary heart disease: summary of evidence from prospective cohort and randomised controlled trials. Ann Nutr Metab. 2009;55(1–3):173–201. doi:10.1159/000229002

    16. Chen X, Liu L, Palacios G, et al. Plasma metabolomics reveals biomarkers of the atherosclerosis. J Sep Sci. 2010;33(17–18):2776–2783. doi:10.1002/jssc.201000395

    17. Oh PC, Koh KK, Sakuma I, et al. Omega-3 fatty acid therapy dose-dependently and significantly decreased triglycerides and improved flow-mediated dilation, however, did not significantly improve insulin sensitivity in patients with hypertriglyceridemia. Int J Cardiol. 2014;176(3):696–702. doi:10.1016/j.ijcard.2014.07.075

    18. Hamazaki K, Iso H, Eshak ES, et al. Plasma levels of n-3 fatty acids and risk of coronary heart disease among Japanese: the Japan Public Health Center-based (JPHC) study. Atherosclerosis. 2018;272:226–232. doi:10.1016/j.atherosclerosis.2017.12.004

    19. Siasos G, Tousoulis D, Oikonomou E, et al. Effects of omega-3 fatty acids on endothelial function, arterial wall properties, inflammatory and fibrinolytic status in smokers: a cross over study. Int J Cardiol. 2013;166(2):340–346. doi:10.1016/j.ijcard.2011.10.081

    20. Bäck M. Omega-3 fatty acids in atherosclerosis and coronary artery disease. Future Sci OA. 2017;3(4):Fso236. doi:10.4155/fsoa-2017-0067

    21. Harvey KA, Walker CL, Pavlina TM, et al. Long-chain saturated fatty acids induce pro-inflammatory responses and impact endothelial cell growth. Clin Nutr. 2010;29(4):492–500. doi:10.1016/j.clnu.2009.10.008

    22. Shen H, Eguchi K, Kono N, et al. Saturated fatty acid palmitate aggravates neointima formation by promoting smooth muscle phenotypic modulation. Arterioscler Thromb Vasc Biol. 2013;33(11):2596–2607. doi:10.1161/atvbaha.113.302099

    23. Chen Y, Cao Y, Li L, et al. The association between circulating palmitic acid levels and risk of premature coronary artery disease in Chinese patients: a case-control study. BMC Cardiovasc Disord. 2025;25(1):412. doi:10.1186/s12872-025-04873-8

    24. Jensen PN, Fretts AM, Hoofnagle AN, et al. Plasma ceramides and sphingomyelins in relation to atrial fibrillation risk: the cardiovascular health study. J Am Heart Assoc. 2020;9(4):e012853. doi:10.1161/JAHA.119.012853

    25. Annevelink CE, Sapp PA, Petersen KS, et al. Diet-derived and diet-related endogenously produced palmitic acid: effects on metabolic regulation and cardiovascular disease risk. J Clin Lipidol. 2023;17(5):577–586. doi:10.1016/j.jacl.2023.07.005

    26. Gonçalinho GHF, Sampaio GR, Soares-Freitas RAM, Damasceno NRT. Stearic acid, but not palmitic acid, is associated with inflammatory and endothelial dysfunction biomarkers in individuals at cardiovascular risk. Arq Bras Cardiol. 2023;120(8):e20220598. doi:10.36660/abc.20220598

    27. Caspar-Bauguil S, Kolditz CI, Lefort C, et al. Fatty acids from fat cell lipolysis do not activate an inflammatory response but are stored as triacylglycerols in adipose tissue macrophages. Diabetologia. 2015;58(11):2627–2636. doi:10.1007/s00125-015-3719-0

    28. Ng TK, Hassan K, Lim JB, Lye MS, Ishak R. Nonhypercholesterolemic effects of a palm-oil diet in Malaysian volunteers. Am J Clin Nutr. 1991;53(4 Suppl):1015S–1020S. doi:10.1093/ajcn/53.4.1015S

    29. Marzuki A, Arshad F, Razak TA, Jaarin K. Influence of dietary fat on plasma lipid profiles of Malaysian adolescents. Am J Clin Nutr. 1991;53(4 Suppl):1010S–1014S. doi:10.1093/ajcn/53.4.1010S

    30. Brownsey RW, Boone AN, Elliott JE, et al. Regulation of acetyl-CoA carboxylase. Biochem Soc Trans. 2006;34(2):223–227. doi:10.1042/BST0340223

    31. Zaidi N, Swinnen JV, Smans K. ATP-citrate lyase: a key player in cancer metabolism. Cancer Res. 2012;72(15):3709–3714. doi:10.1158/0008-5472.Can-11-4112

    32. Uyeda K, Repa JJ. Carbohydrate response element binding protein, ChREBP, a transcription factor coupling hepatic glucose utilization and lipid synthesis. Cell Metab. 2006;4(2):107–110. doi:10.1016/j.cmet.2006.06.008

    33. Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest. 2002;109(9):1125–1131. doi:10.1172/JCI0215593

    34. Cohen JC, Horton JD, Hobbs HH. Human fatty liver disease: old questions and new insights. Science. 2011;332(6037):1519–1523. doi:10.1126/science.1204265

    35. Sensoy I. A review on the food digestion in the digestive tract and the used in vitro models. Curr Res Food Sci. 2021;4:308–319. doi:10.1016/j.crfs.2021.04.004

    36. Lema I, Araújo JR, Rolhion N, et al. Jejunum: the understudied meeting place of dietary lipids and the microbiota. Biochimie. 2020;178:124–136. doi:10.1016/j.biochi.2020.09.007

    37. Lindquist S, Hernell O. Lipid digestion and absorption in early life: an update. Curr Opin Clin Nutr Metab Care. 2010;13(3):314–320. doi:10.1097/MCO.0b013e328337bbf0

    38. Chadaideh KS, Carmody RN. Host-microbial interactions in the metabolism of different dietary fats. Cell Metab. 2021;33(5):857–872. doi:10.1016/j.cmet.2021.04.011

    39. Hulver MW, Berggren JR, Carper MJ, et al. Elevated stearoyl-CoA desaturase-1 expression in skeletal muscle contributes to abnormal fatty acid partitioning in obese humans. Cell Metab. 2005;2(4):251–261. doi:10.1016/j.cmet.2005.09.002

    40. Peter A, Weigert C, Staiger H, et al. Individual stearoyl-coa desaturase 1 expression modulates endoplasmic reticulum stress and inflammation in human myotubes and is associated with skeletal muscle lipid storage and insulin sensitivity in vivo. Diabetes. 2009;58(8):1757–1765. doi:10.2337/db09-0188

    41. Fritzen AM, Lundsgaard A-M, Kiens B. Tuning fatty acid oxidation in skeletal muscle with dietary fat and exercise. Nat Rev Endocrinol. 2020;16(12):683–696. doi:10.1038/s41574-020-0405-1

    42. Smith CD, Lin C-T, Mcmillin SL, et al. Genetically increasing flux through β-oxidation in skeletal muscle increases mitochondrial reductive stress and glucose intolerance. Am J Physiol Endocrinol Metab. 2021;320(5):E938–E950. doi:10.1152/ajpendo.00010.2021

    43. Koga Y. Thermal adaptation of the archaeal and bacterial lipid membranes. Archaea. 2012;2012:789652. doi:10.1155/2012/789652

    44. Chen X, Li J, Kang R, et al. Ferroptosis: machinery and regulation. Autophagy. 2021;17(9):2054–2081. doi:10.1080/15548627.2020.1810918

    45. Igal RA. Stearoyl-CoA desaturase-1: a novel key player in the mechanisms of cell proliferation, programmed cell death and transformation to cancer. Carcinogenesis. 2010;31(9):1509–1515. doi:10.1093/carcin/bgq131

    46. Collins JM, Neville MJ, Hoppa MB, et al. De novo lipogenesis and stearoyl-CoA desaturase are coordinately regulated in the human adipocyte and protect against palmitate-induced cell injury. J Biol Chem. 2010;285(9):6044–6052. doi:10.1074/jbc.M109.053280

    47. Dambrova M, Zuurbier CJ, Borutaite V, et al. Energy substrate metabolism and mitochondrial oxidative stress in cardiac ischemia/reperfusion injury. Free Radic Biol Med. 2021;165:24–37. doi:10.1016/j.freeradbiomed.2021.01.036

    48. Schmid PC, Deli E, Schmid HH. Generation and remodeling of phospholipid molecular species in rat hepatocytes. Arch Biochem Biophys. 1995;319(1):168–176. doi:10.1006/abbi.1995.1279

    49. Dobrosotskaya IY, Seegmiller AC, Brown MS, et al. Regulation of SREBP processing and membrane lipid production by phospholipids in Drosophila. Science. 2002;296(5569):879–883. doi:10.1126/science.1071124

    50. Donnelly KL, Smith CI, Schwarzenberg SJ, et al. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest. 2005;115(5):1343–1351. doi:10.1172/jci23621

    51. Spady DK, Woollett LA, Dietschy JM. Regulation of plasma LDL-cholesterol levels by dietary cholesterol and fatty acids. Annu Rev Nutr. 1993;13:355–381. doi:10.1146/annurev.nu.13.070193.002035

    52. Psychogios N, Hau DD, Peng J, et al. The human serum metabolome. PLoS One. 2011;6(2):e16957. doi:10.1371/journal.pone.0016957

    53. Lopes SM, Trimbo SL, Mascioli EA, et al. Human plasma fatty acid variations and how they are related to dietary intake. Am J Clin Nutr. 1991;53(3):628–637. doi:10.1093/ajcn/53.3.628

    54. Hoffmann GF, Meier-Augenstein W, Stöckler S, et al. Physiology and pathophysiology of organic acids in cerebrospinal fluid. J Inherit Metab Dis. 1993;16(4):648–669. doi:10.1007/bf00711898

    55. Trombetta A, Togliatto G, Rosso A, et al. Increase of palmitic acid concentration impairs endothelial progenitor cell and bone marrow-derived progenitor cell bioavailability: role of the STAT5/PPARγ transcriptional complex. Diabetes. 2013;62(4):1245–1257. doi:10.2337/db12-0646

    56. Paillard F, Catheline D, Duff FL, et al. Plasma palmitoleic acid, a product of stearoyl-coA desaturase activity, is an independent marker of triglyceridemia and abdominal adiposity. Nutr Metab Cardiovasc Dis. 2008;18(6):436–440. doi:10.1016/j.numecd.2007.02.017

    57. Clore JN, Allred J, White D, et al. The role of plasma fatty acid composition in endogenous glucose production in patients with type 2 diabetes mellitus. Metabolism. 2002;51(11):1471–1477. doi:10.1053/meta.2002.35202

    58. Zhang J, Wu G, Dai H. The matricellular protein CCN1 regulates TNF-α induced vascular endothelial cell apoptosis. Cell Biology International. 2016;40(1):1–6. doi:10.1002/cbin.10469

    59. Mozaffarian D. Trans fatty acids – effects on systemic inflammation and endothelial function. Atheroscler Suppl. 2006;7(2):29–32. doi:10.1016/j.atherosclerosissup.2006.04.007

    60. Libby P. The changing landscape of atherosclerosis. Nature. 2021;592(7855):524–533. doi:10.1038/s41586-021-03392-8

    61. Galkina E, Ley K. Vascular adhesion molecules in atherosclerosis. Arterioscler Thromb Vasc Biol. 2007;27(11):2292–2301. doi:10.1161/atvbaha.107.149179

    62. Wu D, Liu J, Pang X, et al. Palmitic acid exerts pro-inflammatory effects on vascular smooth muscle cells by inducing the expression of C-reactive protein, inducible nitric oxide synthase and tumor necrosis factor-α. Int J Mol Med. 2014;34(6):1706–1712. doi:10.3892/ijmm.2014.1942

    63. Sacks FM, Lichtenstein AH, Wu JHY, et al. Dietary fats and cardiovascular disease: a presidential advisory from the American Heart Association. Circulation. 2017;136(3):e1–e23. doi:10.1161/CIR.0000000000000510

    64. Astrup A, Magkos F, Bier DM, et al. Saturated fats and health: a reassessment and proposal for food-based recommendations: JACC state-of-the-art review. J Am Coll Cardiol. 2020;76(7):844–857. doi:10.1016/j.jacc.2020.05.077

    65. Domínguez-López I, Arancibia-Riveros C, Casas R, et al. Changes in plasma total saturated fatty acids and palmitic acid are related to pro-inflammatory molecule IL-6 concentrations after nutritional intervention for one year. Biomed Pharmacother. 2022;150:113028. doi:10.1016/j.biopha.2022.113028

    66. Zhan W, Tian W, Zhang W, et al. ANGPTL4 attenuates palmitic acid-induced endothelial cell injury by increasing autophagy. Cell Signal. 2022;98:110410. doi:10.1016/j.cellsig.2022.110410

    67. Shi H, Kokoeva MV, Inouye K, et al. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest. 2006;116(11):3015–3025. doi:10.1172/jci28898

    68. Luo G, Shi Y, Zhang J, et al. Palmitic acid suppresses apolipoprotein M gene expression via the pathway of PPARβ/δ in HepG2 cells. Biochem Biophys Res Commun. 2014;445(1):203–207. doi:10.1016/j.bbrc.2014.01.170

    69. Bonanome A, Grundy SM. Effect of dietary stearic acid on plasma cholesterol and lipoprotein levels. New England J Med. 1988;318(19):1244–1248. doi:10.1056/nejm198805123181905

    70. Babayan VK. Plasma cholesterol responsiveness to saturated fatty acids. Am J Clin Nutr. 1988;48(6):1520–1522. doi:10.1093/ajcn/48.6.1520

    71. Hall E, Volkov P, Dayeh T, et al. Effects of palmitate on genome-wide mRNA expression and DNA methylation patterns in human pancreatic islets. BMC Med. 2014;12:103. doi:10.1186/1741-7015-12-103

    72. Breher-Esch S, Sahini N, Trincone A, et al. Genomics of lipid-laden human hepatocyte cultures enables drug target screening for the treatment of non-alcoholic fatty liver disease. BMC Medical Genomics. 2018;11(1):111. doi:10.1186/s12920-018-0438-7

    73. Yao Q, Liu J, Cui Q, et al. CCN1/Integrin α(5)β(1) instigates free fatty acid-induced hepatocyte lipid accumulation and pyroptosis through NLRP3 inflammasome activation. Nutrients. 2022;14(18). doi:10.3390/nu14183871

    74. Ding XQ, Jian TY, Gai YN, et al. Chicoric acid attenuated renal tubular injury in HFD-induced chronic kidney disease mice through the promotion of mitophagy via the Nrf2/PINK/parkin pathway. J Agric Food Chem. 2022;70(9):2923–2935. doi:10.1021/acs.jafc.1c07795

    75. Cook S, Konrad S, Goh Y, et al. Palmitic acid does not increase lipoprotein cholesterol levels when the diet contains recommended levels of linoleic acid. In: Proceedings of the Essential Fatty Acids and Eicosanoids Proceedings from the Fourth International Congress on Essential Fatty Acids and Eicosanoids. Edinburgh, Scotland: AOCS Press; 1997.

    76. Tholstrup T, Marckmann P, Jespersen J, et al. Fat high in stearic acid favorably affects blood lipids and factor VII coagulant activity in comparison with fats high in palmitic acid or high in myristic and lauric acids. Am J Clin Nutr. 1994;59(2):371–377. doi:10.1093/ajcn/59.2.371

    77. Hegsted DM, Mcgandy RB, Myers ML, et al. Quantitative effects of dietary fat on serum cholesterol in man. Am J Clin Nutr. 1965;17(5):281–295. doi:10.1093/ajcn/17.5.281

    78. Keys A, Anderson JT, Grande F. Prediction of serum-cholesterol responses of man to changes in fats in the diet. Lancet. 1957;273(7003):959–966. doi:10.1016/s0140-6736(57)91998-0

    79. Collins SC, Salehi A, Eliasson L, et al. Long-term exposure of mouse pancreatic islets to oleate or palmitate results in reduced glucose-induced somatostatin and oversecretion of glucagon. Diabetologia. 2008;51(9):1689–1693. doi:10.1007/s00125-008-1082-0

    80. Moullé VS, Vivot K, Tremblay C, et al. Glucose and fatty acids synergistically and reversibly promote beta cell proliferation in rats. Diabetologia. 2017;60(5):879–888. doi:10.1007/s00125-016-4197-8

    81. Poitout V, Amyot J, Semache M, et al. Glucolipotoxicity of the pancreatic beta cell. Biochimica et Biophysica Acta. 2010;1801(3):289–298. doi:10.1016/j.bbalip.2009.08.006

    82. Aggarwal R, Peng Z, Zeng N, et al. Chronic exposure to palmitic acid down-regulates AKT in beta-cells through activation of mTOR. Am J Pathol. 2022;192(1):130–145. doi:10.1016/j.ajpath.2021.09.008

    83. Gustavo Vazquez-Jimenez J, Chavez-Reyes J, Romero-Garcia T, et al. Palmitic acid but not palmitoleic acid induces insulin resistance in a human endothelial cell line by decreasing SERCA pump expression. Cell Signal. 2016;28(1):53–59. doi:10.1016/j.cellsig.2015.10.001

    84. Vazquez-Jimenez JG, Corpus-Navarro MS, Rodriguez-Chavez JM, et al. The increased expression of regulator of G-Protein Signaling 2 (RGS2) inhibits insulin-induced Akt phosphorylation and is associated with uncontrolled glycemia in patients with type 2 diabetes. Metabolites. 2021;11(2). doi:10.3390/metabo11020091

    85. Zhou BR, Zhang JA, Zhang Q, et al. Palmitic acid induces production of proinflammatory cytokines interleukin-6, interleukin-1β, and tumor necrosis factor-α via a NF-κB-dependent mechanism in HaCaT keratinocytes. Mediators Inflamm. 2013;2013:530429. doi:10.1155/2013/530429

    86. Shirasuna K, Takano H, Seno K, et al. Palmitic acid induces interleukin-1β secretion via NLRP3 inflammasomes and inflammatory responses through ROS production in human placental cells. J Reprod Immunol. 2016;116:104–112. doi:10.1016/j.jri.2016.06.001

    87. Bunn RC, Cockrell GE, Ou Y, et al. Palmitate and insulin synergistically induce IL-6 expression in human monocytes. Cardiovasc Diabetol. 2010;9:73. doi:10.1186/1475-2840-9-73

    88. Balta I, Stef L, Pet I, et al. Essential fatty acids as biomedicines in cardiac health. Biomedicines. 2021;9(10). doi:10.3390/biomedicines9101466

    89. Gan YR, Wei L, Wang YZ, et al. Dickkopf‑1/cysteine‑rich angiogenic inducer 61 axis mediates palmitic acid‑induced inflammation and apoptosis of vascular endothelial cells. Mol Med Rep. 2021;23(2). doi:10.3892/mmr.2020.11761

    90. Afonso MS, Lavrador MS, Koike MK, et al. Dietary interesterified fat enriched with palmitic acid induces atherosclerosis by impairing macrophage cholesterol efflux and eliciting inflammation. J Nutr Biochem. 2016;32:91–100. doi:10.1016/j.jnutbio.2016.01.005

    91. Karasawa T, Kawashima A, Usui-Kawanishi F, et al. Saturated fatty acids undergo intracellular crystallization and activate the NLRP3 inflammasome in macrophages. Arterioscler Thromb Vasc Biol. 2018;38(4):744–756. doi:10.1161/atvbaha.117.310581

    92. Sergi D, Morris AC, Kahn DE, et al. Palmitic acid triggers inflammatory responses in N42 cultured hypothalamic cells partially via ceramide synthesis but not via TLR4. Nutr Neurosci. 2020;23(4):321–334. doi:10.1080/1028415x.2018.1501533

    93. Hu X, Fatima S, Chen M, et al. Toll-like receptor 4 is a master regulator for colorectal cancer growth under high-fat diet by programming cancer metabolism. Cell Death Dis. 2021;12(8):791. doi:10.1038/s41419-021-04076-x

    94. Amine H, Benomar Y, Taouis M. Palmitic acid promotes resistin-induced insulin resistance and inflammation in SH-SY5Y human neuroblastoma. Sci Rep. 2021;11(1):5427. doi:10.1038/s41598-021-85018-7

    95. Huang S, Rutkowsky JM, Snodgrass RG, et al. Saturated fatty acids activate TLR-mediated proinflammatory signaling pathways. J Lipid Res. 2012;53(9):2002–2013. doi:10.1194/jlr.D029546

    96. Wang Z, Liu D, Wang F, et al. Saturated fatty acids activate microglia via toll-like receptor 4/NF-κB signalling. Br J Nutr. 2012;107(2):229–241. doi:10.1017/s0007114511002868

    97. Lancaster GI, Langley KG, Berglund NA, et al. Evidence that TLR4 is not a receptor for saturated fatty acids but mediates lipid-induced inflammation by reprogramming macrophage metabolism. Cell Metab. 2018;27(5):1096–1110.e1095. doi:10.1016/j.cmet.2018.03.014

    98. Teusch N, Lombardo E, Eddleston J, et al. The low molecular weight GTPase RhoA and atypical protein kinase Czeta are required for TLR2-mediated gene transcription. J Immunol. 2004;173(1):507–514. doi:10.4049/jimmunol.173.1.507

    99. Li J, Mao YS, Chen F, et al. Palmitic acid up regulates Gal-3 and induces insulin resistance in macrophages by mediating the balance between KLF4 and NF-κB. Exp Ther Med. 2021;22(3):1028. doi:10.3892/etm.2021.10460

    100. Hansson GK, Holm J, Jonasson L. Detection of activated T lymphocytes in the human atherosclerotic plaque. Am J Pathol. 1989;135(1):169–175.

    101. Jonasson L, Holm J, Skalli O, et al. Regional accumulations of T cells, macrophages, and smooth muscle cells in the human atherosclerotic plaque. Arteriosclerosis. 1986;6(2):131–138. doi:10.1161/01.atv.6.2.131

    102. Fernandez DM, Rahman AH, Fernandez NF, et al. Single-cell immune landscape of human atherosclerotic plaques. Nat Med. 2019;25(10):1576–1588. doi:10.1038/s41591-019-0590-4

    103. Depuydt MAC, Prange KHM, Slenders L, et al. Microanatomy of the human atherosclerotic plaque by single-cell transcriptomics. Circ Res. 2020;127(11):1437–1455. doi:10.1161/circresaha.120.316770

    104. Zernecke A, Winkels H, Cochain C, et al. Meta-analysis of leukocyte diversity in atherosclerotic mouse aortas. Circ Res. 2020;127(3):402–426. doi:10.1161/circresaha.120.316903

    105. Zhou X, Nicoletti A, Elhage R, et al. Transfer of CD4(+) T cells aggravates atherosclerosis in immunodeficient apolipoprotein E knockout mice. Circulation. 2000;102(24):2919–2922. doi:10.1161/01.cir.102.24.2919

    106. Winkels H, Ehinger E, Vassallo M, et al. Atlas of the immune cell repertoire in mouse atherosclerosis defined by single-cell RNA-sequencing and mass cytometry. Circ Res. 2018;122(12):1675–1688. doi:10.1161/circresaha.117.312513

    107. Reilly NA, Lutgens E, Kuiper J, et al. Effects of fatty acids on T cell function: role in atherosclerosis. Nat Rev Cardiol. 2021;18(12):824–837. doi:10.1038/s41569-021-00582-9

    108. Wang L, Folsom AR, Eckfeldt JH. Plasma fatty acid composition and incidence of coronary heart disease in middle aged adults: the Atherosclerosis Risk in Communities (ARIC) Study. Nutr Metab Cardiovasc Dis. 2003;13(5):256–266. doi:10.1016/s0939-4753(03)80029-7

    109. Cani PD, Amar J, Iglesias MA, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56(7):1761–1772. doi:10.2337/db06-1491

    110. Lu Z, Li Y, Jin J, et al. Palmitic acid enhances the stimulatory effect of lipopolysaccharide on interleukin 6 expression in cardiac microvascular endothelial cells by increasing ceramide production via both de novo synthesis and sphingomyelin hydrolysis. Am Heart Assoc. 2013;2013:1.

    111. Lu Z, Li Y, Brinson CW, et al. Cooperative stimulation of atherogenesis by lipopolysaccharide and palmitic acid-rich high fat diet in low-density lipoprotein receptor-deficient mice. Atherosclerosis. 2017;265:231–241. doi:10.1016/j.atherosclerosis.2017.09.008

    112. Furuhashi M, Ishimura S, Ota H, et al. Lipid chaperones and metabolic inflammation. Int J Inflam. 2011;2011:1.

    113. Furuhashi M, Hotamisligil GS. Fatty acid-binding proteins: role in metabolic diseases and potential as drug targets. Nat Rev Drug Discov. 2008;7(6):489–503. doi:10.1038/nrd2589

    114. Furuhashi M, Saitoh S, Shimamoto K, et al. Fatty Acid-Binding Protein 4 (FABP4): pathophysiological insights and potent clinical biomarker of metabolic and cardiovascular diseases. Clin Med Insights Cardiol. 2014;8(Suppl 3):23–33. doi:10.4137/cmc.S17067

    115. Hotamisligil GS, Bernlohr DA. Metabolic functions of FABPs–mechanisms and therapeutic implications. Nat Rev Endocrinol. 2015;11(10):592–605. doi:10.1038/nrendo.2015.122

    116. Furuhashi M. Fatty acid-binding protein 4 in cardiovascular and metabolic diseases. J Atheroscler Thromb. 2019;26(3):216–232. doi:10.5551/jat.48710

    117. Erbay E, Babaev VR, Mayers JR, et al. Reducing endoplasmic reticulum stress through a macrophage lipid chaperone alleviates atherosclerosis. Nat Med. 2009;15(12):1383–1391. doi:10.1038/nm.2067

    118. Li H, Xiao Y, Tang L, et al. Adipocyte fatty acid-binding protein promotes palmitate-induced mitochondrial dysfunction and apoptosis in macrophages. Front Immunol. 2018;9:81. doi:10.3389/fimmu.2018.00081

    119. Chmurzyńska A. The multigene family of fatty acid-binding proteins (FABPs): function, structure and polymorphism. J Appl Genet. 2006;47(1):39–48. doi:10.1007/bf03194597

    120. Makowski L, Brittingham KC, Reynolds JM, et al. The fatty acid-binding protein, aP2, coordinates macrophage cholesterol trafficking and inflammatory activity. Macrophage expression of aP2 impacts peroxisome proliferator-activated receptor gamma and IkappaB kinase activities. J Biol Chem. 2005;280(13):12888–12895. doi:10.1074/jbc.M413788200

    121. Xu H, Hertzel AV, Steen KA, et al. Loss of fatty acid binding protein 4/aP2 reduces macrophage inflammation through activation of SIRT3. Mol Endocrinol. 2016;30(3):325–334. doi:10.1210/me.2015-1301

    122. Gan L, Liu Z, Cao W, et al. FABP4 reversed the regulation of leptin on mitochondrial fatty acid oxidation in mice adipocytes. Sci Rep. 2015;5:13588. doi:10.1038/srep13588

    123. Xu H, Hertzel AV, Steen KA, et al. Uncoupling lipid metabolism from inflammation through fatty acid binding protein-dependent expression of UCP2. Mol Cell Biol. 2015;35(6):1055–1065. doi:10.1128/mcb.01122-14

    124. Kim SK, Oh E, Yun M, et al. Palmitate induces cisternal ER expansion via the activation of XBP-1/CCTα-mediated phospholipid accumulation in RAW 264.7 cells. Lipids Health Dis. 2015;14(1):1–12. doi:10.1186/s12944-015-0077-3

    125. Akoumi A, Haffar T, Mousterji M, et al. Palmitate mediated diacylglycerol accumulation causes endoplasmic reticulum stress, Plin2 degradation, and cell death in H9C2 cardiomyoblasts. Exp Cell Res. 2017;354(2):85–94. doi:10.1016/j.yexcr.2017.03.032

    126. Borradaile NM, Han X, Harp JD, et al. Disruption of endoplasmic reticulum structure and integrity in lipotoxic cell death. J Lipid Res. 2006;47(12):2726–2737. doi:10.1194/jlr.M600299-JLR200

    127. Kim SK, Oh E, Yun M, et al. Palmitate induces cisternal ER expansion via the activation of XBP-1/CCTα-mediated phospholipid accumulation in RAW 264.7 cells. Lipids Health Dis. 2015;14:73. doi:10.1186/s12944-015-0077-3

    128. Kim DH, Cho YM, Lee KH, et al. Oleate protects macrophages from palmitate-induced apoptosis through the downregulation of CD36 expression. Biochem Biophys Res Commun. 2017;488(3):477–482. doi:10.1016/j.bbrc.2017.05.066

    129. Go DH, Lee YG, Lee DH, et al. 3-Decylcatechol induces autophagy-mediated cell death through the IRE1α/JNK/p62 in hepatocellular carcinoma cells. Oncotarget. 2017;8(35):58790–58800. doi:10.18632/oncotarget.17732

    130. Zezina E, Snodgrass RG, Schreiber Y, et al. Mitochondrial fragmentation in human macrophages attenuates palmitate-induced inflammatory responses. Biochim Biophys Acta Mol Cell Biol Lipids. 2018;1863(4):433–446. doi:10.1016/j.bbalip.2018.01.009

    131. Pardo V, González-Rodríguez Á, Muntané J, et al. Role of hepatocyte S6K1 in palmitic acid-induced endoplasmic reticulum stress, lipotoxicity, insulin resistance and in oleic acid-induced protection. Food Chem Toxicol. 2015;80:298–309. doi:10.1016/j.fct.2015.03.029

    132. Ozcan U, Cao Q, Yilmaz E, et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 2004;306(5695):457–461. doi:10.1126/science.1103160

    133. Ozcan L, Ergin AS, Lu A, et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009;9(1):35–51. doi:10.1016/j.cmet.2008.12.004

    134. Biden TJ, Boslem E, Chu KY, et al. Lipotoxic endoplasmic reticulum stress, β cell failure, and type 2 diabetes mellitus. Trends Endocrinol Metab. 2014;25(8):389–398. doi:10.1016/j.tem.2014.02.003

    135. Rashid HO, Yadav RK, Kim HR, et al. ER stress: autophagy induction, inhibition and selection. Autophagy. 2015;11(11):1956–1977. doi:10.1080/15548627.2015.1091141

    136. Yang HY, Chen JY, Huo YN, et al. The role of sirtuin 1 in palmitic acid-induced endoplasmic reticulum stress in cardiac myoblasts. Life. 2022;12(2):182. doi:10.3390/life12020182

    137. Han J, Kaufman RJ. The role of ER stress in lipid metabolism and lipotoxicity. J Lipid Res. 2016;57(8):1329–1338. doi:10.1194/jlr.R067595

    138. Chen P, Liu H, Xiang H, et al. Palmitic acid-induced autophagy increases reactive oxygen species via the Ca(2+)/PKCα/NOX4 pathway and impairs endothelial function in human umbilical vein endothelial cells. Exp Ther Med. 2019;17(4):2425–2432. doi:10.3892/etm.2019.7269

    139. Hua W, Huang HZ, Tan LT, et al. CD36 mediated fatty acid-induced podocyte apoptosis via oxidative stress. PLoS One. 2015;10(5):e0127507. doi:10.1371/journal.pone.0127507

    140. Jiang XS, Chen XM, Hua W, et al. PINK1/Parkin mediated mitophagy ameliorates palmitic acid-induced apoptosis through reducing mitochondrial ROS production in podocytes. Biochem Biophys Res Commun. 2020;525(4):954–961. doi:10.1016/j.bbrc.2020.02.170

    141. Rosa Neto JC, Calder PC, Curi R, et al. The immunometabolic roles of various fatty acids in macrophages and lymphocytes. Int J Mol Sci. 2021;22(16). doi:10.3390/ijms22168460

    142. Xu W, Guo YB, Li X, et al. [Palmitic acid induces hepatocellular oxidative stress and activation of inflammasomes]. Nan Fang Yi Ke Da Xue Xue Bao. 2016;36(5):655–659. Sesotho

    143. Prabhahar A, Batta A, Hatwal J, et al. Endothelial dysfunction in the kidney transplant population: current evidence and management strategies. World J Transplant. 2025;15(1):97458. doi:10.5500/wjt.v15.i1.97458

    144. Schmidt-Lucke C, Rössig L, Fichtlscherer S, et al. Reduced number of circulating endothelial progenitor cells predicts future cardiovascular events: proof of concept for the clinical importance of endogenous vascular repair. Circulation. 2005;111(22):2981–2987. doi:10.1161/circulationaha.104.504340

    145. Frietze KK, Brown AM, Das D, et al. Lipotoxicity reduces DDX58/Rig-1 expression and activity leading to impaired autophagy and cell death. Autophagy. 2022;18(1):142–160. doi:10.1080/15548627.2021.1920818

    146. Stentz FB, Kitabchi AE. Palmitic acid-induced activation of human T-lymphocytes and aortic endothelial cells with production of insulin receptors, reactive oxygen species, cytokines, and lipid peroxidation. Biochem Biophys Res Commun. 2006;346(3):721–726. doi:10.1016/j.bbrc.2006.05.159

    147. Moers A, Schrezenmeir J. Palmitic acid but not stearic acid inhibits NO-production in endothelial cells. Exp Clin Endocrinol Diabetes. 1997;105(Suppl 2):78–80. doi:10.1055/s-0029-1211804

    148. Mao Y, Luo W, Zhang L, et al. STING-IRF3 triggers endothelial inflammation in response to free fatty acid-induced mitochondrial damage in diet-induced obesity. Arterioscler Thromb Vasc Biol. 2017;37(5):920–929. doi:10.1161/atvbaha.117.309017

    149. Carlström M, Weitzberg E, Lundberg JO. Nitric oxide signaling and regulation in the cardiovascular system: recent advances. Pharmacol Rev. 2024;76(6):1038–1062. doi:10.1124/pharmrev.124.001060

    150. Wang XL, Zhang L, Youker K, et al. Free fatty acids inhibit insulin signaling-stimulated endothelial nitric oxide synthase activation through upregulating PTEN or inhibiting Akt kinase. Diabetes. 2006;55(8):2301–2310. doi:10.2337/db05-1574

    151. Libby P, Buring JE, Badimon L, et al. Atherosclerosis. Nat Rev Dis Primers. 2019;5(1):56. doi:10.1038/s41572-019-0106-z

    152. Liu Q, Cheng Z, Huang B, et al. Palmitic acid promotes endothelial-to-mesenchymal transition via activation of the cytosolic DNA-sensing cGAS-STING pathway. Arch Biochem Biophys. 2022;727:109321. doi:10.1016/j.abb.2022.109321

    153. Seeger FH, Haendeler J, Walter DH, et al. p38 mitogen-activated protein kinase downregulates endothelial progenitor cells. Circulation. 2005;111(9):1184–1191. doi:10.1161/01.Cir.0000157156.85397.A1

    154. Jiang H, Liang C, Liu X, et al. Palmitic acid promotes endothelial progenitor cells apoptosis via p38 and JNK mitogen-activated protein kinase pathways. Atherosclerosis. 2010;210(1):71–77. doi:10.1016/j.atherosclerosis.2009.10.032

    155. Murphy JE, Tedbury PR, Homer-Vanniasinkam S, et al. Biochemistry and cell biology of mammalian scavenger receptors. Atherosclerosis. 2005;182(1):1–15. doi:10.1016/j.atherosclerosis.2005.03.036

    156. Ishiyama J, Taguchi R, Yamamoto A, et al. Palmitic acid enhances lectin-like oxidized LDL receptor (LOX-1) expression and promotes uptake of oxidized LDL in macrophage cells. Atherosclerosis. 2010;209(1):118–124. doi:10.1016/j.atherosclerosis.2009.09.004

    157. Murphy DL, Lesch KP. Targeting the murine serotonin transporter: insights into human neurobiology. Nat Rev Neurosci. 2008;9(2):85–96. doi:10.1038/nrn2284

    158. Caruso G, Fresta CG, Grasso M, et al. Inflammation as the common biological link between depression and cardiovascular diseases: can carnosine exert a protective role? Curr Med Chem. 2020;27(11):1782–1800. doi:10.2174/0929867326666190712091515

    159. Ma Y, Liang X, Li C, et al. 5-HT(2A) receptor and 5-HT degradation play a crucial role in atherosclerosis by modulating macrophage foam cell formation, vascular endothelial cell inflammation, and hepatic steatosis. J Atheroscler Thromb. 2022;29(3):322–336. doi:10.5551/jat.58305

    160. Luo Y, Duan H, Qian Y, et al. Macrophagic CD146 promotes foam cell formation and retention during atherosclerosis. Cell Research. 2017;27(3):352–372. doi:10.1038/cr.2017.8

    161. Duan H, Jing L, Xiang J, et al. CD146 associates with Gp130 to control a macrophage pro‐inflammatory program that regulates the metabolic response to obesity. Adv Sci. 2022;9(13):2103719. doi:10.1002/advs.202103719

    162. Cheng G, Zheng L. Regulation of the apolipoprotein M signaling pathway: a review. J Recept Signal Transduct Res. 2022;42(3):285–292. doi:10.1080/10799893.2021.1924203

    163. Zhang XY, Dong X, Zheng L, et al. Specific tissue expression and cellular localization of human apolipoprotein M as determined by in situ hybridization. Acta Histochem. 2003;105(1):67–72. doi:10.1078/0065-1281-00687

    164. Luo G, Zhang X, Mu Q, et al. Expression and localization of apolipoprotein M in human colorectal tissues. Lipids Health Dis. 2010;9:102. doi:10.1186/1476-511x-9-102

    165. Christoffersen C, Obinata H, Kumaraswamy SB, et al. Endothelium-protective sphingosine-1-phosphate provided by HDL-associated apolipoprotein M. Proc Natl Acad Sci U S A. 2011;108(23):9613–9618. doi:10.1073/pnas.1103187108

    166. Mousa H, Elrayess MA, Diboun I, et al. Metabolomics profiling of vitamin D status in relation to dyslipidemia. Metabolites. 2022;12(8). doi:10.3390/metabo12080771

    167. Elsøe S, Ahnström J, Christoffersen C, et al. Apolipoprotein M binds oxidized phospholipids and increases the antioxidant effect of HDL. Atherosclerosis. 2012;221(1):91–97. doi:10.1016/j.atherosclerosis.2011.11.031

    168. Wolfrum C, Poy MN, Stoffel M. Apolipoprotein M is required for prebeta-HDL formation and cholesterol efflux to HDL and protects against atherosclerosis. Nat Med. 2005;11(4):418–422. doi:10.1038/nm1211

    169. Ali FY, Armstrong PC, Dhanji AR, et al. Antiplatelet actions of statins and fibrates are mediated by PPARs. Arterioscler Thromb Vasc Biol. 2009;29(5):706–711. doi:10.1161/atvbaha.108.183160

    170. Yeung J, Adili R, Yamaguchi A, et al. Omega-6 DPA and its 12-lipoxygenase-oxidized lipids regulate platelet reactivity in a nongenomic PPARα-dependent manner. Blood Adv. 2020;4(18):4522–4537. doi:10.1182/bloodadvances.2020002493

    171. Yang ZH, Emma-Okon B, Remaley AT. Dietary marine-derived long-chain monounsaturated fatty acids and cardiovascular disease risk: a mini review. Lipids Health Dis. 2016;15(1):201. doi:10.1186/s12944-016-0366-5

    172. Shramko VS, Striukova EV, Polonskaya YV, et al. Associations of antioxidant enzymes with the concentration of fatty acids in the blood of men with coronary artery atherosclerosis. J Pers Med. 2021;11(12). doi:10.3390/jpm11121281

    173. Ebbesson SO, Tejero ME, López-Alvarenga JC, et al. Individual saturated fatty acids are associated with different components of insulin resistance and glucose metabolism: the GOCADAN study. Int J Circumpolar Health. 2010;69(4):344–351. doi:10.3402/ijch.v69i4.17669

    174. Li Y, Hruby A, Bernstein AM, et al. Saturated fats compared with unsaturated fats and sources of carbohydrates in relation to risk of coronary heart disease: a prospective cohort study. J Am Coll Cardiol. 2015;66(14):1538–1548. doi:10.1016/j.jacc.2015.07.055

    175. S AIS, B CA, S AJ. Changes in plasma free fatty acids associated with type-2 diabetes. Nutrients. 2019;11(9). doi:10.3390/nu11092022

    176. Alsharari ZD, Risérus U, Leander K, et al. Serum fatty acids, desaturase activities and abdominal obesity – a population-based study of 60-year old men and women. PLoS One. 2017;12(1):e0170684. doi:10.1371/journal.pone.0170684

    177. Bucalossi A, Mori S. Fatty acid composition of adipose tissue in ischemic heart disease and stroke. Gerontol Clin. 1972;14(6):339–345. doi:10.1159/000245419

    178. Insull W, Lang P, Hsi B. Adipose tissue fatty acids and extent of coronary atherosclerosis. In: Proceedings of the Circulation; Philadelphia, PA: Lippincott Williams & Wilkins 1968:19106.

    179. Lee K, Shaper A, Scott R, et al. Geographic studies pertaining to arteriosclerosis: comparison of fatty acid patterns of adipose tissue and plasma lipids in East Africans with those of North American white and Negro groups. Arch Pathol. 1962;74:481–488.

    180. Lopaschuk GD, Ussher JR, Folmes CD, et al. Myocardial fatty acid metabolism in health and disease. Physiol Rev. 2010;90(1):207–258. doi:10.1152/physrev.00015.2009

    181. Tomata Y, Wang Y, Hägg S, et al. Fatty acids and frailty: a mendelian randomization study. Nutrients. 2021;13(10). doi:10.3390/nu13103539

    182. Cetrullo S, Tantini B, Flamigni F, et al. Antiapoptotic and antiautophagic effects of eicosapentaenoic acid in cardiac myoblasts exposed to palmitic acid. Nutrients. 2012;4(2):78–90. doi:10.3390/nu4020078

    183. Dyntar D, Eppenberger-Eberhardt M, Maedler K, et al. Glucose and palmitic acid induce degeneration of myofibrils and modulate apoptosis in rat adult cardiomyocytes. Diabetes. 2001;50(9):2105–2113. doi:10.2337/diabetes.50.9.2105

    184. Lemaitre RN, Jensen PN, Hoofnagle A, et al. Plasma ceramides and sphingomyelins in relation to heart failure risk. Circ Heart Fail. 2019;12(7):e005708. doi:10.1161/circheartfailure.118.005708

    185. Hooper L, Martin N, Jimoh OF, et al. Reduction in saturated fat intake for cardiovascular disease. Cochrane Database Syst Rev. 2020;5(5):Cd011737. doi:10.1002/14651858.CD011737.pub2

    186. Wang DD, Li Y, Chiuve SE, et al. Association of specific dietary fats with total and cause-specific mortality. JAMA Intern Med. 2016;176(8):1134–1145. doi:10.1001/jamainternmed.2016.2417

    187. Duda MK, O’shea KM, Stanley WC. omega-3 polyunsaturated fatty acid supplementation for the treatment of heart failure: mechanisms and clinical potential. Cardiovasc Res. 2009;84(1):33–41. doi:10.1093/cvr/cvp169

    188. Cerf ME, Louw J. Islet cell response to high fat programming in neonate, weanling and adolescent Wistar rats. JOP. 2014;15(3):228–236. doi:10.6092/1590-8577/1534

    189. Taegtmeyer H, Stanley WC. Too much or not enough of a good thing? Cardiac glucolipotoxicity versus lipoprotection. J Mol Cell Cardiol. 2011;50(1):2–5. doi:10.1016/j.yjmcc.2010.09.014

    190. Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–1072. doi:10.1016/j.cell.2012.03.042

    191. Stockwell BR, Friedmann Angeli JP, Bayir H, et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 2017;171(2):273–285. doi:10.1016/j.cell.2017.09.021

    192. Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22(4):266–282. doi:10.1038/s41580-020-00324-8

    193. Ke C, Pan CW, Zhang Y, et al. Metabolomics facilitates the discovery of metabolic biomarkers and pathways for ischemic stroke: a systematic review. Metabolomics. 2019;15(12):152. doi:10.1007/s11306-019-1615-1

    194. Fielding CJ, Fielding PE. Cholesterol transport between cells and body fluids. Role of plasma lipoproteins and the plasma cholesterol esterification system. Med Clin North Am. 1982;66(2):363–373. doi:10.1016/s0025-7125(16)31425-0

    195. Satizabal CL, Samieri C, Davis-Plourde KL, et al. APOE and the association of fatty acids with the risk of stroke, coronary heart disease, and mortality. Stroke. 2018;49(12):2822–2829. doi:10.1161/strokeaha.118.022132

    196. Yamagishi K, Folsom AR, Steffen LM, et al. Plasma fatty acid composition and incident ischemic stroke in middle-aged adults: the Atherosclerosis Risk in Communities (ARIC) Study. Cerebrovasc Dis. 2013;36(1):38–46. doi:10.1159/000351205

    197. Thaler JP, Yi CX, Schur EA, et al. Obesity is associated with hypothalamic injury in rodents and humans. J Clin Invest. 2012;122(1):153–162. doi:10.1172/jci59660

    198. Moraes JC, Coope A, Morari J, et al. High-fat diet induces apoptosis of hypothalamic neurons. PLoS One. 2009;4(4):e5045. doi:10.1371/journal.pone.0005045

    199. Ricchi M, Odoardi MR, Carulli L, et al. Differential effect of oleic and palmitic acid on lipid accumulation and apoptosis in cultured hepatocytes. J Gastroenterol Hepatol. 2009;24(5):830–840. doi:10.1111/j.1440-1746.2008.05733.x

    200. Mayer CM, Belsham DD. Palmitate attenuates insulin signaling and induces endoplasmic reticulum stress and apoptosis in hypothalamic neurons: rescue of resistance and apoptosis through adenosine 5’ monophosphate-activated protein kinase activation. Endocrinology. 2010;151(2):576–585. doi:10.1210/en.2009-1122

    201. Truran S, Karamanova N, Serrano G, et al. Palmitic acid-induced endothelial dysfunction in human leptomeningeal and adipose arterioles. Circulation. 2015;132(suppl_3):A18245–A18245.

    202. Karamanova N, Truran S, Madine J, et al. Medin amyloid, but not β-amyloid, induces pro-inflammatory signaling in endothelial cells that is synergistic with palmitic acid. Circulation. 2017;136(suppl_1):A14270–A14270.

    203. Posey KA, Clegg DJ, Printz RL, et al. Hypothalamic proinflammatory lipid accumulation, inflammation, and insulin resistance in rats fed a high-fat diet. Am J Physiol Endocrinol Metab. 2009;296(5):E1003–1012. doi:10.1152/ajpendo.90377.2008

    204. Milanski M, Degasperi G, Coope A, et al. Saturated fatty acids produce an inflammatory response predominantly through the activation of TLR4 signaling in hypothalamus: implications for the pathogenesis of obesity. J Neurosci. 2009;29(2):359–370. doi:10.1523/jneurosci.2760-08.2009

    205. Lu Z, Liu S, Lopes-Virella MF, et al. LPS and palmitic acid Co-upregulate microglia activation and neuroinflammatory response. Compr Psychoneuroendocrinol. 2021;6:100048. doi:10.1016/j.cpnec.2021.100048

    206. Zhang Y, Dong L, Yang X, et al. α-Linolenic acid prevents endoplasmic reticulum stress-mediated apoptosis of stearic acid lipotoxicity on primary rat hepatocytes. Lipids Health Dis. 2011;10:81. doi:10.1186/1476-511x-10-81

    207. Ng YW, Say YH. Palmitic acid induces neurotoxicity and gliatoxicity in SH-SY5Y human neuroblastoma and T98G human glioblastoma cells. PeerJ. 2018;6:e4696. doi:10.7717/peerj.4696

    208. Xue X, Li F, Cai M, et al. Interactions between endoplasmic reticulum stress and autophagy: implications for apoptosis and neuroplasticity-related proteins in palmitic acid-treated prefrontal cells. Neural Plast. 2021;2021:8851327. doi:10.1155/2021/8851327

    209. Khan M, Rutten BPF, Kim MO. MST1 regulates neuronal cell death via JNK/Casp3 signaling pathway in HFD mouse brain and HT22 cells. Int J Mol Sci. 2019;20(10). doi:10.3390/ijms20102504

    210. Hernández-Cáceres MP, Cereceda K, Hernández S, et al. Palmitic acid reduces the autophagic flux in hypothalamic neurons by impairing autophagosome-lysosome fusion and endolysosomal dynamics. Mol Cell Oncol. 2020;7(5):1789418. doi:10.1080/23723556.2020.1789418

    211. Fu CN, Wei H, Gao WS, et al. Obesity increases neuropathic pain via the AMPK-ERK-NOX4 pathway in rats. Aging. 2021;13(14):18606–18619. doi:10.18632/aging.203305

    212. Deng W, Mandeville E, Terasaki Y, et al. Transcriptomic characterization of microglia activation in a rat model of ischemic stroke. J Cereb Blood Flow Metab. 2020;40(1_suppl):S34–s48. doi:10.1177/0271678×20932870

    213. Zhou YD. Glial regulation of energy metabolism. Adv Exp Med Biol. 2018;1090:105–121. doi:10.1007/978-981-13-1286-1_6

    214. Yang C, Sui G, Wang L, et al. MiR-124 prevents the microglial proinflammatory response by inhibiting the activities of TLR4 and downstream NLRP3 in palmitic acid-treated BV2 cells. J Mol Neurosci. 2022;72(3):496–506. doi:10.1007/s12031-021-01921-8

    215. Chowen JA, Frago LM, Fernández-Alfonso MS. Physiological and pathophysiological roles of hypothalamic astrocytes in metabolism. J Neuroendocrinol. 2019;31(5):e12671. doi:10.1111/jne.12671

    216. Joyal JS, Sun Y, Gantner ML, et al. Retinal lipid and glucose metabolism dictates angiogenesis through the lipid sensor Ffar1. Nat Med. 2016;22(4):439–445. doi:10.1038/nm.4059

    Continue Reading

  • Art Basel Paris 2025 cheat sheet

    Art Basel Paris 2025 cheat sheet

    Become a Vogue Business Member to receive unlimited access to Member-only reporting and insights, our Beauty and TikTok Trend Trackers, Member-only newsletters and exclusive event invitations.

    If you think you’ve been busy lately, spare a…

    Continue Reading

  • Insulin Resistance as a Biomarker for Pelvic Organ Prolapse in Gestati

    Insulin Resistance as a Biomarker for Pelvic Organ Prolapse in Gestati

    Introduction

    Gestational diabetes mellitus (GDM) rates have increased steadily along with obesity and type 2 diabetes mellitus incidence worldwide.1 Pregnant women are susceptible to GDM, which is a metabolic disorder specific to pregnancy that…

    Continue Reading