1
|
Huang W, Zhu W, Lin Y, Chan FKL, Xu Z, Ng SC. Roseburia hominis improves host metabolism in diet-induced obesity. Gut Microbes 2025; 17:2467193. [PMID: 39976263 PMCID: PMC11845086 DOI: 10.1080/19490976.2025.2467193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025] Open
Abstract
Next-generation live biotherapeutics are promising to aid the treatment of obesity and metabolic diseases. Here, we reported a novel anti-obesity probiotic candidate, Roseburia hominis, that was depleted in stool samples of obese subjects compared with lean controls, and its abundance was negatively correlated with body mass index and serum triglycerides. Supplementation of R. hominis prevented body weight gain and disorders of glucose and lipid metabolism, prevented fatty liver, inhibited white adipose tissue expansion and brown adipose tissue whitening in mice fed with high-fat diet, and boosted the abundance of lean-related species. The effects of R. hominis could be partially attributed to the production of nicotinamide riboside and upregulation of the Sirtuin1/mTOR signaling pathway. These results indicated that R. hominis is a promising candidate for the development of next-generation live biotherapeutics for the prevention of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Wenli Huang
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Wenyi Zhu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Lin
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Francis K. L. Chan
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhilu Xu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Siew C. Ng
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
2
|
Wang H, Qin Y, Niu J, Chen H, Lu X, Wang R, Han J. Evolving perspectives on evaluating obesity: from traditional methods to cutting-edge techniques. Ann Med 2025; 57:2472856. [PMID: 40077889 PMCID: PMC11912248 DOI: 10.1080/07853890.2025.2472856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Objective: This review examines the evolution of obesity evaluation methods, from traditional anthropometric indices to advanced imaging techniques, focusing on their clinical utility, limitations, and potential for personalized assessment of visceral adiposity and associated metabolic risks. Methods: A comprehensive analysis of existing literature was conducted, encompassing anthropometric indices (BMI, WC, WHR, WHtR, NC), lipid-related metrics (LAP, VAI, CVAI, mBMI), and imaging technologies (3D scanning, BIA, ultrasound, DXA, CT, MRI). The study highlights the biological roles of white, brown, and beige adipocytes, emphasizing visceral adipose tissue (VAT) as a critical mediator of metabolic diseases. Conclusion: Although BMI and other anthropometric measurements are still included in the guidelines, indicators that incorporate lipid metabolism information can more accurately reflect the relationship between metabolic diseases and visceral obesity. At the same time, the use of more modern medical equipment, such as ultrasound, X-rays, and CT scans, allows for a more intuitive assessment of the extent of visceral obesity.
Collapse
Affiliation(s)
- Heyue Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaxin Qin
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinzhu Niu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haowen Chen
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinda Lu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianli Han
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
3
|
Terada H, Kojima T, Takasu C, Kawabata S, Shimada N, Nihei K, Takayanagi K, Kanemura N, Murata K. Fibrosis of the infrapatellar fat pad induces gradual cartilage degeneration in a rat model. Tissue Cell 2025; 95:102851. [PMID: 40086109 DOI: 10.1016/j.tice.2025.102851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Knee osteoarthritis (OA), a degenerative joint disease, causes pain and reduced activity levels, with prevalence increasing with age. Knee OA is recognized as a "joint organ disease," where interactions between the articular cartilage, meniscus, synovium, and infrapatellar fat pad (IFP) are critical. Obesity contributes to OA not only through mechanical stress but also via systemic and local inflammation mediated by adipose tissue. Adiponectin, an adipokine with anti-inflammatory and chondroprotective effects, shows levels negatively correlated with obesity and positively correlated with OA severity. This study explored the role of IFP in OA progression using a rat model of monoiodoacetic acid-induced IFP fibrosis. In vivo, IFP and cartilage degeneration were assessed via histological analysis, real-time PCR, and RNA sequencing. Ex vivo, the effects of conditioned media from synovial cells and IFP on chondrocytes were evaluated. Findings revealed that fibrosis reduced adiponectin expression and accelerated OA progression. Gene sequencing showed significant metabolic pathway changes, particularly in fatty acid β-oxidation, indicating impaired IFP function. Conditioned media from inflamed IFP negatively affected chondrocytes by reducing anabolic factors and increasing cartilage-degrading enzymes. These results suggest that IFP degeneration contributes to OA by promoting inflammation and cartilage degradation. Targeting IFP fibrosis and fatty acid metabolism may offer therapeutic potential to mitigate OA progression.
Collapse
Affiliation(s)
- Hidenobu Terada
- Clinical Center for Sports Medicine and Sports Dentistry, Institute of Science Tokyo Hospital, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8519, Japan
| | - Takuma Kojima
- Department of Rehabilitation, Soka Orthopedics Internal Medicine, 1-1-18 Chuo, Soka-shi, Saitama 340-0016, Japan
| | - Chiharu Takasu
- Department of Health and Social Services, Graduate School of Saitama Prefectural University, 820 Sannomiya, Koshigaya-shi, Saitama 343-8540, Japan
| | - Sora Kawabata
- Department of Health and Social Services, Graduate School of Saitama Prefectural University, 820 Sannomiya, Koshigaya-shi, Saitama 343-8540, Japan
| | - Naoki Shimada
- Department of Health and Social Services, Graduate School of Saitama Prefectural University, 820 Sannomiya, Koshigaya-shi, Saitama 343-8540, Japan
| | - Kota Nihei
- Department of Health and Social Services, Graduate School of Saitama Prefectural University, 820 Sannomiya, Koshigaya-shi, Saitama 343-8540, Japan
| | - Kiyomi Takayanagi
- Toto Rehabilitation Academy, 2-4-2 Ohashi, Meguro-Ku, Tokyo 153-0044, Japan
| | - Naohiko Kanemura
- Department of Physical Therapy, School of Health and Social Services, Saitama Prefectural University, 820 Sannomiya, Koshigaya-shi, Saitama 343-8540, Japan
| | - Kenji Murata
- Department of Physical Therapy, School of Health and Social Services, Saitama Prefectural University, 820 Sannomiya, Koshigaya-shi, Saitama 343-8540, Japan.
| |
Collapse
|
4
|
Vargas MR, Ferreira MDR, Collins P, D'Alessandro ME. Astaxanthin obtained from freshwater crustaceans mitigates visceral adiposity by modulating adipose tissue lipogenesis and ameliorates dyslipidemia in high-sucrose diet fed rats. J Nutr Biochem 2025; 142:109924. [PMID: 40245956 DOI: 10.1016/j.jnutbio.2025.109924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/28/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
The incidence of overweight and obesity continues to grow at alarming rates around the world. The search for foods with potential benefits in the prevention/treatment of overweight/obesity and related disorders has great relevance. The aim of this study was to evaluate the effect of astaxanthin (ASTX) from freshwater crustaceans (crabs) upon visceral adiposity, adipose tissue lipid metabolism disorders and dyslipidemia present in a Metabolic Syndrome rodent model. Male Wistar rats were fed for 90 days with 1 of 4 experimental diets: a-Reference group (RD) received a standard commercial rodent diet, b- High-sucrose diet (HSD) group received a HSD, c- RD+ASTX group received a standard commercial rodent diet plus ASTX, d- HSD+ASTX group received a HSD plus ASTX. The rats were given orally either ASTX (10 mg/kg body weight/day in sunflower oil) or only sunflower oil. Compared with HSD-fed rats, HSD+ASTX group had lower body weight gain (19%) and both reduced abdominal circumference (5%) and visceral adiposity index (5%). Energy intake was 24% lower at the middle of the experimental period. Epididymal adipocytes size and triglyceride (TG) content was reduced by 14%. Besides, fatty acid synthase, malic enzyme and glucose-6-phosphate dehydrogenase activities in epididymal adipose tissue were 43%, 28% and 38% lower respectively. These changes were accompanied by lower TG (25%) and cholesterol (27%) serum levels, atherogenic index (31%) and reduced Systolic (12%) and Diastolic (15%) blood pressure. The results show that ASTX could be a potential strategy to prevent/attenuate the incidence of metabolic risk factors such as overweight/adiposity and dyslipidemia.
Collapse
Affiliation(s)
- Matias Rodrigo Vargas
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición. Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, Santa Fe, 3000, Argentina
| | - María Del Rosario Ferreira
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición. Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, Santa Fe, 3000, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina.
| | - Pablo Collins
- Departamento de Acuicultura, COE INTA Ángel Gallardo (EEA Rafaela), Universidad Nacional del Litoral- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - María Eugenia D'Alessandro
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición. Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, Santa Fe, 3000, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina.
| |
Collapse
|
5
|
Vratarić M, Teofilović A, Milutinović DV, Veličković N, Vučićević L, Đmura G, Djordjevic A. Changes in lipid metabolism in the visceral rather than the subcutaneous adipose tissue depot attenuate metabolic disturbances in obesity-resistant mice fed a high-fat diet. J Nutr Biochem 2025; 141:109912. [PMID: 40174754 DOI: 10.1016/j.jnutbio.2025.109912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Obesity is characterized by an enlargement of white adipose tissue caused by caloric excess. The depot-specific adaptation of white adipose tissue in individuals resistant to obesity despite a high-calorie diet is crucial for understanding the pathogenesis of obesity and related metabolic disorders. Our aim was to characterize the metabolic and morphological state of obesity resistance and to investigate depot-specific changes in signaling pathways in epididymal visceral (eVAT) and inguinal subcutaneous (iSAT) white adipose tissue of C57BL/6J male mice on a high-fat diet (60 kcal% fats). After 14 weeks, the mice were categorized as obese (at least 30% higher body mass compared to the control group) or obesity-resistant (weight gain below 30%). Biochemical and morphological parameters, as well as histology, and signaling pathways involved in lipid metabolism, inflammation, and insulin sensitivity were investigated in eVAT and iSAT. The results showed unaltered body, total VAT and iSAT mass in obesity-resistant mice despite increased caloric intake. Leptin levels and glucose homeostasis were improved in these animals compared to the obese mice. In both eVAT and iSAT of the obesity-resistant mice, adipocyte size and lipolytic capacity were retained at control levels, while compared to the obese mice, preserved capacity for adipogenesis, improved local insulin sensitivity and the absence of inflammation were observed only in the eVAT. In conclusion, metabolic adaptation of eVAT rather than iSAT may have a substantial impact on the maintenance of the obesity-resistant phenotype with fewer metabolic complications, which could contribute to the improvement of existing obesity therapies.
Collapse
Affiliation(s)
- Miloš Vratarić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Teofilović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Danijela Vojnović Milutinović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nataša Veličković
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ljubica Vučićević
- Department of Neurophysiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Goran Đmura
- Animal Facility, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Djordjevic
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
6
|
Shen X, Liu Q, Lin T, Zheng D, He Q. Association between Chinese visceral adiposity index and cardiovascular events risk in individuals with cardiovascular-kidney-metabolic syndrome stage 0-3: a nationwide cohort study. Int Urol Nephrol 2025; 57:2255-2269. [PMID: 39934555 DOI: 10.1007/s11255-025-04403-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND The population with cardiovascular-kidney-metabolic (CKM) syndrome has a higher risk of cardiovascular events. Chinese visceral adiposity index (CVAI), an index of both visceral obesity and surrogate insulin resistance, has been linked to cardiovascular events. However, the nature of this relationship remains unclear in individuals with CKM syndrome. METHODS All data came from the China Health and Retirement Longitudinal Study (CHARLS). The association between CVAI and cardiovascular events risk was explored using Cox regression models, restricted cubic spline (RCS) curves, and multiple sensitivity analyses. To compare the predictive abilities of various indices, receiver operating characteristic (ROC) analyses were employed. RESULTS 7744 participants were in final analysis. During 9 year follow-up, 1679 cases of cardiovascular disease (CVD), 1,255 cases of heart disease, and 604 cases of stroke were recorded. Cox regression analyses revealed that per-SD (standard deviation) increase in CVAI, the risk of CVD, heart disease, and stroke increased by 22% (95% CI 1.13-1.32), 22% (95% CI 1.13-1.32), and 32% (95% CI 1.19-1.47). In participants at CKM stage 3 with CVD, a J-shaped curve was observed in the RCS analyses (P non-linearity = 0.036). Subgroup analysis revealed an interaction between age and each 10-unit increase in CVAI in CVD (P interaction = 0.0173) and stroke risk (P interaction = 0.028). The AUC (area under curve) value for CVAI was highest compared to other indicators (all DeLong Test P values < 0.05). CONCLUSIONS This research demonstrates a higher CVAI was linked to increased cardiovascular risk in individuals with CKM syndrome stage 0-3. Monitoring CVAI can help identify high-risk individuals early and improve the effectiveness of disease management.
Collapse
Affiliation(s)
- Xiaobo Shen
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Qi Liu
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Tianchen Lin
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Danna Zheng
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiang He
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Tran NNQ, Choi H, Sactivel B, Oh YJ, Maeng HJ, Kim MK, Lee J, Kim YB, Lee DH, Oh BC, Jun HS, Chun KH. The dual targeting effects of KD025 on casein kinase 2 and ROCK2 in a mouse model of diet-induced obesity. Biochem Pharmacol 2025; 237:116933. [PMID: 40210126 DOI: 10.1016/j.bcp.2025.116933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/16/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
KD025(belumosudil), a selective ROCK2 inhibitor, exhibits unique anti-adipogenic activity through inhibition of casein kinase 2 (CK2). This study investigated the dual inhibitory effects of KD025 on metabolism in a diet-induced obese model. C57BL/6 mice on a high fat diet (HFD) were treated with KD025 for 4 weeks, while fasudil (a pan-ROCK inhibitor) and CX-4945 (a CK2-specific inhibitor) served as comparison treatments. KD025 significantly reduced body weight gain without affecting food intake, serum insulin, or fasting blood glucose levels. In contrast, while both CX-4945 and fasudil treatments showed a trend toward weight reduction, these results were not statistically significant. KD025 improved lipid metabolism by significantly lowering LDL cholesterol and triglyceride, although it slightly impaired glucose metabolism, as observed in insulin and glucose tolerance tests. Weight reduction in the KD025- and CX-4945-treated groups was attributed to decreased adipose tissue mass, particularly in inguinal (ingWAT) and epididymal (epiWAT) fat depots. Hematoxylin and eosin (H&E) staining confirmed smaller adipocyte size in these groups. KD025 had no significant effect on serum levels of tumor necrosis factor-α (TNF-α), interleukin 6 (IL-6), or monocyte chemoattractant protein-1 (MCP-1) with varied inflammatory responses. Furthermore, KD025 and CX-4945 upregulated adipogenic and browning markers, such as Cebpa, Cidea, and Pparg in the epiWAT, though without significant UCP1 expression. Overall, KD025 effectively reduced weight gain in HFD-fed mice through dual inhibition of CK2 and ROCK2, highlighting its potential as a therapeutic agent for obesity-related conditions.
Collapse
Affiliation(s)
- Nhu Nguyen Quynh Tran
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Hojung Choi
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Bathiga Sactivel
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Yu Jin Oh
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Han-Joo Maeng
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Min Kyung Kim
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Jeongmi Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, United States
| | - Dae Ho Lee
- Department of Internal Medicine, Gachon University College of Medicine, Incheon 21565, Republic of Korea
| | - Byung-Chul Oh
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea
| | - Hee-Sook Jun
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea
| | - Kwang-Hoon Chun
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea.
| |
Collapse
|
8
|
Son YL, Hou J, Kato-Suzuki M, Okamatsu-Ogura Y, Watase M, Kiyonari H, Kondo T. Eva1 deficiency prevents obesity-induced metabolic disorders by reducing visceral adipose dysfunction. Metabolism 2025; 168:156235. [PMID: 40118448 DOI: 10.1016/j.metabol.2025.156235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
AIMS Epithelial V-like antigen 1 (Eva1) is a highly specific marker for brown adipose tissue (BAT) in both mice and humans, but its metabolic function remains unclear. We investigated the impact of Eva1 deletion on the development of obesity. METHODS To assess the metabolic role of Eva1, we generated whole-body and adipocyte-specific Eva1knockout (KO) mice, which were subjected to a high-fat diet (HFD) for 12 weeks and characterized metabolic phenotypes. To further elucidate the depot-dependent impact of Eva1 deficiency, we performed histological analysis and 3' mRNA-seq of BAT and epididymal visceral white adipose tissue (eWAT). To investigate the role of macrophage-derived Eva1 in obesity development, we transplanted wild-type (WT) or Eva1KO macrophages into Eva1KO mice fed an HFD. RESULTS We found that whole-body Eva1KO mice are resistant to HFD-induced obesity, insulin resistance and visceral adipose inflammation. However, Eva1 deletion in adipocytes, both brown and white, did not phenocopy these protective effects. Notably, whole-body Eva1 deficiency triggers functional changes in eWAT, but not in BAT. These results led us to investigate a possible involvement of macrophages in Eva1-mediated obesity regulation. We found that Eva1 is expressed in macrophages and plays a role in lipopolysaccharide (LPS)-induced inflammatory responses, possibly through the direct interaction with toll-like receptor 4 (TLR4). Moreover, Eva1KO mice exhibited improved survival rates in the face of severe sepsis induced by LPS. Importantly, transplantation of WT macrophages to Eva1KO mice abolished the beneficial effects of whole-body Eva1 deletion against obesity and visceral adipose inflammation. CONCLUSION Our findings highlight macrophage-derived Eva1 as an important mediator in obesity-induced eWAT remodeling, suggesting that targeting Eva1 could offer a novel therapeutic strategy for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- You Lee Son
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
| | - Jiahui Hou
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Mira Kato-Suzuki
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Megumi Watase
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Toru Kondo
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
9
|
Alsenousy AHA, Sharker SA, Gowayed MA, Elblehi SS, Kamel MA. Aptamer-targeted anti-miR RNA construct based on 3WJ as a new approach for the treatment of chronic kidney disease in an experimental model. Gene Ther 2025:10.1038/s41434-025-00544-7. [PMID: 40514411 DOI: 10.1038/s41434-025-00544-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 05/06/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025]
Abstract
The treatment of chronic disease (CKD) is a great challenge in healthcare that requires an innovative approach to address its complex nature. RNA nanotechnology has emerged rapidly and received attention in the last few years because of its significant aptitude for therapies. Hence, the present study aimed to design, construct, and characterize a multifunctional (anti-miR-34a DNA aptamer-kidney targeted) RNA nanoparticle (RNPs) based on bacteriophage phi29 packaging RNA three-way junction (pRNA-3WJ), and then explore their in vivo toxicity and therapeutic potentials in mice model of CKD. After confirming the safety and specific targeting capability of the prepared core 3WJ (3WJ) and the therapeutic 3WJ (3WJ-Kapt/anti-miR-34a) RNPs to renal tissue using healthy mice, CKD was induced in C57BL/6 mice using adenine. CKD mice were treated with a single intravenous injection of 3WJ or 3WJ-Kapt/anti-miR-34a. Every week, 5 mice of each group were selected randomly for sample collection for 4 weeks post-treatment. The anti-miR-34a 3WJ-RNPs have shown stability, safety, and efficacy in renal targeting using DNA aptamer, by targeting miR-34a in renal tissue, 3WJ-Kapt/anti-miR-34a suppressed profibrotic gene expression and induced anti-fibrotic pathways' expression. Our present study provides preliminary and pioneering evidence for the promising treatment of renal fibrosis and CKD through targeting miR-34a in the renal tissue by 3WJ-RNPs. The CKD mice showed marked time-dependent up-regulation of the renal profibrotic pathways, including TGF-β, FGF2, and WNT/β-catenin pathways. The same mice showed suppressed renal expression of the antifibrotic pathways, including α and β Klotho, SMAD7, and SIRT1. The prepared anti-miR-34a 3WJ-RNPs have shown stability, safety, and efficacy in renal targeting using DNA aptamer. By targeting miR-34a in renal tissue, 3WJ-Kapt/anti-miR-34a suppressed profibrotic gene expression and induced anti-fibrotic pathways' expression. Our present study provides preliminary and pioneer evidence for the promising treatment of renal fibrosis and CKD through targeting miR-34a in the renal tissue by 3WJ-RNPs.
Collapse
Affiliation(s)
- Aisha H A Alsenousy
- Department of Biochemistry, Medical Research Institute, University of Alexandria, Alexandria, Egypt.
| | - Sara A Sharker
- Department of Biochemistry, Medical Research Institute, University of Alexandria, Alexandria, Egypt
| | - Mennatallah A Gowayed
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Samar S Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Maher A Kamel
- Department of Biochemistry, Medical Research Institute, University of Alexandria, Alexandria, Egypt
- Research Projects unit, Pharos University in Alexandria, 21648, Alexandria, Egypt
| |
Collapse
|
10
|
Mohaghegh N, Iyer A, Wang E, Balajam NZ, Kang H, Akbari M, Barnhill MS, Khademhosseini A, Pearson RM, Hassani Najafabadi A. Apigenin-loaded nanoparticles for obesity intervention through immunomodulation and adipocyte browning. J Control Release 2025; 382:113670. [PMID: 40187647 DOI: 10.1016/j.jconrel.2025.113670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/04/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
Obesity is characterized by a significant imbalance in adipose tissue macrophages (ATMs), shifting from anti-inflammatory M2 to pro-inflammatory M1 phenotypes, contributing to chronic low-grade inflammation and metabolic dysfunction. This study explores the potential of nanoparticle (NP)-mediated immunomodulation to address obesity-related inflammation, adipocyte browning, and metabolic dysfunction. Apigenin (Api), a natural compound with notable anti-inflammatory properties, was encapsulated within poly(lactic-co-glycolic acid) (PLGA) NPs (Api-NPs) for localized delivery to adipose tissues (ATs). Api-NPs demonstrated favorable physicochemical properties and sustained release profiles. In vitro, Api-NPs, increased M2 macrophage (MΦ) polarization, reduced inflammatory markers, and promoted adipocyte browning. In a high-fat diet (HFD)-induced obesity mouse model, Api-NP administration effectively modulated MΦ polarization and enhanced AT browning, leading to marked reductions in body weight and AT mass. Our findings indicate that Api-NP treatment mitigates obesity-related inflammation and promotes beneficial changes in AT composition and function. Importantly, histological evaluations confirmed the absence of toxicity in major organs, reinforcing the safety profile of this approach. These results position Api-NPs as a promising novel therapeutic strategy for obesity management, integrating immune modulation and localized drug delivery to address the complexities of obesity and its associated inflammatory processes.
Collapse
Affiliation(s)
- Neda Mohaghegh
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Anjali Iyer
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Ethan Wang
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Heemin Kang
- Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Mohsen Akbari
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA.
| | | |
Collapse
|
11
|
Hamari N, Blaak EE, Canfora EE. The impact of butyrate on glycemic control in animals and humans: a comprehensive semi-systemic review. Front Nutr 2025; 12:1603490. [PMID: 40557239 PMCID: PMC12185432 DOI: 10.3389/fnut.2025.1603490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/17/2025] [Accepted: 05/27/2025] [Indexed: 06/28/2025] Open
Abstract
The gut microbiome has been identified as a significant factor in host metabolism, playing a key role in the etiology of obesity, type 2 diabetes and cardiometabolic risk. Butyrate, produced by the gut microbiome from indigestible carbohydrates, has been shown to have beneficial effects on body weight control, inflammation, and insulin resistance, primarily evidenced by animal studies and in vitro experiments. However, translating these benefits to humans remains challenging due to variability in mode of butyrate administration or production upon fermentation of dietary fibers, as well as in butyrate absorption, and its metabolism. For instance, oral butyrate supplementation can directly increase circulating butyrate levels, thereby targeting peripheral tissues. In contrast, butyrate produced by the gut microbiome may also influence metabolism through local signaling mechanisms affecting peripheral tissues. Additionally, there may be large heterogeneity in the response of the individuals to butyrate interventions. Future research should aim to better understand butyrate kinetics and dynamics and its mechanisms in regulating intestinal and metabolic health. In human studies, longer-term, placebo-controlled trials are needed to establish the efficacy of either targeting butyrate production or supplementation in individuals with obesity and/or metabolic disturbances. Personalized dietary interventions based on individual microbiota composition and/or function and metabolic profiles may optimize butyrate production and its metabolic benefits. This could pave the way for effective butyrate-based interventions to improve metabolic health and prevent obesity-related complications.
Collapse
Affiliation(s)
| | | | - Emanuel E. Canfora
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center +, Maastricht, Netherlands
| |
Collapse
|
12
|
Troshina EA, Panevin TS, Briskman TD. [The role of obesity in the development and progression of osteoarthritis: the influence of medical and surgical therapies for obesity on the course of inflammatory arthritis: A review]. TERAPEVT ARKH 2025; 97:449-454. [PMID: 40561489 DOI: 10.26442/00403660.2025.05.203230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/15/2024] [Indexed: 06/28/2025]
Abstract
Obesity is considered the most important risk factor for the development of osteoarthritis (ОА) - progressive inflammatory disease of the joints, that is one of the causes of disability and long-term immobilization. Excessively developed adipose tissue not only increases the mechanical load on the joints, but also participates in the maintenance of chronic low-grade inflammation through the production of adipokines, cytokines, hemokines, complement factors and hormones. Adipokines influence cells of synovial tissue, cartilage and bone, which in turn produce some adipokines locally, maintaining an inflammatory microenvironment intraarticularly. Adipokines, including leptin, adiponectin, chemerin, and resistin, regulate inflammatory immune responses in cartilage, also affecting synovial tissue cells and bone. In turn, chondrocytes, osteoblasts and osteoclasts produce some adipokines locally, maintaining an inflammatory microenvironment intra-articularly. Weight loss in OA can improve the patient's quality of life, physical function, lead to reduce pain, and slow or halt the progression of structural degenerative changes. The purpose of this article is to clearly describe the pathogenetic ways between obesity and inflammation, to reveal the mechanisms of the pathological state of adipokines and proinflammatory mediators (IL-6, TNF-á, etc.) on cartilage and bone homeostasis and, as expected, to evaluate their participation in the development of OA. So understanding immune regulation and resolution of inflammation in obesity is critical to developing treatments approaches to OA for these patients. The article also analyzes current researches on the effect of drug therapy (liraglutide, orlistat, sibutramine) and bariatric surgery of obesity on the course of inflammatory joint diseases.
Collapse
Affiliation(s)
- E A Troshina
- National Medical Research Center for Endocrinology
| | - T S Panevin
- Nasonova Research Institute of Rheumatology
- Far Eastern State Medical University
| | - T D Briskman
- National Medical Research Center for Endocrinology
| |
Collapse
|
13
|
Wang Y, Wang Y, He X, Li X. Sodium-glucose transporter 2 inhibitors and cardiovascular-kidney-metabolic syndrome: a narrative review. Front Endocrinol (Lausanne) 2025; 16:1554637. [PMID: 40538814 PMCID: PMC12176599 DOI: 10.3389/fendo.2025.1554637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 05/19/2025] [Indexed: 06/22/2025] Open
Abstract
The Cardiovascular-Kidney-Metabolic (CKM) syndrome is a systemic disorder involving obesity, diabetes, chronic kidney disease (CKD), and cardiovascular disease, characterized by complex pathophysiological mechanisms that interact and lead to increased morbidity and mortality. In recent years, sodium-glucose transport protein 2 inhibitors (SGLT2i), as a new class of antidiabetic medications, have shown remarkable efficacy in the management of diabetes, renal and cardiovascular diseases. Research has confirmed their ability to reduce cardiovascular events and all-cause mortality. These inhibitors lower blood glucose levels by decreasing renal reabsorption of glucose and sodium, and offer multiple benefits, including lowering blood pressure, reducing body weight, exerting antioxidant, anti-inflammatory, and anti-fibrotic effects, as well as reducing proteinuria and improving glomerular filtration rate. These effects collectively contribute to the improvement of cardiovascular and renal health. Furthermore, SGLT2i have shown potential therapeutic roles at various stages of CKM syndrome, including improving cardiac function, slowing CKD progression, promoting weight loss, and improving lipid profiles. However, the precise mechanisms of action and off-target effects of SGLT2i still require further investigation to evaluate their efficacy and safety under different clinical conditions. Future research directions should include strategies for multiple disease management, combination therapy effects, interdisciplinary collaboration, and long-term follow-up studies to fully understand and optimize the application of SGLT2i in the treatment of CKM syndrome.
Collapse
Affiliation(s)
- Yuqing Wang
- Graduate School of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yaqing Wang
- Graduate School of Chengde Medical University, Chengde, Hebei, China
| | - Xiaojie He
- Graduate School of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaodong Li
- Department of Nephrology, Baoding No.1 Central Hospital of Hebei Medical University, Baoding, Hebei, China
| |
Collapse
|
14
|
Ponirakis G, Peerzada L, Petropoulos IN, Gad H, Abdulshakoor S, Concepcion JM, Khalfalla SH, Elamin ISA, AlZawqari ATH, Elgassim E, Baraka A, Mahfoud ZR, El Deeb MA, Afifi N, Malik RA. Differential Effects of Visceral and Subcutaneous Adiposity on Peripheral Neuropathy. J Peripher Nerv Syst 2025; 30:e70025. [PMID: 40345154 DOI: 10.1111/jns.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025]
Abstract
OBJECTIVE Obesity increases the risk of diabetic neuropathy. This study investigates the impact of visceral (VAT) and subcutaneous adipose tissue (SAT) volume on peripheral neuropathy. METHODS A total of 302 adults from the Qatar Biobank (QBB) underwent iDXA to measure VAT and SAT volumes, intima media thickness (IMT), and peripheral neuropathy assessments using corneal confocal microscopy (CCM), vibration perception threshold (VPT), and the DN4 questionnaire. RESULTS The QBB cohort was aged 43.9 ± 12.9 years, of whom 43.7% were women, 42.1% had obesity, 17.4% had type 2 diabetes (T2D) and 10.9% had hypertension. VAT was associated with T2D, hypertension, higher HbA1c, diastolic blood pressure, triglycerides, and inflammatory markers, and lower HDL (p < 0.0001). There were no significant associations between SAT and these cardiovascular risk factors. VAT volume was associated with lower corneal nerve inferior whorl length (IWL) (p < 0.05) and higher VPT (p = 0.01), partially mediated by elevated HbA1c (p < 0.05, p = 0.001) and IMT (p < 0.0001), while its association with neuropathic symptoms was fully mediated by systolic blood pressure (p < 0.05), T2D (p < 0.01), and triglycerides (p = 0.05). SAT showed no associations with measures of neuropathy. CONCLUSIONS VAT but not SAT is associated with peripheral neuropathy. This study underscores the need to target VAT to improve neuropathy.
Collapse
Affiliation(s)
- Georgios Ponirakis
- Department of Medicine, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Leeza Peerzada
- Department of Medicine, CUNY School of Medicine/Sophie Davis Biomedical Education, The City College of New York, New York, New York, USA
| | | | - Hoda Gad
- Department of Medicine, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | | | | | - Sara H Khalfalla
- Qatar Biobank, Qatar Foundation, Hamad Medical City, Doha, Qatar
| | - Iynas S A Elamin
- Qatar Biobank, Qatar Foundation, Hamad Medical City, Doha, Qatar
| | | | - Einas Elgassim
- Department of Medicine, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Areej Baraka
- Department of Medicine, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Ziyad R Mahfoud
- Department of Medicine, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Marwa A El Deeb
- Qatar Biobank, Qatar Foundation, Hamad Medical City, Doha, Qatar
| | - Nahla Afifi
- Qatar Biobank, Qatar Foundation, Hamad Medical City, Doha, Qatar
| | - Rayaz A Malik
- Department of Medicine, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| |
Collapse
|
15
|
Li Y, Zhang Z, Han Q, Liu G, Yin Y, Yin J. Lactobacillus johnsonii-derived leucic acid promotes fatty acid absorption and deposition by targeting CD36. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1727-1739. [PMID: 40120026 DOI: 10.1007/s11427-024-2794-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/25/2024] [Indexed: 03/25/2025]
Abstract
Lactobacillus johnsonii is a microbial biomarker associated with lipid deposition, but the mechanism by which it accelerates fatty acid absorption and deposition remains unclear. In this study, we isolated a strain of L. johnsonii MS0621 from the feces of Ningxiang pigs, an obese animal model, and evaluated its probiotic properties with high resistance to temperature and intestinal fluids. Colonization by L. johnsonii MS0621 increased the abundance of gut Lactobacillus in lean DLY pigs, concomitant with increases in fatty acid absorption in the intestine and lipid depositions in the fat and muscle tissues. The lipid absorption-promoting effect was further detected in IPEC-J2 cells treated with live L. johnsonii MS0621 and the bacteria-free supernatants, as evidenced by high triglyceride synthesis and the expression of CD36, a key lipid transporter. Metabolomics analysis showed that (R)-leucic acid is a potential metabolite targeting CD36 expression to guarantee lipid absorption and deposition. The mechanism might involve direct interaction with CD36, as molecular docking and inhibition of CD36 blocked L. johnsonii MS0621 or derived metabolite-mediated lipid absorption. In conclusion, we uncovered an important role of L. johnsonii MS0621 derived (R)-leucic acid in regulating the absorption and deposition of intestinal fatty acids via regulation of CD36 expression.
Collapse
Affiliation(s)
- Yunxia Li
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, 410128, China
- Yuelushan Laboratory, Changsha, 410128, China
| | - Zhiming Zhang
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, 410128, China
- Yuelushan Laboratory, Changsha, 410128, China
| | - Qi Han
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, 410128, China
- Yuelushan Laboratory, Changsha, 410128, China
| | - Gang Liu
- Yuelushan Laboratory, Changsha, 410128, China
- College of Bioscience and Biotechnology, Hunan Agriculture University, Changsha, 410128, China
| | - Yulong Yin
- Yuelushan Laboratory, Changsha, 410128, China
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, 410128, China.
- Yuelushan Laboratory, Changsha, 410128, China.
| |
Collapse
|
16
|
Wu L, Coletta DK. Obesity and type 2 diabetes mellitus: insights from skeletal muscle extracellular matrix remodeling. Am J Physiol Cell Physiol 2025; 328:C1752-C1763. [PMID: 40244268 DOI: 10.1152/ajpcell.00154.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/23/2024] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are metabolic diseases at epidemic proportions. The economic burden for these diseases is at an all-time high, and as such, there is an urgent need for advancements in identifying targets for treating these complex disorders. The extracellular matrix (ECM), comprising collagen, fibronectin, laminin, elastin, and proteoglycan, surrounds skeletal muscles and plays a critical role in maintaining tissue homeostasis by providing structural support and facilitating cell-to-cell communication. Disruption of the ECM signaling results in changes to its micro/macroenvironment, thereby modifying tissue homeostasis. Skeletal muscle ECM remodeling has been shown to be associated with insulin resistance, an underlying feature of obesity and T2DM. This narrative review explores the critical components of skeletal muscle ECM and its accumulation and remodeling in metabolic diseases. In addition, we discuss potential treatments to mitigate the effects of ECM remodeling in skeletal muscle. We conclude that targeting ECM remodeling in skeletal muscle represents a promising yet underexplored therapeutic avenue in the management of metabolic disorders.
Collapse
Affiliation(s)
- Linda Wu
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
| | - Dawn K Coletta
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, Arizona, United States
- Center for Disparities in Diabetes, Obesity, and Metabolism, University of Arizona, Tucson, Arizona, United States
| |
Collapse
|
17
|
Zong J, Wu X, Huang X, Yuan L, Yuan K, Zhang Z, Jiang M, Ping Z, Cheong LY, Xu A, Hoo RLC. Adipocyte-derived shed Syndecan-4 suppresses lipolysis contributing to impaired adipose tissue browning and adaptive thermogenesis. Mol Metab 2025; 96:102133. [PMID: 40180176 PMCID: PMC12004711 DOI: 10.1016/j.molmet.2025.102133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025] Open
Abstract
Lipolysis in white adipose tissue (WAT) provides fatty acids as energy substrates for thermogenesis to increase energy expenditure. Syndecan-4 (Sdc4) is a transmembrane proteoglycan bearing heparan sulfate chains. Although single nucleotide polymorphisms (SNPs) of the Sdc4 gene have been identified linking to metabolic syndromes, its specific function in adipose tissue remains obscure. Here, we show that Sdc4 serves as a regulator of lipid metabolism and adaptive thermogenesis. Sdc4 expression and shedding are elevated in the white adipose tissue (WAT) of diet-induced obese mice. Adipocyte-specific deletion of Sdc4 promotes lipolysis and WAT browning, thereby raising whole-body energy expenditure to protect against diet-induced obesity. Mechanistically, fibroblast growth factor 2 (FGF2) is a paracrine factor that maintains energy homeostasis. Elevated shed Sdc4 concentrates and delivers FGF2 to fibroblast growth factor receptor 1 (FGFR1) on adipocytes, which in turn suppresses lipolysis by reducing hormone-sensitive lipase (HSL) activity, thus exaggerating adipose tissue dysfunction upon high-fat diet induction. Sdc4-deficient adipocytes show higher lipolytic and thermogenic capacity by enhancing HSL phosphorylation and UCP1 expression. Overall, our study reveals that adipocyte-derived shed Sdc4 is a novel suppressor of lipolysis, contributing to decreased energy expenditure, thus exaggerating obesity. Targeting shed Sdc4 is a potential therapeutic strategy for obesity.
Collapse
Affiliation(s)
- Jiuyu Zong
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiaoping Wu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiaowen Huang
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Lufengzi Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kai Yuan
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zixuan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Mengxue Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Zhihui Ping
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Lai Yee Cheong
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ruby Lai Chong Hoo
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
18
|
Jeong H, Hwang US, Choi H, Park YS. Assessing the Anti-Obesity Potential of Lactococcus lactis subsp. lactis CAB701: Modulation of Adipocyte Differentiation and Lipid Metabolism in In Vitro and In Vivo Models. Probiotics Antimicrob Proteins 2025; 17:1203-1216. [PMID: 38064160 DOI: 10.1007/s12602-023-10198-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 05/07/2025]
Abstract
In this study, the potential anti-obesity effects of Lactococcus lactis subsp. lactis CAB701, a probiotic strain isolated from cabbage, were investigated using in vitro and in vivo assays. L. lactis subsp. lactis CAB701 inhibited adipocyte differentiation of 3T3-L1 cells by 67%. In an in vivo model of high-fat diet-induced obese mice, treatment with L. lactis subsp. lactis CAB701 markedly reduced body weight and peri-epididymal fat mass, and significantly reduced serum total cholesterol, triglycerides, and low-density lipoprotein cholesterol levels. Molecular analysis revealed a significant modulation of key genes and proteins involved in lipid metabolism and adipogenesis. Specifically, fatty acid synthase and peroxisome proliferator-activated receptor gamma were significantly downregulated in peri-epididymal adipose tissue, alluding to the molecular mechanism underlying the anti-obesity effects exerted by L. lactis subsp. lactis CAB701. Furthermore, histological examination revealed a significant reduction in adipocyte size in the treated group, indicating effective adipose tissue remodeling. Our findings suggest that L. lactis subsp. lactis CAB701 mediates anti-obesity effects through the modulation of critical molecular markers and lipid profiles. L. lactis subsp. lactis CAB701 thus represents a promising candidate for combating obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Huijin Jeong
- Department of Food Science and Biotechnology, Gachon University, Seongnam, 13120, Republic of Korea
| | | | | | - Young-Seo Park
- Department of Food Science and Biotechnology, Gachon University, Seongnam, 13120, Republic of Korea.
| |
Collapse
|
19
|
Guglielmi V, Colangeli L, Parrotta ME, Ciammariconi A, Milani I, D'Adamo M, Sbraccia P, Capoccia D. Social isolation and loneliness in non-communicable chronic diseases: Impact of COVID-19 pandemic, population aging and technological progress. Nutr Metab Cardiovasc Dis 2025; 35:104015. [PMID: 40189996 DOI: 10.1016/j.numecd.2025.104015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/29/2025]
Abstract
AIMS Social isolation and loneliness have increasingly emerged as closely linked to onset and progression of non-communicable chronic diseases (NCDs). The aim of this review is to highlight the importance of addressing social isolation in the prevention and management of NCDs such as obesity, type 2 diabetes, and cardiovascular diseases in order to hinder their development and improve their outcomes. DATA SYNTHESIS Social isolation and loneliness affect a significant portion of the older adult population, due to decrease in social interactions, chronic illnesses and sensory impairments. However, many other vulnerable populations may experience social isolation because of psychiatric or disabling health conditions, substances abuse, low socioeconomic status, unemployment and belonging to ethnic or marginalized minorities. The unprecedented COVID-19-related social distancing can be taken as a proof-of-concept of the detrimental role of poor interactions in NCDs prevention and management not only at individual level but also in a public health perspective. Indeed, social isolation has been linked to unhealthy lifestyle choices, disrupted sleep quality, low utilization of healthcare, preventive services and adherence to treatments. Underlying mechanisms like inflammation and stress responses may also play a role in the association between social isolation and worse NCDs outcomes. CONCLUSIONS Social isolation negatively impacts on the development, progression and management of NCDs. Effective interventions for social isolation should address both societal factors and healthcare-related needs. To counteract the detrimental effects of social distancing during COVID-19 pandemic, the use of telemedicine was implemented. However, telemedicine is not always available, and legislative and age-related barriers persist.
Collapse
Affiliation(s)
- Valeria Guglielmi
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy.
| | - Luca Colangeli
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Maria Eugenia Parrotta
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Azzurra Ciammariconi
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Ilaria Milani
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Monica D'Adamo
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Danila Capoccia
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
20
|
Sriboonaied P, Phuangbubpha P, Saetan P, Charoensuksai P, Charoenpanich A. Dual Modulation of Adipogenesis and Apoptosis by PPARG Agonist Rosiglitazone and Antagonist Betulinic Acid in 3T3-L1 Cells. Biomedicines 2025; 13:1340. [PMID: 40564064 PMCID: PMC12190099 DOI: 10.3390/biomedicines13061340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Revised: 05/27/2025] [Accepted: 05/28/2025] [Indexed: 06/28/2025] Open
Abstract
Background/Objectives: Disruptions in adipose tissue dynamics contribute to obesity-related metabolic disorders, emphasizing the need for targeted therapies focusing on adipose tissue cells, including progenitor cells and adipocytes. Peroxisome proliferator-activated receptor gamma (PPARG) ligands are potent insulin sensitizers used in type 2 diabetes treatment. This study investigated the effects of rosiglitazone, a PPARG agonist, and betulinic acid, a PPARG antagonist, on adipogenesis and apoptosis in 3T3-L1 pre-adipocytes. Method: 3T3-L1 pre-adipocytes were treated with rosiglitazone or betulinic acid during adipogenic differentiation. Lipid droplet formation was used to evaluate adipogenesis. Cell growth and cell death were assessed using the resazurin-based cell viability assay, trypan blue exclusion assay, LDH assay, and Annexin V/PI staining. Quantitative PCR was conducted to examine the expression of genes associated with adipogenesis and apoptosis. Results: Betulinic acid reduced adipogenesis only when administered daily for eight days. Rosiglitazone did not alter the overall lipid quantity; however, it promoted a shift toward fewer but larger lipid droplets. Both compounds increased Adipoq and Cfd expression, and betulinic acid also elevated Fabp4. Rosiglitazone induced stronger cell aggregation. Despite increased cell death, overall viability was maintained. Apoptotic cell death was enhanced by both compounds and confirmed via Annexin V/PI staining and flow cytometry, accompanied by downregulation of Ccnd1 and Bcl2. Additionally, rosiglitazone markedly increased the expression of Cebpa, a key regulator that can modulate lipid droplet formation and the balance between cell growth and death. Conclusions: Rosiglitazone and betulinic acid differentially modulate adipogenesis and apoptosis in 3T3-L1 cells, revealing a complex interplay between lipid accumulation and programmed cell death. Together, the findings underscore the potential of dual PPARG-targeting approaches for metabolic disease interventions.
Collapse
Affiliation(s)
- Patsawee Sriboonaied
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand; (P.S.); (P.P.); (P.S.)
| | - Pornwipa Phuangbubpha
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand; (P.S.); (P.P.); (P.S.)
| | - Puretat Saetan
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand; (P.S.); (P.P.); (P.S.)
| | - Purin Charoensuksai
- Department of Biomedicine and Health Informatics and Bioactives from Natural Resources Research Collaboration for Excellence in Pharmaceutical Sciences (BNEP), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand;
| | - Adisri Charoenpanich
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand; (P.S.); (P.P.); (P.S.)
| |
Collapse
|
21
|
Wang R, Xu Y, Zhang Y, Wu Y, Zuo A, Liu S, Ba Y, Xu H, Weng S, Zhou Z, Ma H, Luo P, Cheng Q, Han X, Liu Z. Total and regional fat-to-muscle mass ratio and risks of pan-cancer: a prospective cohort study. BMC Med 2025; 23:296. [PMID: 40437455 PMCID: PMC12121253 DOI: 10.1186/s12916-025-04102-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 04/25/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND The fat-to-muscle mass ratio (FMR) has served as a marker for various diseases. This study aimed to explore sex-specific associations between FMR in different body regions (whole body, trunk, arm, and leg) and cancer incidence. METHODS We included 435,986 cancer-free participants (203,133 men and 232,853 women) from the UK Biobank at baseline. FMR was calculated as the ratio of fat mass to muscle mass in each body region. Multivariable Cox proportional hazards models, along with Cox models incorporating restricted cubic splines (RCS) function, were employed to examine both linear and non-linear associations between FMR and cancer risk in men and women. Additionally, a combined grouping of body mass index (BMI) and FMR was used to assess the joint impact of body composition on cancer incidence. RESULTS During the follow-up period, 62,060 new cancer cases were recorded. Our analysis showed significant associations between both total and regional FMR and the risk of several cancers. In men, higher whole body FMR was associated with an increased risk of esophagus, stomach, colorectal, liver, pancreas, and kidney cancers, while a decreased risk was observed for prostate and non-melanoma skin cancers (FDR < 0.05). In women, higher FMR was associated with a higher incidence of gallbladder, pancreas, kidney, thyroid, breast, and uterus cancers (FDR < 0.05). Non-linear associations were observed for several cancer types, with specific FMR cut-off points presented using RCS curves. The analysis by combining BMI and FMR suggested potential interaction patterns, revealing some masked risks; for example, a significant increase in cancer incidence was also observed in individuals exhibiting high FMRs despite having low BMI. CONCLUSIONS Our findings suggested that both total and regional FMR may serve as potential biomarkers for assessing the risk of overall and site-specific cancers.
Collapse
Affiliation(s)
- Ruizhi Wang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yushuai Wu
- Shanghai Academy of Artificial Intelligence for Science, Shanghai, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Henan, 450052, China
| | - Hongxuan Ma
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
22
|
Guzmán-Herrera N, Ruíz-Madrigal B, Parés-Hipólito J, Salazar-Olivo LA. SERPINA3 is expressed in human adipocytes and modulated by TNF-α and vitamin B6. In Vitro Cell Dev Biol Anim 2025:10.1007/s11626-025-01053-y. [PMID: 40425899 DOI: 10.1007/s11626-025-01053-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 04/10/2025] [Indexed: 05/29/2025]
Abstract
SERPINA3G participates in the antiadipogenesis and insulin resistance induced by TNF-α in 3T3-F442A murine cells. Here, we show that the human orthologue SERPINA3 is expressed in human subcutaneous and visceral adipose depots of normal-weight individuals and that TNF-α and RA induced the overexpression of SERPINA3 mRNA in cultured human subcutaneous and visceral adipocytes, although only TNF-α induced the expression of serpin A3 protein. We also demonstrate that vitamin B6 abrogated the expression of the SERPINA3 gene and diminished the anti-adipogenic effects of TNF-α on mature adipocytes. Our results indicate that SERPINA3 is expressed in human adipose tissues and modulates the antiadipogenic effects of TNF-α, and suggest serpin A3 could be a promissory target in the inflammatory processes linked to obesity and other adipose dysfunctions.
Collapse
Affiliation(s)
- Nataly Guzmán-Herrera
- Molecular Biology Division, Instituto Potosino de Investigación Científica y Tecnológica, Camino a La Presa San José 2055, San Luis Potosí, 78216, México
| | - Bertha Ruíz-Madrigal
- Microbiology Research Laboratory, Department of Microbiology and Pathology, University Center for Health Sciences, University of Guadalajara, Sierra Mojada 950, Guadalajara, 44340, Jalisco, México
| | - Jaime Parés-Hipólito
- Health Sciences Division, Universidad Autónoma del Estado de Quintana Roo, Blvd. Bahía S/N del Bosque, Quintana Roo, 77019, Chetumal, México
- Present Address: Hospital Militar de Especialidades Oftalmológicas, Alcaldía Miguel Hidalgo, Av Constituyentes 240, Col. Ampliación Daniel Garza, CP, 11830, Mexico City, México
| | - Luis A Salazar-Olivo
- Molecular Biology Division, Instituto Potosino de Investigación Científica y Tecnológica, Camino a La Presa San José 2055, San Luis Potosí, 78216, México.
| |
Collapse
|
23
|
Terreros G, Munoz F, Magdalena M, Soto-Donoso M, Torres N, D’Espessailles A. Adipose Tissue Dysfunction Induced by High-Fat Diet Consumption Is Associated with Higher Otoacoustic Emissions Threshold in Mice C57BL/6. Nutrients 2025; 17:1786. [PMID: 40507055 PMCID: PMC12156954 DOI: 10.3390/nu17111786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Revised: 05/19/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025] Open
Abstract
Background/Objectives: Obesity is a risk factor for several diseases; however, less has been researched about how diet-induced obesity may affect the auditory system. In this sense, the purpose of this study was to evaluate the effect of diet-induced obesity on the functionality and integrity of the outer hair cells, a key component of the organ of Corti, inside the cochlea. Furthermore, we hypothesized that adipose tissue (AT) status is associated with impaired outer hair cell auditory amplification in young C57BL/6 mice, contributing to increased vulnerability to hearing damage. Methods: Weaning male C57BL/6J mice (7 weeks old) weighing 22-23 g were divided into two diet groups: (i) a control diet or (ii) a high-fat diet (HFD) for 12 or 16 weeks. Metabolic parameters (body and AT weight, glucose tolerance test), AT dysfunction markers (AT remodeling, adipocyte size, crown-like structures), and outer hair cell function (distortion products otoacoustic emissions (DPOAEs) threshold and amplitudes) and integrity (hair cells cell count) were evaluated. Results: We observed that mice fed an HFD for 16 weeks showed a higher DPOAE threshold against stimuli at 16 KHz and a lower count of outer hair cells in the medial section of the cochlea. These results demonstrate a correlation between body and AT weight specifically at 16 weeks of treatment, the time point at which we observed a marked AT dysfunction. Conclusions: Taken together, our results suggest that obese mice with AT dysfunction have an altered auditory efferent system, characterized by a higher DPOAE threshold and a lower outer hair cell count in the medial section, which may impact signal transduction.
Collapse
Affiliation(s)
- Gonzalo Terreros
- Laboratorio de Neurociencia Sensorial, Perceptual y Cognitiva, Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile; (G.T.); (F.M.); (M.S.-D.)
| | - Felipe Munoz
- Laboratorio de Neurociencia Sensorial, Perceptual y Cognitiva, Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile; (G.T.); (F.M.); (M.S.-D.)
| | - Matías Magdalena
- Laboratorio de Neurociencia Sensorial, Perceptual y Cognitiva, Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile; (G.T.); (F.M.); (M.S.-D.)
- Escuela de Salud, Universidad de O’Higgins, Rancagua 2820000, Chile
| | - Manuel Soto-Donoso
- Laboratorio de Neurociencia Sensorial, Perceptual y Cognitiva, Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile; (G.T.); (F.M.); (M.S.-D.)
- Escuela de Salud, Universidad de O’Higgins, Rancagua 2820000, Chile
| | - Nairo Torres
- Doctorado en Ciencias de la Bioingeniería, Universidad de O’Higgins, Rancagua 2820000, Chile
| | - Amanda D’Espessailles
- Laboratorio de Neurociencia Sensorial, Perceptual y Cognitiva, Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile; (G.T.); (F.M.); (M.S.-D.)
| |
Collapse
|
24
|
Juguilon C, Khosravi R, Radisic M, Wu JC. In Vitro Modeling of Interorgan Crosstalk: Multi-Organ-on-a-Chip for Studying Cardiovascular-Kidney-Metabolic Syndrome. Circ Res 2025; 136:1476-1493. [PMID: 40403116 PMCID: PMC12180411 DOI: 10.1161/circresaha.125.325497] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/24/2025]
Abstract
Cardiovascular-kidney-metabolic syndrome is a progressive disorder driven by perturbed interorgan crosstalk among adipose, liver, kidney, and heart, leading to multiorgan dysfunction. Capturing the complexity of human cardiovascular-kidney-metabolic syndrome pathophysiology using conventional models has been challenging. Multi-organ-on-a-chip platforms offer a versatile means to study underlying interorgan signaling at different stages of cardiovascular-kidney-metabolic syndrome and bolster clinical translation.
Collapse
Affiliation(s)
- Cody Juguilon
- Stanford Cardiovascular Institute (C.J., J.C.W.), Stanford University, Stanford, CA
- Division of Cardiovascular Medicine, Department of Medicine (C.J., J.C.W.), Stanford University, Stanford, CA
| | - Ramak Khosravi
- Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (R.K., M.R.)
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC (R.K.)
| | - Milica Radisic
- Stanford Cardiovascular Institute (C.J., J.C.W.), Stanford University, Stanford, CA
- Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (R.K., M.R.)
- Institute of Biomedical Engineering (M.R.)
- University of Toronto, Ontario, Canada (M.R.)
| | - Joseph C Wu
- Stanford Cardiovascular Institute (C.J., J.C.W.), Stanford University, Stanford, CA
- Division of Cardiovascular Medicine, Department of Medicine (C.J., J.C.W.), Stanford University, Stanford, CA
- Greenstone Biosciences, Palo Alto, CA (J.C.W.)
| |
Collapse
|
25
|
Hade MD, Butsch BL, Loreto Palacio P, Nguyen KT, Shantaram D, Noria SF, Brethauer SA, Needleman BJ, Hsueh W, Reátegui E, Magaña SM. Human Differentiated Adipocytes as Surrogate Mature Adipocytes for Adipocyte-Derived Extracellular Vesicle Analysis. Cells 2025; 14:757. [PMID: 40497934 PMCID: PMC12153563 DOI: 10.3390/cells14110757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/08/2025] [Accepted: 05/16/2025] [Indexed: 06/18/2025] Open
Abstract
Obesity is a growing global health concern, contributing to diseases such as cancer, autoimmune disorders, and neurodegenerative conditions. Adipose tissue dysfunction, characterized by abnormal adipokine secretion and chronic inflammation, plays a key role in these conditions. Adipose-derived extracellular vesicles (ADEVs) have emerged as critical mediators in obesity-related diseases. However, the study of mature adipocyte-derived EVs (mAdipo-EVs) is limited due to the short lifespan of mature adipocytes in culture, low EV yields, and the low abundance of these EV subpopulations in the circulation. Additionally, most studies rely on rodent models, which have differences in adipose tissue biology compared to humans. To overcome these challenges, we developed a standardized approach for differentiating human preadipocytes (preAdipos) into mature differentiated adipocytes (difAdipos), which produce high-yield, human adipocyte EVs (Adipo-EVs). Using visceral adipose tissue from bariatric surgical patients, we isolated the stromal vascular fraction (SVF) and differentiated preAdipos into difAdipos. Brightfield microscopy revealed that difAdipos exhibited morphological characteristics comparable to mature adipocytes (mAdipos) directly isolated from visceral adipose tissue, confirming their structural similarity. Additionally, qPCR analysis demonstrated decreased preadipocyte markers and increased mature adipocyte markers, further validating successful differentiation. Functionally, difAdipos exhibited lipolytic activity comparable to mAdipos, supporting their functional resemblance to native adipocytes. We then isolated preAdipo-EVs and difAdipo-EVs using tangential flow filtration and characterized them using bulk and single EV analysis. DifAdipo-EVs displayed classical EV and adipocyte-specific markers, with significant differences in biomarker expression compared to preAdipo-EVs. These findings demonstrate that difAdipos serve as a reliable surrogate for mature adipocytes, offering a consistent and scalable source of adipocyte-derived EVs for studying obesity and its associated disorders.
Collapse
Affiliation(s)
- Mangesh Dattu Hade
- Department of Pediatrics, Center for Clinical and Translational Research, Nationwide Children’s Hospital, Columbus, OH 43202, USA; (M.D.H.); (B.L.B.); (P.L.P.)
| | - Bradley L. Butsch
- Department of Pediatrics, Center for Clinical and Translational Research, Nationwide Children’s Hospital, Columbus, OH 43202, USA; (M.D.H.); (B.L.B.); (P.L.P.)
| | - Paola Loreto Palacio
- Department of Pediatrics, Center for Clinical and Translational Research, Nationwide Children’s Hospital, Columbus, OH 43202, USA; (M.D.H.); (B.L.B.); (P.L.P.)
| | - Kim Truc Nguyen
- Department of Chemical and Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; (K.T.N.); (E.R.)
| | - Dharti Shantaram
- Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH 43210, USA; (D.S.); (W.H.)
| | - Sabrena F. Noria
- Center for Minimally Invasive Surgery, Department of Surgery, Division of General and GI Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (S.F.N.); (S.A.B.); (B.J.N.)
| | - Stacy A. Brethauer
- Center for Minimally Invasive Surgery, Department of Surgery, Division of General and GI Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (S.F.N.); (S.A.B.); (B.J.N.)
| | - Bradley J. Needleman
- Center for Minimally Invasive Surgery, Department of Surgery, Division of General and GI Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (S.F.N.); (S.A.B.); (B.J.N.)
| | - Willa Hsueh
- Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH 43210, USA; (D.S.); (W.H.)
| | - Eduardo Reátegui
- Department of Chemical and Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; (K.T.N.); (E.R.)
| | - Setty M. Magaña
- Department of Pediatrics, Center for Clinical and Translational Research, Nationwide Children’s Hospital, Columbus, OH 43202, USA; (M.D.H.); (B.L.B.); (P.L.P.)
| |
Collapse
|
26
|
Iqbal M, Yu Q, Tang J, Xiang J. Unraveling the gut microbiota's role in obesity: key metabolites, microbial species, and therapeutic insights. J Bacteriol 2025; 207:e0047924. [PMID: 40183584 PMCID: PMC12096833 DOI: 10.1128/jb.00479-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Obesity, characterized by excessive fat accumulation, stems from an imbalance between energy intake and expenditure, with the gut microbiota playing a crucial role. This review highlights how gut microbiota influences metabolic pathways, inflammation, and adipose tissue regulation in obesity. Specific bacteria and metabolites, such as lipopolysaccharides (LPS) and short-chain fatty acids (SCFAs), modulate gut permeability, inflammation, and energy harvest, impacting obesity development. Certain gut bacteria, including Clostridium XIVb, Dorea spp., Enterobacter cloacae, and Collinsella aerofaciens, promote obesity by increasing energy harvest, gut permeability, and inflammatory response through LPS translocation into the bloodstream. Conversely, beneficial bacteria like Akkermansia muciniphila, Lactobacillus spp., and Bifidobacterium spp. enhance gut barrier integrity, regulate SCFA production, and modulate fasting-induced adipose factor, which collectively support metabolic health by reducing fat storage and inflammation. Metabolites such as SCFAs (acetate, propionate, and butyrate) interact with G-protein coupled receptors to regulate lipid metabolism and promote the browning of white adipose tissue (WAT), thus enhancing thermogenesis and energy expenditure. However, LPS contributes to insulin resistance and fat accumulation, highlighting the dual roles of these microbial metabolites in both supporting and disrupting metabolic function. Therapeutic interventions targeting gut microbiota, such as promoting WAT browning and activating brown adipose tissue (BAT), hold promise for obesity management. However, personalized approaches are necessary due to individual microbiome variability. Further research is essential to translate these insights into microbiota-based clinical therapies.
Collapse
Affiliation(s)
- Majid Iqbal
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Yu
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingqun Tang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juanjuan Xiang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
27
|
Cheng J, Xu D, Zhang D, Huang K, Zhang Y, Li X, Zhao Y, Zhao L, Xu Q, Yang X, Ma Z, Tian H, Zhang X, Wang W. Exploring the cecal microbial community associated with fat deposition in sheep and its possible pathways of action. Microbiol Spectr 2025:e0148824. [PMID: 40401954 DOI: 10.1128/spectrum.01488-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 04/14/2025] [Indexed: 05/23/2025] Open
Abstract
Fat deposition is a crucial economic trait during sheep growth and development, closely linked to economic returns. Current research on sheep's digestive tract predominantly focuses on the rumen, but the composition of the cecal microbiota and its relationship with host fat deposition remains largely unexplored. In this study, we sequenced the cecal microbiota of 60 Hu sheep, exhibiting marked differences in traits. The most abundant species in the sheep cecum were Firmicutes and Bacteroidota. Statistical analyses revealed significant differences in microbial community structures among different fat-deposition groups (P < 0.05). Using a random forest regression model and linear regression, 15 microbial biomarkers, including Lachnospiraceae_NK3A20_group, Turicibacter, and Bacteroides, were identified as key contributors to fat deposition. Additionally, volatile fatty acids (VFAs) in the cecum and biochemical indices in serum were measured. Acetic acid was the most abundant VFA in the cecum, while isobutyric acid levels were significantly higher in the low-fat group than in other groups (P < 0.05). Serum triglyceride (TG) levels were significantly higher in the high-fat group (P < 0.05). Correlation analysis revealed a significant association between Lachnospiraceae_NK3A20_group and acetic acid levels, as well as between TG levels and fat deposition traits (P < 0.05). TG levels were negatively correlated with acetic acid concentrations (P < 0.05). These findings suggest that the cecal microbiota influences fat deposition in sheep, potentially via the VFAs-TG metabolic pathway. IMPORTANCE Compared with muscle development, fat deposition consumes more energy, and controlling the fat deposition process can effectively reduce energy waste. Current research on rumination mainly focuses on the rumen but lacks research on the hindgut. This study identifies differences in the cecal microbiota of sheep with varying fat deposition levels and highlights significant correlations between specific microorganisms, cecal metabolites, and host traits. Therefore, the regulation of cecal microorganisms can help improve the fat deposition characteristics of sheep.
Collapse
Affiliation(s)
- Jiangbo Cheng
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Dan Xu
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Deyin Zhang
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Kai Huang
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Yukun Zhang
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaolong Li
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Yuan Zhao
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Liming Zhao
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Quanzhong Xu
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaobin Yang
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Zongwu Ma
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Huibin Tian
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoxue Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Weimin Wang
- The State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
28
|
Peng H, Cheng Q, Chen J, Wang Y, Du M, Lin X, Zhao Q, Chen S, Zhang J, Wang X. Green Tea Epigallocatechin-3-gallate Ameliorates Lipid Accumulation and Obesity-Associated Metabolic Syndrome via Regulating Autophagy and Lipolysis in Preadipocytes and Adipose Tissue. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12272-12291. [PMID: 40347183 DOI: 10.1021/acs.jafc.5c00973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2025]
Abstract
Previous studies have shown that epigallocatechin-3-gallate (EGCG), the most abundant catechin in green tea, demonstrates promising antiobesity effects. While autophagy mediates obesity via preadipocyte differentiation and lipogenesis, EGCG's potential autophagy-dependent antiobesity mechanism remains unclear. We used 3T3-L1 cells and high-fat-diet (HFD)-fed mice to examine how EGCG inhibits adipogenesis and lipogenesis via autophagy. EGCG (50 or 100 mg/kg) significantly attenuated HFD-induced weight gain, fat accumulation, hyperlipidemia, and glucose intolerance in mice. It also enhanced autophagy and lipolysis in white adipose tissue (WAT). EGCG profoundly inhibited terminal preadipocyte differentiation and lipid droplet formation in 3T3-L1 cells accompanied by reduced PPARγ, C/EBPα, and FASN expressions. Mechanistically, EGCG inhibited autophagy during the early stage of preadipocyte differentiation, as evidenced by increased autophagosome accumulation and impaired autophagic flux. The antiadipogenic effect of EGCG was further aggravated by the autophagy inhibitor chloroquine. Meanwhile, EGCG increased p38 and AMPK/ACC phosphorylation while inhibiting JNK phosphorylation in 3T3-L1 cells at an early stage of adipogenesis. Interestingly, EGCG reduced the expression of lipolytic enzymes HSL and ATGL, and it decreased glycerol contents in differentiated 3T3-L1 cells. These findings provide novel insights into the mechanism of using green tea EGCG in functional foods to combat obesity by targeting autophagy and lipolysis.
Collapse
Affiliation(s)
- He Peng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Qi Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Jiajun Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Ying Wang
- College of Food and Health, Zhejiang A&F University, Hangzhou 311300, P. R. China
| | - Menghao Du
- Research Institute of Subtropical Forestry, Chinese Academy of Forestry, Hangzhou 311400, P. R. China
| | - Xiaojian Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Qian Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Shengjia Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Jingsa Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Xingya Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
- School of Pharmaceutical Sciences & Institute of Advanced Studies, Taizhou University, Taizhou 318000, P. R. China
| |
Collapse
|
29
|
Bhattacharjee P, Iyer MR. Therapeutic Potential of Vascular Adhesion Protein-1 (VAP-1)/Semicarbazide-Sensitive Amine Oxidase (SSAO) Inhibitors: Current Medicinal Chemistry and Emerging Opportunities. Med Res Rev 2025. [PMID: 40396328 DOI: 10.1002/med.22118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 03/14/2025] [Accepted: 04/27/2025] [Indexed: 05/22/2025]
Abstract
Semicarbazide-sensitive amine oxidase (SSAO) is a vascular enzyme and expressed in high concentrations in vascular smooth muscle cells (VSMCs), localized in the caveolae of the plasma membrane, and the endothelial cells. SSAO is classified as a copper amine oxidase and encoded by the amine oxidase copper-containing 3 gene. SSAO exists both as a soluble protein and as a tissue-bound transmembrane protein. The latter is often called vascular adhesion protein 1. Vascular adhesion protein-1 or VAP-1, encoded by the AOC3 gene, is a pro-inflammatory and multifunctional molecule belonging to the SSAO family. It assists the transformation of primary amines to aldehydes resulting in the production of hydrogen peroxide and ammonia. Work from the last two decades, has shown that VAP-1/SSAO plays a role in several physiological and pathological processes, making it a potentially valuable target for therapeutic development. In this review, we provide a detailed overview of the inhibitors of VAP-1/SSAO that are being developed specifically for the treatment of inflammatory diseases. Here in we have highlighted important aspects of the compounds investigated in therapeutic applications. Furthermore, we have outlined potential avenues for innovation with the aim of maximizing the therapeutic efficacy of VAP-1/SSAO inhibitors in clinical settings.
Collapse
Affiliation(s)
- Pinaki Bhattacharjee
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, USA
| | - Malliga R Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
30
|
Pan W, Ji TF, Hu BT, Yang J, Lu L, Wei J. Association between triglyceride glucose body mass index and 1 year all cause mortality in stage 4 CKM syndrome patients. Sci Rep 2025; 15:17019. [PMID: 40379745 PMCID: PMC12084581 DOI: 10.1038/s41598-025-01549-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 05/07/2025] [Indexed: 05/19/2025] Open
Abstract
The triglyceride-glucose body mass index (TyG-BMI) is acknowledged as a dependable surrogate biomarker for the evaluation of insulin resistance (IR). Current research indicates a significant correlation between TyG-BMI and the risk of subsequent cardiovascular events in individuals diagnosed with cardiovascular-kidney-metabolic syndrome (CKM) at stages 0-3. Nevertheless, the prognostic significance of TyG-BMI in patients with CKM stage 4 has not been extensively investigated, and there is a paucity of evidence available on this topic. The study utilized patient data from the Medical Information Mart for Intensive Care (MIMIC-IV) database, categorizing the data into quartiles based on the TyG-BMI index. The primary outcomes of interest were all-cause mortality at 180 days and at one year. To assess the relationship between the TyG-BMI index and these outcomes in patients diagnosed with stage 4 CKM, a Cox proportional hazards model was employed. Additionally, a restricted cubic splines(RCS) model was applied to further investigate the associations between the TyG-BMI index and the specified outcomes. A total of 1,885 patients participated in the study, with 62.49% of the cohort being male. The all-cause mortality rates were recorded at 30.50% at 180 days and 35.12% at one year. Analysis using a multivariate Cox proportional hazards model revealed that an increase in the TyG-BMI index was significantly correlated with a reduction in the risk of all-cause mortality at both the 180-day and one-year marks. Specifically, for each standard deviation increase in the TyG-BMI index, the risk of all-cause mortality decreased by 17% within 180 days (HR = 0.83, 95% CI: 0.76-0.91) and by 21% within one year (HR = 0.79, 95% CI: 0.71-0.87). Furthermore, regression analysis utilizing RCS indicated a linear decrease in all-cause mortality rates associated with increasing TyG-BMI index values over both the 180-day and one-year periods (P for nonlinearity = 0.171 and P for nonlinearity = 0.141, respectively). In patients diagnosed with stage 4 CKM syndrome, a reduced TyG-BMI index was found to be significantly correlated with a heightened risk of all-cause mortality within both 180 days and one year. Consequently, the TyG-BMI index may be utilized as an effective instrument for risk stratification and prognostic assessment in this patient population.
Collapse
Affiliation(s)
- Wen Pan
- Department of Cardiovascular Internal Medicine, The Second Affiliated Hospital of Xi 'an Jiaotong University, Xi 'an, China
| | - Teng-Fei Ji
- Department of Cardiovascular Internal Medicine, The Second Affiliated Hospital of Xi 'an Jiaotong University, Xi 'an, China
| | - Bing-Tao Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Xi 'an Jiaotong University, Xi 'an, China
| | - Jing Yang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Xi 'an Jiaotong University, Xi 'an, China
| | - Lei Lu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Xi 'an Jiaotong University, Xi 'an, China
| | - Jin Wei
- Department of Cardiovascular Internal Medicine, The Second Affiliated Hospital of Xi 'an Jiaotong University, Xi 'an, China.
| |
Collapse
|
31
|
Lu G, Tian J, Shi F, Zhang DG, Wang D. Association of cardiovascular-kidney-metabolic syndrome stages with kidney stone prevalence: a population-based analysis of NHANES 2007-2020. BMJ Open 2025; 15:e096533. [PMID: 40379326 PMCID: PMC12086886 DOI: 10.1136/bmjopen-2024-096533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/22/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND The prevalence of kidney stones (KSs) has been increasing globally, and their association with cardiovascular disease and metabolic syndrome suggests a shared underlying pathophysiology. However, the impact of different stages of cardiovascular-kidney-metabolic (CKM) syndrome on KS prevalence remains unclear. OBJECTIVE This study aimed to investigate the association between the stages of CKM syndrome and the prevalence of KS in a nationally representative sample of adults in the USA. METHODS A total of 15 568 participants aged ≥20 years were included in the National Health and Nutrition Examination Survey 2007-2020 fasting subsample. CKM syndrome stages (0-4) were defined based on the 2023 American Heart Association Presidential Advisory on CKM Health. The KS history was determined using self-reported data. Multivariable logistic regression models were used to assess the association between the CKM syndrome stage and KS prevalence. RESULTS Of the 15 568 participants, 1501 (9.64%) reported a history of KS. The KS prevalence increased progressively with advancing CKM stage, rising from 5.10% in stage 0 to 16.55% in stage 4 (p<0.001). In the fully adjusted model, the ORs for KS were 1.18 (95% CI 0.83-1.68) for stage 1, 1.72 (95% CI 1.28 to 2.32) for stage 2, 2.00 (95% CI 1.29 to 3.10) for stage 3 and 2.36 (95% CI 1.64 to 3.40) for stage 4, compared with stage 0 (P for trend <0.001). Stratified analyses revealed no significant interactions between age, sex, race/ethnicity or other subgroups. CONCLUSION This study demonstrated a significant stepwise increase in KS prevalence with the advancing stages of CKM syndrome. These findings highlight the importance of monitoring and managing CKM syndromes to mitigate the risks of KS.
Collapse
Affiliation(s)
- Guoliang Lu
- Department of Urology, Ruijin Hospital, Shanghai, China
| | - Jinjun Tian
- Department of Urology, Shanghai Pudong New Area people's Hospital, Shanghai, China
| | - Feng Shi
- Civil Aviation Shanghai Hospital, Shanghai, China
| | - Ding-Guo Zhang
- Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Dawei Wang
- Department of Urology, Ruijin Hospital, Shanghai, China
| |
Collapse
|
32
|
Liu Y, Yu Z, Wang X, Yuan MQ, Lu MJ, Gong MR, Li Q, Xia YB, Yang GH, Xu B, Litscher G, Xu TC. Neurophysiological mechanisms of electroacupuncture in regulating pancreatic function and adipose tissue expansion. World J Diabetes 2025; 16. [DOI: doi:10.4239/wjd.v16.i5.101354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND
Electroacupuncture (EA) has been recognized for its beneficial effects on glucolipid metabolism, potentially through the regulation of sensory nerve coordination. The expandability of peripancreatic adipose tissue (PAT) is implicated in the transition from obesity to type 2 diabetes mellitus (T2DM). However, the specific pancreatic responses to EA require further elucidation.
AIM
To investigate the influence of EA on pancreatic glucolipid reduction level in a high-fat diet (HFD) rat model.
METHODS
To delineate the precise pathway through which EA mediates interactions between PAT and islets, we assessed the expression levels of NGF, TRPV1, insulin, as well as other proteins in the pancreas and PAT. This approach enabled us to identify the acupoints that are most conducive to optimizing glycolipid metabolism.
RESULTS
The ST25, LI11 and ST37 groups attenuated HFD-induced obesity and insulin resistance (IR) to distinct degrees, with ST25 group having the greatest effect. EA at ST25 was found to modify the local regulatory influence of PAT on the pancreatic intrinsic nervous system. Specifically, EA at ST25 obviously activated the TRPV1-CGRP-islet beta cell pathway, contributing to the relief of glucolipid metabolic stress. The beneficial effects were abrogated following the chemical silencing of TRPV1 sensory afferents, confirming their indispensable role in EA-mediated regulation of islet and PAT function. Furthermore, in TRPV1 knockout mice, a reduction in PAT inflammation was observed, along with the recovery of islet beta cell function. EA at LI11 and ST37 demonstrated anti-inflammatory properties and helped ameliorate IR.
CONCLUSION
The PAT ecological niche influenced the progression from obesity to T2DM through various immunometabolic pathways. EA at ST25 could regulate glucolipid metabolism via the TRPV1-CGRP-islet beta cell pathway.
Collapse
|
33
|
Liu Y, Yu Z, Wang X, Yuan MQ, Lu MJ, Gong MR, Li Q, Xia YB, Yang GH, Xu B, Litscher G, Xu TC. Neurophysiological mechanisms of electroacupuncture in regulating pancreatic function and adipose tissue expansion. World J Diabetes 2025; 16:101354. [DOI: 10.4239/wjd.v16.i5.101354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/12/2025] [Accepted: 03/14/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Electroacupuncture (EA) has been recognized for its beneficial effects on glucolipid metabolism, potentially through the regulation of sensory nerve coordination. The expandability of peripancreatic adipose tissue (PAT) is implicated in the transition from obesity to type 2 diabetes mellitus (T2DM). However, the specific pancreatic responses to EA require further elucidation.
AIM To investigate the influence of EA on pancreatic glucolipid reduction level in a high-fat diet (HFD) rat model.
METHODS To delineate the precise pathway through which EA mediates interactions between PAT and islets, we assessed the expression levels of NGF, TRPV1, insulin, as well as other proteins in the pancreas and PAT. This approach enabled us to identify the acupoints that are most conducive to optimizing glycolipid metabolism.
RESULTS The ST25, LI11 and ST37 groups attenuated HFD-induced obesity and insulin resistance (IR) to distinct degrees, with ST25 group having the greatest effect. EA at ST25 was found to modify the local regulatory influence of PAT on the pancreatic intrinsic nervous system. Specifically, EA at ST25 obviously activated the TRPV1-CGRP-islet beta cell pathway, contributing to the relief of glucolipid metabolic stress. The beneficial effects were abrogated following the chemical silencing of TRPV1 sensory afferents, confirming their indispensable role in EA-mediated regulation of islet and PAT function. Furthermore, in TRPV1 knockout mice, a reduction in PAT inflammation was observed, along with the recovery of islet beta cell function. EA at LI11 and ST37 demonstrated anti-inflammatory properties and helped ameliorate IR.
CONCLUSION The PAT ecological niche influenced the progression from obesity to T2DM through various immunometabolic pathways. EA at ST25 could regulate glucolipid metabolism via the TRPV1-CGRP-islet beta cell pathway.
Collapse
Affiliation(s)
- Yun Liu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Zhi Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Xuan Wang
- College of Traditional Chinese Medicine, Jiangsu Vocational College of Medicine, Yancheng 224000, Jiangsu Province, China
| | - Ming-Qian Yuan
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Meng-Jiang Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Mei-Rong Gong
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Qian Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - You-Bing Xia
- Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Guan-Hu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH 45701, United States
| | - Bin Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Gerhard Litscher
- High-Tech Acupuncture and Digital Chinese Medicine, Swiss University of Traditional Chinese Medicine, Bad Zurzach 5530, Switzerland
| | - Tian-Cheng Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| |
Collapse
|
34
|
Yu H, Guo B, Miao Z, Chen C, Song Y, Yang J. A high-fat diet suppresses growth hormone synthesis and secretion by influencing the Vit D receptor and Pit1. Endocrine 2025:10.1007/s12020-025-04270-3. [PMID: 40369297 DOI: 10.1007/s12020-025-04270-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND A long-term high-fat diet (HFD) leads to excessive lipid deposition, which may cause many diseases, including NAFLD, diabetes, and thyroid dysfunction. In addition, HFD leads to a decrease in serum growth hormone (GH) levels to further increase lipid deposition and obesity. However, the mechanism of such reduction of GH has not been fully elucidated. METHODS Male Sprague-Dawley rats were fed a regular diet (CD) or a high-fat diet (HFD) for 29 weeks. GH synthesis and secretion were evaluated in pituitary and blood samples, respectively. An in vitro model was constructed by treating cultured cells with palmitic acid (PA). Vit D receptor (VDR) plasmids (OE-VDR), paricalcitol and VDR knockdown virus (sh-VDR) were used to overexpress or depress the activation of VDR during PA treatment of GH3 cells. The GH content, lipid content, and relevant expression of different molecules were measured in pituitary and cell samples. RESULTS A HFD decreased the levels of circulating GH and the expression of Gh in the anterior pituitary gland tissues of rats. In vitro, PA treatment decreased Pit1 and Gh expression in cultured GH3 cells. VDR expression was reduced in the rat pituitary tissues under HFD conditions and in PA-treated GH3 cells. The overexpression and knockdown of VDR increased and decreased the expression of Pit1 and Gh, respectively. Paricalcitol antagonized the decrease in the expression of Pit1 and Gh caused by PA treatment. CONCLUSIONS HFD induced lipid deposition in the pituitary may cause GH deficiency, and VDR - Pit1 may be at least partially involved in the process.
Collapse
Affiliation(s)
- Huimin Yu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Key Laboratery of Endocine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Jinan, China
| | - Boning Guo
- Key Laboratery of Endocine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Jinan, China
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zhiwei Miao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Key Laboratery of Endocine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Jinan, China
| | - Chen Chen
- Endocrinology, SBMS, Faculty of Medicine, The University of Queensland, St Lucia, Qld, Australia
| | - Yongfeng Song
- Department of Endocrinology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Jianmei Yang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
35
|
Lu L, Chen Y, Liu B, Li X, Wang J, Nie Z, Fu X. Association between cumulative changes of the triglyceride glucose index and incidence of stroke in a population with cardiovascular-kidney-metabolic syndrome stage 0-3: a nationwide prospective cohort study. Cardiovasc Diabetol 2025; 24:202. [PMID: 40355933 PMCID: PMC12070779 DOI: 10.1186/s12933-025-02754-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND The triglyceride-glucose (TyG) index was associated with higher risk of mortality in individuals with Cardiovascular-Kidney-Metabolic (CKM) syndrome stages 0-3. However, the relationship between cumulative of TyG (cumTyG) and incidence of stroke remains unclear in individuals with CKM syndrome stages 0-3. METHOD Participants with CKM syndrome stage 0-3 were enrolled from the China Health and Retirement Longitudinal Study (CHARLS) from 2011 to 2015. TyG was calculated as ln [fasting triglyceride (mg/dL)×fasting glucose (mg/dL)/2], and the cumTyG, as an area-under-the-curve estimate (mean TyG × time span), was calculated as (TyG2012 + TyG 2015)/2 * time (2015-2012). TyG control levels were classified using k-mean clustering analysis. Logistic regression was used to analyze the effect of cumTyG and TyG control levels on the incidence of stroke. Restricted cubic spline models (RCS) were performed to explore the potential non-linear relationship between cumTyG and stroke risk at different CKM syndrome stages 0-3. RESULTS A total of 4,700 CKM syndrome stages 0-3 participants were enrolled, among 280 patients had developed stroke during the 3-year follow-up period. After adjusting for confounders, compared to class 1 group, the odds ratio (OR) of incidents of stroke for class 2 was 1.39 [95% confidence interval (CI) 1.003, 1.92], P = 0.046; the OR of incidents of stroke for class 3 was 1.28 (95% CI 0.92-1.77), P = 0.147, the OR of incidents of stroke for class 4 was 1.28 (95% CI 0.84-1.94), P = 0.238. Elevated cumTyG was associated with an increase in incidence of stroke (OR 1.13, 95% CI 1.05, 1.22, P = 0.002). The relationship between the cumTyG index and stroke was linear in restricted cubic spline regression. CONCLUSIONS Elevated cumTyG was associated with an increased risk of stroke events in the population of CKM syndrome stages 0-3. Long-term dynamic monitoring of changes of TyG may help in the early identification of patients at high risk of developing stroke in the individuals with CKM syndrome stages 0-3.
Collapse
Affiliation(s)
- Lifei Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Yubiao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
- Guangzhou National Laboratory, Guangzhou, People's Republic of China
| | - Baiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
- Guangzhou National Laboratory, Guangzhou, People's Republic of China
| | - Xicong Li
- 920th Hospital of Joint Logistics Support Force, PLA, Kunming, Yunnan, People's Republic of China
- Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China
| | - Jiale Wang
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital, Guangzhou Medical University, 1st Xinzao Rd, Xinzao, Panyu District, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Zhengchang Nie
- 920th Hospital of Joint Logistics Support Force, PLA, Kunming, Yunnan, People's Republic of China
- Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China
| | - Xiaodong Fu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital, Guangzhou Medical University, 1st Xinzao Rd, Xinzao, Panyu District, Guangzhou, 510260, Guangdong, People's Republic of China.
| |
Collapse
|
36
|
Lee CW, Yoon YS, Yoon YS, Chung KS, Kim MJ, Park G, Choi M, Jang YP, Lee KT. Protective Effects of a Standardized Water Extract from the Stem of Ipomoea batatas L. Against High-Fat Diet-Induced Obesity. Nutrients 2025; 17:1643. [PMID: 40431382 PMCID: PMC12113841 DOI: 10.3390/nu17101643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Obesity is a major health concern that can lead to various chronic diseases. Little is known about the anti-obesity effect of a standardized hot water extract from the stems of Ipomoea batatas (WIB). This study aimed to evaluate the therapeutic potential of WIB as a natural alternative to conventional anti-obesity treatments by assessing its effects on body weight, fat accumulation, and key metabolic biomarkers in a high-fat diet-induced obesity model. Methods: A high-fat diet (HFD) induced obesity in C57BL/6 mice. The mice were then treated orally with either orlistat (positive control) or WIB. Changes in body weight, food intake, and fat weight were measured, along with blood lipid profiles and adipokines. Western blot analyses were conducted to determine protein levels in each tissue. H&E staining in white adipose tissue and liver, and the gut microbiota composition were analyzed. Results: WIB treatment significantly reduced body weight and fat mass compared to the HFD group and demonstrated comparable effects to orlistat. WIB improved blood lipid profiles and adipokine levels. H&E staining revealed reduced fat accumulation in the white adipose tissue and liver. Also in those tissues, WIB restored expression levels of sterol regulatory element-binding protein-1 (SREBP-1) and CCAAT/enhancer-binding protein α (C/EBPα) and increased AMP-activated protein kinase (AMPK) phosphorylation. In brown adipose tissue, WIB enhanced AMPK phosphorylation and upregulated thermogenic-related proteins, including peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), peroxisome proliferator-activated receptor α (PPARα), sirtuin 1 (SIRT1), uncoupling protein-1 (UCP-1), and cytochrome C oxidase subunit 4 (COX-IV). Analysis of gut microbiota revealed that WIB normalized β-diversity and reversed HFD-induced phyla imbalances (notably in Bacteroidetes, Firmicutes, and Proteobacteria). Conclusions: By reducing adiposity under the conditions tested in a murine model, improving metabolic markers, and favorably modulating gut microbiota, WIB demonstrates potential in mitigating obesity-related risks. These findings suggest that WIB may serve as a promising natural substance for the management of obesity. Further studies are warranted to confirm its efficacy and explore the potential underlying mechanisms in overweight or obese humans as a health supplement to help manage or prevent obesity.
Collapse
Affiliation(s)
- Chae-Won Lee
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ye Seul Yoon
- Development Department, Dalim Biotech Co., Ltd., Wonju-si 26348, Gangwon-do, Republic of Korea; (Y.S.Y.); (M.-j.K.)
| | - Young-Seo Yoon
- BELABEL BIO. Inc., Gimhae-si 50969, Gyeongsangnam-do, Republic of Korea; (Y.-S.Y.); (K.-S.C.)
| | - Kyung-Sook Chung
- BELABEL BIO. Inc., Gimhae-si 50969, Gyeongsangnam-do, Republic of Korea; (Y.-S.Y.); (K.-S.C.)
| | - Mi-ju Kim
- Development Department, Dalim Biotech Co., Ltd., Wonju-si 26348, Gangwon-do, Republic of Korea; (Y.S.Y.); (M.-j.K.)
| | - Geonha Park
- Division of Pharmacognosy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (G.P.); (M.C.)
- Department of Oriental Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Minsik Choi
- Division of Pharmacognosy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (G.P.); (M.C.)
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Young-Pyo Jang
- Division of Pharmacognosy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (G.P.); (M.C.)
- Department of Oriental Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Tae Lee
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
37
|
Constant B, Kamzolas I, Yang X, Guo J, Rodriguez-Fdez S, Mali I, Rodriguez-Cuenca S, Petsalaki E, Vidal-Puig A, Li W. Distinct signalling dynamics of BMP4 and BMP9 in brown versus white adipocytes. Sci Rep 2025; 15:15971. [PMID: 40335635 PMCID: PMC12059129 DOI: 10.1038/s41598-025-99122-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
Adipocyte dysfunction contributes to lipotoxicity and cardiometabolic diseases. Bone morphogenetic protein 4 (BMP4) is expressed in white adipocytes and remodels white adipose tissue, while liver-derived BMP9, a key circulating BMP, influences adipocyte lipid metabolism. The gene sets regulated by BMP4 and BMP9 signalling in mature adipocytes remain unclear. Here, we directly compare BMP4 and BMP9 signalling in mature brown and white adipocytes. While both BMPs showed comparable potency across adipocyte types, RNA sequencing analysis revealed extensive gene regulation, with many more differentially expressed genes and suppression of critical metabolic pathways in white adipocytes. Although BMP4 and BMP9 induced inhibitors of BMP and GDF signalling in both adipocytes, they selectively upregulated several TGF-β family receptors and BMP4 expression only in white adipocytes. These findings underscore a central role of BMP signalling in adipocyte homeostasis and suggest both BMP4 and BMP9 as regulators of white adipocyte plasticity with potential therapeutic implications.
Collapse
Affiliation(s)
- Benjamin Constant
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Ioannis Kamzolas
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Xudong Yang
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Jingxu Guo
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Sonia Rodriguez-Fdez
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
| | - Iman Mali
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
| | - Sergio Rodriguez-Cuenca
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Antonio Vidal-Puig
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK.
- CIBERDEN, Centro de Investigacion Principe Felipe, Valencia, Spain.
| | - Wei Li
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
| |
Collapse
|
38
|
Gu Q, Wang L, Xu M, Zhou W, Liu G, Tian H, Efferth T, Wang C, Fu Y. The natural dihydrochalcone phloretin reduces lipid accumulation via downregulation of IIS and sbp-1/ SREBP pathways in HepG2 cells and Caenorhabditis elegans. Food Funct 2025. [PMID: 40326995 DOI: 10.1039/d5fo01105a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Phloretin, a natural dihydrochalcone, exhibits significant potential in modulating lipid metabolism both in vitro and in vivo. This study investigated the effects of phloretin on lipid accumulation in HepG2 cells and Caenorhabditis elegans. In HepG2 cells, phloretin reduced lipid accumulation, ROS levels, and lipid peroxidation while ameliorating mitochondrial dysfunction. It downregulated lipid synthesis genes (SREBP, FASN) and upregulated PI3K-AKT pathway genes (AKT, FOXO, MTOR). In C. elegans, phloretin alleviated lipid accumulation-induced growth and locomotor impairments, reduced lipofuscin, ROS, glucose, and triglyceride levels, and modulated amino acid and lipid metabolism pathways. Gene expression analysis revealed downregulation of sbp-1, mdt-15, fat-5, fat-6, and fat-7, and upregulation of daf-16, age-1, and skn-1. Mutant studies confirmed that phloretin's lipid-lowering effects were mediated through the IIS and sbp-1/SREBP pathways. These findings suggest phloretin is a promising candidate for regulating lipid metabolism and preventing hyperlipidemia.
Collapse
Affiliation(s)
- Qi Gu
- The College of Forestry, Beijing Forestry University, 100083, Beijing, PR China.
| | - Litao Wang
- The College of Forestry, Beijing Forestry University, 100083, Beijing, PR China.
| | - Mingyue Xu
- The College of Forestry, Beijing Forestry University, 100083, Beijing, PR China.
| | - Wanmei Zhou
- The College of Forestry, Beijing Forestry University, 100083, Beijing, PR China.
| | - Guosheng Liu
- The College of Forestry, Beijing Forestry University, 100083, Beijing, PR China.
| | - Haiting Tian
- The College of Forestry, Beijing Forestry University, 100083, Beijing, PR China.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Chenlu Wang
- The College of Forestry, Beijing Forestry University, 100083, Beijing, PR China.
| | - Yujie Fu
- The College of Forestry, Beijing Forestry University, 100083, Beijing, PR China.
- Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, 150040, Harbin, PR China
| |
Collapse
|
39
|
Santos L, Fort RS, Schlapp G, Cal K, Perez-Torrado V, Meikle MN, Mulet AP, Espasandín C, Chiesa C, Sotelo-Silveira JR, Verdes JM, Contreras P, Calliari AJ, Crispo M, Badano JL, Escande C. Adipocyte-specific deletion of Dbc1 does not recapitulate healthy obesity phenotype but suggests regulation of inflammation signaling. PLoS One 2025; 20:e0322732. [PMID: 40315412 PMCID: PMC12048159 DOI: 10.1371/journal.pone.0322732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/25/2025] [Indexed: 05/04/2025] Open
Abstract
The protein Deleted in Breast Cancer 1 (Dbc1) is an important regulator of various transcription factors and epigenetic modulators, significantly influencing metabolism, obesity, and aging-related processes. Knockout mice lacking Dbc1 exhibit severe obesity but remain protected from liver steatosis, insulin resistance, and atherosclerosis. We hypothesized that this phenotype of "healthy obesity" results from adipose tissue expansion, which prevents free fatty acid spillover and subsequent metabolic damage to peripheral tissues. To further investigate the putative role of Dbc1 in adipose cells during obesity and its effects on metabolic dysregulation, we generated conditional Dbc1 knockout (KO) mice by backcrossing with AdipoQ-CRE transgenic mice to selectively abrogate Dbc1 expression in all mature adipocytes (Dbc1LoxP/LoxP;CRE). These mice demonstrated effective deletion of Dbc1 in mature adipocytes across various fat depots. We assessed the impact of Dbc1 deletion on metabolic regulation in male and female mice fed standard chow and high-fat diets. Our findings revealed that Dbc1 knockout in mature adipocytes did not influence weight gain, glucose tolerance, or other metabolic dysregulation markers, irrespective of sex. However, Dbc1 KO adipocytes exhibited an mRNA expression profile indicative of heightened inflammation during obesity. These results suggest that the protective phenotype observed in whole-body Dbc1 KO obese mice is not attributable to Dbc1's function within mature adipocytes but likely involves other cell types in adipose tissue. Moreover, the specific deletion of Dbc1 in mature adipocytes unveils a novel role of Dbc1 in inflammation signaling during obesity.
Collapse
Affiliation(s)
- Leonardo Santos
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Rafael Sebastián Fort
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
- Sección Genómica Funcional, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Geraldine Schlapp
- Unidad de Biotecnología en Animales de Laboratorio, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Karina Cal
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Valentina Perez-Torrado
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Maria Noel Meikle
- Unidad de Biotecnología en Animales de Laboratorio, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Ana Paula Mulet
- Unidad de Biotecnología en Animales de Laboratorio, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Camila Espasandín
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Department of Biosciences, Facultad de Veterinaria, Universidad de la República, Universidad de la República, Montevideo, Uruguay
| | - Camila Chiesa
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - José R. Sotelo-Silveira
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
- Departamento de Biología Celular y Molecular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Jose M. Verdes
- Unidad Patología, Departamento de Patobiología, Facultad de Veterinaria, Universidad de la República, Montevideo, Uruguay
| | - Paola Contreras
- Departamento de Fisiología, Facultad de Medicina, Udelar, Uruguay
| | - Aldo J. Calliari
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Department of Biosciences, Facultad de Veterinaria, Universidad de la República, Universidad de la República, Montevideo, Uruguay
| | - Martina Crispo
- Unidad de Biotecnología en Animales de Laboratorio, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Jose L. Badano
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Carlos Escande
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur de Montevideo, Montevideo, Uruguay
| |
Collapse
|
40
|
Khalaf F, Barayan D, Saldanha S, Jeschke MG. Metabolaging: a new geroscience perspective linking aging pathologies and metabolic dysfunction. Metabolism 2025; 166:156158. [PMID: 39947519 DOI: 10.1016/j.metabol.2025.156158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
With age, our metabolic systems undergo significant alterations, which can lead to a cascade of adverse effects that are implicated in both metabolic disorders, such as diabetes, and in the body's ability to respond to acute stress and trauma. To elucidate the metabolic imbalances arising from aging, we introduce the concept of "metabolaging." This framework encompasses the broad spectrum of metabolic disruptions associated with the hallmarks of aging, including the functional decline of key metabolically active organs, like the adipose tissue. By examining how these organs interact with essential nutrient-sensing pathways, "metabolaging" provides a more comprehensive view of the systemic metabolic imbalances that occur with age. This concept extends to understanding how age-related metabolic disturbances can influence the response to acute stressors, like burn injuries, highlighting the interplay between metabolic dysfunction and the ability to handle severe physiological challenges. Finally, we propose potential interventions that hold promise in mitigating the effects of metabolaging and its downstream consequences.
Collapse
Affiliation(s)
- Fadi Khalaf
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Dalia Barayan
- David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Sean Saldanha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Marc G Jeschke
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
41
|
Martins FF, Amarante MDSM, Oliveira DS, Vasques‐Monteiro IML, Souza‐Mello V, Daleprane JB, Camillo CDS. Obesity, White Adipose Tissue, and Adipokines Signaling in Male Reproduction. Mol Nutr Food Res 2025; 69:e70054. [PMID: 40195898 PMCID: PMC12087738 DOI: 10.1002/mnfr.70054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/19/2025] [Accepted: 03/17/2025] [Indexed: 04/09/2025]
Abstract
Currently, obesity is a global pandemic characterized by systemic metabolic complications that negatively impact several organs, including white adipose tissue (WAT) and the tissues of the male reproductive system. Since the discovery of leptin in 1994, WAT has been recognized as a dynamic endocrine organ for secreting a series of molecules with hormonal functions, collectively called adipokines. The link between obesity, WAT, adipokines, and the male reproductive system is direct and little explored. With changes in nutritional status, WAT undergoes morphofunctional changes, and the secretion of adipokines is altered, negatively impacting reproductive mechanisms, including steroidogenesis and spermatogenesis. In this review, we address in an updated way the structural and functional characteristics of WAT as well as the link between obesity and changes in the signaling pathways of the adipokines leptin, adiponectin, resistin, visfatin, apelin, chemerin, omentin-1, vaspin, and asprosin in male reproduction. Understanding the relationship between obesity, these adipokines, and reproductive dysfunction can contribute to new strategies for the treatment of subfertility and male infertility.
Collapse
Affiliation(s)
| | | | - Daiana Santana Oliveira
- Laboratory of MorphometryMetabolism and Cardiovascular DiseasesBiomedical CenterInstitute of BiologyRio de Janeiro State UniversityRio de JaneiroBrazil
| | - Isabela Macedo Lopes Vasques‐Monteiro
- Department of Basic and Experimental NutritionLaboratory for Studies of Interactions Between Nutrition and GeneticsLEINGRio de Janeiro State UniversityRio de JaneiroBrazil
| | - Vanessa Souza‐Mello
- Laboratory of MorphometryMetabolism and Cardiovascular DiseasesBiomedical CenterInstitute of BiologyRio de Janeiro State UniversityRio de JaneiroBrazil
| | - Julio Beltrame Daleprane
- Department of Basic and Experimental NutritionLaboratory for Studies of Interactions Between Nutrition and GeneticsLEINGRio de Janeiro State UniversityRio de JaneiroBrazil
| | | |
Collapse
|
42
|
Liu Z, Li Q, Zhao F, Chen J. A decade review on phytochemistry and pharmacological activities of Cynomorium songaricum Rupr.: Insights into metabolic syndrome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156602. [PMID: 40058318 DOI: 10.1016/j.phymed.2025.156602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/12/2025] [Accepted: 03/01/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Cynomorium songaricum Rupr. (CSR), a perennial herb with a rich history in traditional medicine, has demonstrated therapeutic potential against metabolic syndrome (MetS) through its active compounds, including proanthocyanidins, polysaccharides, and triterpenoids. MetS, a global health concern, encompasses interlinked conditions such as obesity, type 2 diabetes mellitus (T2DM), and inflammation. This review synthesizes recent findings on CSR's pharmacological and phytochemical properties, focusing on its role in ameliorating MetS. METHODS Following Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines, relevant studies were retrieved from PubMed, Web of Science, and CNKI databases up to December 2024. Keywords included "Cynomorium Songaricum Rupr.", "Cynomorii Herba", "Suoyang", "Suo Yang", "Metabolic syndrome", "Proanthocyanidins", "Polysaccharides" and "Triterpenoids" and their combinations. Inclusion criteria emphasized studies exploring CSR's impact on MetS, while duplicate, low-quality studies and studies not written in Chinese, English, or unrelated were excluded. RESULTS A total of 92 studies were analyzed, revealing that CSR's active components exhibit multi-target effects. Proanthocyanidins reduce glucose absorption and oxidative stress, polysaccharides enhance insulin sensitivity and gut microbiota composition, and triterpenoids mitigate obesity and mitochondria damage. These mechanisms collectively contribute to the beneficial effects of CSR against MetS. CONCLUSION CSR presents a promising natural therapy for MetS, utilizing its pharmacologically active compounds to address core metabolic dysfunctions. Future studies should focus on clinical validation and safety assessments to facilitate CSR's integration into modern therapeutic regimens.
Collapse
Affiliation(s)
- Zhihao Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China; The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, China
| | - Qihao Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Fu Zhao
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Jihang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China; The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, China.
| |
Collapse
|
43
|
Zhang GL, Lee J. Associations of Combined Physical Activity and Body Mass Index with Metabolic Syndrome in American Adults: From the National Health and Nutrition Examination Survey, 2017-2020. Am J Health Promot 2025; 39:598-608. [PMID: 39653592 DOI: 10.1177/08901171241307434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
PurposeThe purpose of this study is to examine the combined associations of physical activity and body mass index with the prevalence of metabolic syndrome in a population-based sample of American adults.DesignCross-sectional study.SettingData from the 2017-2020 National Health and Nutrition Examination Survey (NHANES).SubjectsA total of 2945 American adults who participated in the NHANES 2017-2020.MeasuresParticipants' physical activity levels were categorized based on the Physical Activity Guidelines for Americans. Metabolic syndrome was determined using the National Cholesterol Education Program Adult Treatment Panel III guidelines.AnalysisThe prevalence of metabolic syndrome among the various groups was analyzed using binary logistic regression to observe differences in the associated risks of developing metabolic syndrome.ResultsAn increase in body mass index and a decrease in physical activity are both risk factors for metabolic syndrome. Statistical results show that the prevalence of metabolic syndrome in the obese and sedentary group is 22.31 times higher than that in the normal weight and active group (P = 0.000).ConclusionA combination of at least 600 MET-minutes of physical activity per week and a lower body mass index is significantly associated with the lowest prevalence of metabolic syndrome.
Collapse
Affiliation(s)
- Guang-Lei Zhang
- Graduate School of Sport Sceince, Kyung Hee University, Yongin-si, Republic of Korea
| | - Junga Lee
- Graduate School of Sport Sceince, Kyung Hee University, Yongin-si, Republic of Korea
| |
Collapse
|
44
|
Lacruz-Pleguezuelos B, Bazán GX, Romero-Tapiador S, Freixer G, Tolosana R, Daza R, Fernández-Díaz CM, Molina S, Crespo MC, Laguna T, Marcos-Zambrano LJ, Aguilar-Aguilar E, Fernández-Cabezas J, Cruz-Gil S, Fernández LP, Vera-Rodriguez R, Fierrez J, Ramírez de Molina A, Ortega-Garcia J, Morales A, Carrillo de Santa Pau E, Espinosa-Salinas I. AI4Food, a feasibility study for the implementation of automated devices in the nutritional advice and follow up within a weight loss intervention. Clin Nutr 2025; 48:80-89. [PMID: 40168934 DOI: 10.1016/j.clnu.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/17/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND & AIMS The widespread prevalence of NCDs calls for an improvement in their prevention and treatment. Wearable technologies can be an important asset in the development of precision nutrition strategies, for both health professionals and patients. However, their clinical use is hindered by a lack of validation against current methodologies or appropriate tools to deliver nutritional strategies based on their data. Our study includes manual and automatic data capture methods within a weight loss intervention with the aim to create an essential asset for the implementation, validation, and benchmarking of AI-based tools in nutritional clinical practice. METHODS This is a feasibility prospective and crossover controlled trial for weight loss in overweight and obese participants, randomized into two groups: Group 1 used manual data collection methods based on validated questionnaires for the first two weeks; while Group 2 started with automatic data collection methods consisting of wearable sensors. After two weeks, the two groups switched data collection methods. Lifestyle data, anthropometric measurements and biological samples were collected from all participants. RESULTS A total of 93 participants completed the nutritional intervention designed for weight loss, achieving a mean reduction of 2 kg (V1: 84.99 SD ± 13.69, V3: 82.72 SD ± 13.32, p < 0.001). Significant reductions were observed in body mass index, visceral fat, waist circumference, total cholesterol, and HbA1c levels. The use of electronic devices proved satisfactory among the participants (System Usability Scale score 78.27 ± 12.86). We also report the presence of distinct patient groups based on continuous glucose measurements. CONCLUSION This study has yielded a large amount of data and has showcased how automatic data collection devices can be employed to gather data in the context of a nutritional intervention. This will enable the implementation of AI-based tools in nutritional clinical practice. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, NCT05807243.
Collapse
Affiliation(s)
- Blanca Lacruz-Pleguezuelos
- Computational Biology Group, IMDEA Food, CEI UAM+CSIC, Carretera de Cantoblanco, 8, 28049 Madrid Spain; UAM Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain
| | - Guadalupe X Bazán
- GENYAL Platform, IMDEA Food, CEI UAM+CSIC, Carretera de Cantoblanco, 8, 28049 Madrid Spain
| | - Sergio Romero-Tapiador
- Biometrics and Data Pattern Analytics Lab, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Spain
| | - Gala Freixer
- GENYAL Platform, IMDEA Food, CEI UAM+CSIC, Carretera de Cantoblanco, 8, 28049 Madrid Spain
| | - Ruben Tolosana
- Biometrics and Data Pattern Analytics Lab, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Spain
| | - Roberto Daza
- Biometrics and Data Pattern Analytics Lab, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Spain
| | | | - Susana Molina
- GENYAL Platform, IMDEA Food, CEI UAM+CSIC, Carretera de Cantoblanco, 8, 28049 Madrid Spain
| | - María Carmen Crespo
- GENYAL Platform, IMDEA Food, CEI UAM+CSIC, Carretera de Cantoblanco, 8, 28049 Madrid Spain
| | - Teresa Laguna
- Computational Biology Group, IMDEA Food, CEI UAM+CSIC, Carretera de Cantoblanco, 8, 28049 Madrid Spain
| | | | - Elena Aguilar-Aguilar
- GENYAL Platform, IMDEA Food, CEI UAM+CSIC, Carretera de Cantoblanco, 8, 28049 Madrid Spain; Department of Pharmacy and Nutrition, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Calle Tajo s/n, Villaviciosa de Odon, 28670, Spain
| | | | - Silvia Cruz-Gil
- Molecular Oncology Group, IMDEA Food, CEI UAM+CSIC, Carretera de Cantoblanco, 8, 28049 Madrid Spain
| | - Lara P Fernández
- Molecular Oncology Group, IMDEA Food, CEI UAM+CSIC, Carretera de Cantoblanco, 8, 28049 Madrid Spain
| | - Ruben Vera-Rodriguez
- Biometrics and Data Pattern Analytics Lab, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Spain
| | - Julian Fierrez
- Biometrics and Data Pattern Analytics Lab, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Spain
| | - Ana Ramírez de Molina
- GENYAL Platform, IMDEA Food, CEI UAM+CSIC, Carretera de Cantoblanco, 8, 28049 Madrid Spain
| | - Javier Ortega-Garcia
- Biometrics and Data Pattern Analytics Lab, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Spain
| | - Aythami Morales
- Biometrics and Data Pattern Analytics Lab, Escuela Politécnica Superior, Universidad Autónoma de Madrid, Spain
| | | | | |
Collapse
|
45
|
Chen D, Yang YY, Yang Y. Astrocyte Loss Augments Body Weight Through Reduction in Adipose Sympathetic Outflows. Glia 2025; 73:1068-1076. [PMID: 39780483 PMCID: PMC11922664 DOI: 10.1002/glia.24673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Emerging evidence indicates that astrocytes modulate energy metabolism and homeostasis. However, one important but poorly understood element is the necessity of astrocytes in the control of body weight. Here, we apply viral vector-assisted brain-region selective loss of astrocytes to define physiological roles played by astrocytes in the arcuate nucleus of the hypothalamus (ARH) and to elucidate the involved mechanism. We find that astrocyte loss potently augments body weight in adult mice fed chow or high-fat diet. Mechanistically, we find that the loss of astrocytes reduces adipose tissue norepinephrine (NE) contents and chemogenetic stimulation of adipose tissue sympathetic inputs reverses the astrocyte loss-induced increase in body weight. Collectively, our findings in this study suggest a crucial physiological role of astrocytes in preventing diet-induced energy surfeit and obesity by modulating adipose tissue lipid metabolism through central sympathetic outflows to adipose tissues.
Collapse
Affiliation(s)
- Dan Chen
- Department of Medicine Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | | | - Yunlei Yang
- Department of Medicine Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
46
|
Solsona‐Vilarrasa E, Vousden KH. Obesity, white adipose tissue and cancer. FEBS J 2025; 292:2189-2207. [PMID: 39496581 PMCID: PMC12062788 DOI: 10.1111/febs.17312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024]
Abstract
White adipose tissue (WAT) is crucial for whole-body energy homeostasis and plays an important role in metabolic and hormonal regulation. While healthy WAT undergoes controlled expansion and contraction to meet the body's requirements, dysfunctional WAT in conditions like obesity is characterized by excessive tissue expansion, alterations in lipid homeostasis, inflammation, hypoxia, and fibrosis. Obesity is strongly associated with an increased risk of numerous cancers, with obesity-induced WAT dysfunction influencing cancer development through various mechanisms involving both systemic and local interactions between adipose tissue and tumors. Unhealthy obese WAT affects circulating levels of free fatty acids and factors like leptin, adiponectin, and insulin, altering systemic lipid metabolism and inducing inflammation that supports tumor growth. Similar mechanisms are observed locally in an adipose-rich tumor microenvironment (TME), where WAT cells can also trigger extracellular matrix remodeling, thereby enhancing the TME's ability to promote tumor growth. Moreover, tumors reciprocally interact with WAT, creating a bidirectional communication that further enhances tumorigenesis. This review focuses on the complex interplay between obesity, WAT dysfunction, and primary tumor growth, highlighting potential targets for therapeutic intervention.
Collapse
|
47
|
Wang Q, Wang S, Zhuang Z, Wu X, Gao H, Zhang T, Zou G, Ge X, Liu Y. Identification of potential crucial genes and mechanisms associated with metabolically unhealthy obesity based on the gene expression profile. Front Genet 2025; 16:1540721. [PMID: 40376303 PMCID: PMC12078199 DOI: 10.3389/fgene.2025.1540721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/10/2025] [Indexed: 05/18/2025] Open
Abstract
Background Obesity is an epidemic and systemic metabolic disease that seriously endangers human health. This study aimed to understand the transcriptomic characteristics of the blood of metabolically unhealthy obesity (MUO) and provide insight into the target genes of differently expressed microRNAs in the occurrence and development of MUO. Methods The GSE146869, GSE145412, GSE23561, and GSE169290 datasets were analyzed to understand the transcriptome characteristics of the blood of MUO and provide insights into the target genes of differently expressed microRNAs (DEMs) in MUO. Functional and pathway enrichment analyses and gene interaction network analyses were performed to profile the function of differentially expressed genes (DEGs). In addition, miRNet 2.0, TransmiR v2.0, RNA22, TargetScan 7.2, miRDB, and miRWalk databases were used to predict the target genes of effective microRNAs. Results A total of 189 co-DEGs were identified in at least two datasets. The 156 co-upregulated genes were enriched into 29 biological process (BP) terms and 12 KEGG pathways. Among the 29 BP terms, the immune- and metabolism-related BP terms were enriched. The 33 co-downregulated genes were enriched into two BP terms, including apoptotic process and regulation of the apoptotic process, with no KEGG pathway. The hub genes EGF, STAT3, IL1B, PF4, SELP, and ITGA2B in the gene interaction network might play important roles in abnormal BP in MUO. Among the 19 DEMs identified in the blood of the MUO group by the GSE169290 dataset, 18 microRNAs targeted 85 genes as risk factors in MUO. Conclusion A network consisting of 18 microRNAs and 85 target genes might serve as a risk factor for metabolically unhealthy obesity.
Collapse
Affiliation(s)
- Qingqing Wang
- Department of Nephrology, Xuzhou Children’s Hospital, Xuzhou, Jiangsu, China
| | - Silu Wang
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhanyu Zhuang
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, Jiangsu, China
| | - Xueting Wu
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, Jiangsu, China
| | - Hongkun Gao
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tianyi Zhang
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, Jiangsu, China
| | - Guorong Zou
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, Jiangsu, China
| | - Xing Ge
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, Jiangsu, China
| | - Yapeng Liu
- Yunlong District Center for Disease Control and Prevention, Xuzhou, Jiangsu, China
| |
Collapse
|
48
|
Bahrami LS, Rahnama I, Chambari M, Norouzy A, Karav S, Arabi SM, Sahebkar A. The Effects of Curcuminoids Supplementation on Serum Adipokines: An Umbrella Review of Meta-Analyses of Randomized Controlled Trials. Phytother Res 2025; 39:2250-2261. [PMID: 40109154 DOI: 10.1002/ptr.8471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 01/08/2025] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
This umbrella review of randomized clinical trials aims to provide a unique and detailed understanding of curcumin's effects on adipokines, adding a novel perspective to the existing body of research. We carried out a thorough search of international databases up to April 2024, including MEDLINE, SciVerse Scopus, and Clarivate Analytics Web of Science. A random-effects model was utilized to evaluate the impact of curcuminoid on adipokines. The umbrella review incorporated meta-analyses that examined the effects of curcuminoid supplementation on adipokines, presenting associated effect sizes (ES) and confidence intervals (CI). We applied the GRADE and AMSTAR (A Tool for Assessing the Risk of Bias in Systematic Reviews system) to assess the certainty of the evidence and the quality of the systematic reviews. Our analysis of one meta-analysis, including 14 RCTs plus 1 RCT not included in meta-analyses, revealed significant and impactful findings. We found a substantial increase in serum adiponectin levels with curcuminoid supplementation, indicating a positive effect (SMD: 0.9; 95% CI, 0.4 to 1.3, p < 0.001; I2 = 92.2%). However, we did not observe a significant impact on serum leptin. The GRADE assessment supports the effect of curcuminoids on adiponectin with moderate evidence, while the impact on leptin was supported by low evidence. Curcuminoid supplementation significantly increases serum adiponectin levels with moderate-quality evidence and has no significant impact on serum leptin. This provides evidence as to the safety and effectiveness of curcuminoids in enhancing adiponectin without adverse effects, reassuring the audience about their potential in adipokine research.
Collapse
Affiliation(s)
- Leila Sadat Bahrami
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Iman Rahnama
- Binaloud Institute of Higher Education, Mashhad, Iran
| | - Mahla Chambari
- Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Abdolreza Norouzy
- Department of Clinical Nutrition, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Seyyed Mostafa Arabi
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
49
|
Sleiman L, Dinescu S. Role of Non-Coding RNAs in White and Brown Adipose Tissue Differentiation and Development. Noncoding RNA 2025; 11:30. [PMID: 40407588 PMCID: PMC12101253 DOI: 10.3390/ncrna11030030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/18/2025] [Accepted: 04/28/2025] [Indexed: 05/26/2025] Open
Abstract
Adipocyte differentiation is a complex process in which pluripotent mesenchymal stem cells (MSCs) differentiate and develop into mature fat cells, also known as adipocytes. This process is controlled by various transcription factors, hormones, and signaling molecules that regulate the development of these cells. Recently, an increasing number of non-coding RNAs (ncRNAs), especially microRNAs (miRNAs), have been established to be involved in the regulation of many biological processes, including adipocyte differentiation, development, metabolism, and energy homeostasis of white and brown adipose tissue. Several in vitro and in vivo studies reported the significant role of ncRNAs in either promoting or inhibiting adipocyte differentiation into white or brown fat cells by targeting specific transcription factors and regulating the expression of key adipogenic genes. Identifying the function of ncRNAs and their subsequent targets contributes to our understanding of how these molecules can be used as potential biomarkers and tools for therapies against obesity, diabetes, and other diseases related to obesity. This could also contribute to advancements in tissue-engineering based treatments. In this review, we intended to present an up-to-date comprehensive literature overview of the role of ncRNAs, including miRNAs, long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), focusing particularly on miRNAs, in regulating the differentiation and development of cells into white and brown adipose tissue. In addition, we further discuss the potential use of these molecules as biomarkers for the development of novel therapeutic strategies for future personalized treatment options for patients.
Collapse
Affiliation(s)
- Lea Sleiman
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania;
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania;
- Research Institute of the University of Bucharest (ICUB), 050663 Bucharest, Romania
| |
Collapse
|
50
|
Makassy D, Williams K, Karwi QG. The Evolving Role of Macrophage Metabolic Reprogramming in Obesity. Can J Cardiol 2025:S0828-282X(25)00320-4. [PMID: 40311669 DOI: 10.1016/j.cjca.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025] Open
Abstract
Recent research has extensively explored the critical role of energy metabolism in shaping the inflammatory response and polarization of macrophages in obesity. This rapidly growing field emphasizes the need to understand the connection between metabolic processes that support macrophage polarization in obesity. Although most published research in this area has focused on glucose and fatty acids, how the flux through other metabolic pathways (such as ketone and amino acid oxidation) in macrophages is altered in obesity is not well defined. This review summarizes the main alterations in uptake, storage, and oxidation of oxidative substrates (glucose, fatty acids, ketone bodies, and amino acids) in macrophages and how these alterations are linked to macrophage polarization and contribution to augmented inflammatory markers in obesity. The review also discusses how oxidative substrates could modulate macrophage energy metabolism and inflammatory responses via feeding into other nonoxidative pathways (such as the pentose phosphate pathway, triacylglycerol synthesis/accumulation), via acting as signalling molecules, or via mediating post-translational modifications (such as O-GlcNAcylation or β-hydroxybutyrylation). The review also identifies several critical unanswered questions regarding the characteristics (functional and metabolic) of macrophages from different origins (adipose tissue, skeletal muscle, bone marrow) in obesity and how these characteristics contribute to early vs late phases of obesity. We also identified a number of new therapeutic targets that could be evaluated in future investigations. Targeting macrophage metabolism in obesity is an exciting and active area of research with significant potential to help identify new treatments to limit the detrimental effects of inflammation in obesity.
Collapse
Affiliation(s)
- Dorcus Makassy
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, Newfoundland and Labrador, Canada
| | - Kyra Williams
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, Newfoundland and Labrador, Canada
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, Newfoundland and Labrador, Canada.
| |
Collapse
|