1
|
Yıldırım Ayaz E, Mesci B, Üner ÖE, Kaya FN, Dincer B, İşman FK, Oğuz A. The effect of exercise on GDF-15 levels in individuals with prediabetes: A randomized controlled trial. J Diabetes Investig 2025; 16:656-669. [PMID: 39868911 PMCID: PMC11970312 DOI: 10.1111/jdi.14404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/12/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
AIMS Growth differentiation factor-15 (GDF-15) is an inflammatory cytokine that increases in prediabetes and is known for its anorexigenic effects. This study aims to evaluate the effects of a 12-week exercise program on GDF-15 in individuals with prediabetes. MATERIALS AND METHODS In this multicenter, parallel-group, randomized-controlled trial, 64 patients aged 18-60 diagnosed with prediabetes were randomized in a 1:1 ratio into the exercise group (E) and the control group (C). Additionally, 32 patients who were planned to start metformin were included in the metformin group (M). Participants in the exercise group engaged in aerobic exercise at 50-70% of their maximum heart rate for 60 min, 3 days a week. Serum GDF-15 levels were evaluated at the beginning and the end of the 12th week. RESULTS The mean age of the 91 participants who completed the study was 46.13 ± 8.52 years, and 23.1% were male. Basal GDF-15 levels were similar among the groups (E = 668.6 ± 415.1, C = 651.8 ± 352.5, M = 603.6 ± 387.2, P = 0.47). At the 12th week, GDF-15 levels were lower in the E compared to the C, while higher in the M compared to the C (E = 383.1 ± 215.6, C = 556.4 ± 285.6, M = 810.8 ± 498.0, P < 0.001). In inter-group comparisons, no significant change was observed in the C between the 0th and 12th weeks, while GDF-15 decreased in the E (P < 0.001) and increased in the M (P < 0.001). CONCLUSIONS It was determined that in individuals with prediabetes, GDF-15, which serves both as a biomarker of metabolic disorder and has a negative regulatory effect on appetite, decreased with 12 weeks of aerobic exercise and increased with metformin administration.
Collapse
Affiliation(s)
- Elif Yıldırım Ayaz
- Sultan 2. Abdülhamid Han Training and Research HospitalUniversity of Health SciencesÜsküdar, İstanbulTurkey
| | - Banu Mesci
- Göztepe Prof. Dr. Süleyman Yalçın City HospitalIstanbul Medeniyet UniversityİstanbulTurkey
| | - Özden Ezgi Üner
- Fatih Sultan Mehmet Training and Research HospitalUniversity of Health SciencesÜsküdar, İstanbulTurkey
| | | | - Berna Dincer
- Faculty of Health SciencesIstanbul Medeniyet UniversityİstanbulTurkey
| | - Ferruh Kemal İşman
- Göztepe Prof. Dr. Süleyman Yalçın City HospitalIstanbul Medeniyet UniversityİstanbulTurkey
| | - Aytekin Oğuz
- Göztepe Prof. Dr. Süleyman Yalçın City HospitalIstanbul Medeniyet UniversityİstanbulTurkey
| |
Collapse
|
2
|
Xu S, Liu Z, Tian T, Zhao W, Wang Z, Liu M, Xu M, Zhang F, Zhang Z, Chen M, Yin Y, Su M, Fang W, Pan W, Liu S, Li MD, Little PJ, Kamato D, Zhang S, Wang D, Offermanns S, Speakman JR, Weng J. The clinical antiprotozoal drug halofuginone promotes weight loss by elevating GDF15 and FGF21. SCIENCE ADVANCES 2025; 11:eadt3142. [PMID: 40138418 PMCID: PMC11939056 DOI: 10.1126/sciadv.adt3142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/20/2025] [Indexed: 03/29/2025]
Abstract
Obesity is a debilitating global pandemic with a huge cost on health care due to it being a major underlying risk factor for several diseases. Therefore, there is an unmet medical need for pharmacological interventions to curb obesity. Here, we report that halofuginone, a Food and Drug Administration-approved anti-scleroderma and antiprotozoal drug, is a promising anti-obesity agent in preclinical mouse and pig models. Halofuginone suppressed food intake, increased energy expenditure, and resulted in weight loss in diet-induced obese mice while also alleviating insulin resistance and hepatic steatosis. Using molecular and pharmacological tools with transcriptomics, we identified that halofuginone increases fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15) levels via activating integrated stress response. Using Gdf15 and Fgf21 knockout mice, we show that both hormones are necessary to elicit anti-obesity changes. Together, our study reports the beneficial metabolic effects of halofuginone and underscores its utility in treating obesity and its associated metabolic complications, which merits clinical assessment.
Collapse
Affiliation(s)
- Suowen Xu
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei 230001, China
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
- Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei 230001, China
| | - Zhenghong Liu
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Tian Tian
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Wenqi Zhao
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhihua Wang
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Monan Liu
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Mengyun Xu
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Fanshun Zhang
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhidan Zhang
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Meijie Chen
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yanjun Yin
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Meiming Su
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Wenxiang Fang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Wenhao Pan
- Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Shiyong Liu
- Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Min-dian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, MOE Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Peter J. Little
- Department of Pharmacy, Guangzhou Xinhua University, No. 721, Guangshan Road 1, Guangzhou 510520, China
| | - Danielle Kamato
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Queensland 4111, Australia
| | - Songyang Zhang
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Dongdong Wang
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - John R. Speakman
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- School of Biological Sciences, University of Aberdeen, Aberdeen AB24 3FX, UK
| | - Jianping Weng
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei 230001, China
- Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei 230001, China
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| |
Collapse
|
3
|
Chen J, Xiang J, Zhou M, Huang R, Zhang J, Cui Y, Jiang X, Li Y, Zhou R, Xin H, Li J, Li L, Lam SM, Zhu J, Chen Y, Yang Q, Xie Z, Shui G, Deng F, Zhang Z, Li MD. Dietary timing enhances exercise by modulating fat-muscle crosstalk via adipocyte AMPKα2 signaling. Cell Metab 2025:S1550-4131(25)00065-8. [PMID: PMID:40088888 DOI: 10.1016/j.cmet.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/16/2025] [Accepted: 02/22/2025] [Indexed: 03/17/2025]
Abstract
Feeding rhythms regulate exercise performance and muscle energy metabolism. However, the mechanisms regulating adipocyte functions remain unclear. Here, using multi-omics analyses, involving (phospho-)proteomics and lipidomics, we found that day-restricted feeding (DRF) regulates diurnal rhythms of the mitochondrial proteome, neutral lipidome, and nutrient-sensing pathways in mouse gonadal white adipose tissue (GWAT). Adipocyte-specific knockdown of Prkaa2 (the gene encoding AMPKα2) impairs physical endurance. This defect is associated with altered rhythmicity in acyl-coenzyme A (CoA) metabolism-related genes, a loss of rhythmicity in the GWAT lipidome, and circadian remodeling of serum metabolites-in particular, lactate and succinate. We also found that adipocyte Prkaa2 regulates muscle clock genes during DRF. Notably, oral administration of the AMPK activator C29 increases endurance and muscle functions in a time-of-day manner, which requires intact adipocyte AMPKα2 signaling. Collectively, our work defines adipocyte AMPKα2 signaling as a critical regulator of circadian metabolic coordination between fat and muscle, thereby enhancing exercise performance.
Collapse
Affiliation(s)
- Jianghui Chen
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Jing Xiang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Meiyu Zhou
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Rongfeng Huang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China; Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610072, China
| | - Jianxin Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China; Department of Cardiology, The 960th Hospital of the PLA Joint Service Support Force, Jinan 250000, China
| | - Yuanting Cui
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Xiaoqing Jiang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Yang Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Runchao Zhou
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Haoran Xin
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Jie Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Lihua Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; LipidALL Technologies Company Limited, Changzhou, China
| | - Jianfang Zhu
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Yanxiu Chen
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Qingyuan Yang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Zhifu Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fang Deng
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing 400038, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing 400038, China
| | - Zhihui Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China.
| | - Min-Dian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China.
| |
Collapse
|
4
|
Zhou Y, Dou L, Wang L, Chen J, Mao R, Zhu L, Liu D, Zheng K. Growth and differentiation factor 15: An emerging therapeutic target for brain diseases. Biosci Trends 2025; 19:72-86. [PMID: 39864834 DOI: 10.5582/bst.2024.01305] [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: 01/28/2025]
Abstract
Growth and differentiation factor 15 (GDF15), a member of the transforming growth factor-βsuperfamily, is considered a stress response factor and has garnered increasing attention in recent years due to its roles in neurological diseases. Although many studies have suggested that GDF15 expression is elevated in patients with neurodegenerative diseases (NDDs), glioma, and ischemic stroke, the effects of increased GDF15 expression and the potential underlying mechanisms remain unclear. Notably, many experimental studies have shown the multidimensional beneficial effects of GDF15 on NDDs, and GDF15 overexpression is able to rescue NDD-associated pathological changes and phenotypes. In glioma, GDF15 exerts opposite effects, it is both protumorigenic and antitumorigenic. The causes of these conflicting findings are not comprehensively clear, but inhibiting GDF15 is helpful for suppressing tumor progression. GDF15 is also regarded as a biomarker of poor clinical outcomes in ischemic stroke patients, and targeting GDF15 may help prevent this disease. Thus, we systematically reviewed the synthesis, transcriptional regulation, and biological functions of GDF15 and its related signaling pathways within the brain. Furthermore, we explored the potential of GDF15 as a therapeutic target and assessed its clinical applicability in interventions for brain diseases. By integrating the latest research findings, this study provides new insights into the future treatment of neurological diseases.
Collapse
Affiliation(s)
- Yingying Zhou
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Dou
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Luyao Wang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiajie Chen
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruxue Mao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lingqiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dan Liu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kai Zheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
5
|
Wang Q, Leask MP, Lee K, Jaiswal J, Kallingappa P, Dissanayake W, Puli'uvea C, O'Sullivan C, Watson H, Wilcox P, Murphy R, Merry TL, Shepherd PR. The population-specific Thr44Met OCT3 coding variant affects metformin pharmacokinetics with subsequent effects on insulin sensitivity in C57Bl/6J mice. Diabetologia 2025; 68:537-548. [PMID: 39422716 PMCID: PMC11832584 DOI: 10.1007/s00125-024-06287-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/19/2024] [Indexed: 10/19/2024]
Abstract
AIMS/HYPOTHESIS Metformin is an important first-line treatment for type 2 diabetes and acts by increasing the body's ability to dispose of glucose. Metformin's efficacy can be affected by genetic variants in the transporters that regulate its uptake into cells. The SLC22A3 gene (also known as EMT; EMTH; OCT3) codes for organic cation transporter 3 (OCT3), which is a broad-specificity cation transporter that also transports metformin. Most SLC22A3 variants reduce the rate of metformin transport but the rs8187715 variant (p.Thr44Met) is reported to increase uptake of metformin in vitro. However, the impact of this on in vivo metformin transport and efficacy is unknown. Very few carriers of this variant have been reported globally, but, notably, all were of Pacific Island descent. Therefore, this study aims to understand the prevalence of this variant in Polynesian peoples (Māori and Pacific peoples) and to understand its impact on metformin transport and efficacy in vivo. METHODS rs8187715 was genotyped in 310 individuals with Māori and Pacific ancestry recruited in Aotearoa New Zealand. To study this variant in a physiological context, an orthologous knockin mouse model with C57BL/6J background was used. Pharmacokinetic analysis compared uptake rate of metformin into tissues. Plasma growth/differentiation factor 15 (GDF-15) was also measured as a marker of metformin efficacy. Glucose and insulin tolerance was assessed after acute or sustained metformin treatment in knockin and wild-type control mice to examine the impact of the variant on metformin's glycaemic control. RESULTS The minor allele frequency of this variant in the Māori and Pacific participants was 15.4%. There was no association of the variant with common metabolic parameters including diabetes status, BMI, blood pressure, lipids, or blood glucose and HbA1c. However, in the orthologous knockin mouse model, the rate of metformin uptake into the blood and tissues was increased. Acute metformin dosing increased insulin sensitivity in variant knockin mice but this effect was lost after longer-term metformin treatment. Metformin's effects on GDF-15 levels were also lost in variant knockin mice with longer-term metformin treatment. CONCLUSIONS/INTERPRETATION These data provide evidence that the SLC22A3 rs8187715 variant accelerates metformin uptake rate in vivo. While this acutely improves insulin sensitivity, there was no increased effect of metformin with longer-term dosing. Thus, our finding of a high prevalence of this variant specifically in Māori and Pacific peoples identifies it as a potential population-specific pharmacogenetic marker with potential to guide metformin therapy in these peoples.
Collapse
Affiliation(s)
- Qian Wang
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | | | - Kate Lee
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | - Jagdish Jaiswal
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Prasanna Kallingappa
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Waruni Dissanayake
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | - Chris Puli'uvea
- Maurice Wilkins Centre, Auckland, New Zealand
- Department of Biomedicine and Diagnostics, Auckland University of Technology, Auckland, New Zealand
| | | | - Huti Watson
- Paratene Ngata Research Centre, Ngati Porou Oranga, Te Puia Springs, New Zealand
| | - Phillip Wilcox
- Maurice Wilkins Centre, Auckland, New Zealand
- Department of Statistics, University of Otago, Dunedin, New Zealand
| | - Rinki Murphy
- Maurice Wilkins Centre, Auckland, New Zealand
- Department of Medicine, University of Auckland, Auckland, New Zealand
- Auckland Diabetes Centre, Te Whatu Ora Health New Zealand, Te Toka Tumai, New Zealand
| | - Troy L Merry
- Maurice Wilkins Centre, Auckland, New Zealand
- Department of Nutrition, University of Auckland, Auckland, New Zealand
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand.
- Maurice Wilkins Centre, Auckland, New Zealand.
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
6
|
Mohammadi M, Salehi S, Habibzadeh A, Mohammadi A, Mirzaasgari Z. Neuroprotective Effects of Metformin in Stroke Patients: A Systematic Review and Meta-analysis of Cohort Studies. Clin Neuropharmacol 2025; 48:51-59. [PMID: 40072880 DOI: 10.1097/wnf.0000000000000625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
OBJECTIVES People with diabetes are 1.5 times more likely to experience stroke than those without diabetes, underlining the urgent need to address this issue. Metformin is often the initial medication chosen to manage diabetes mellitus (DM). The purpose of our systematic review and meta-analysis is to explore the potential neuroprotective effects of metformin in individuals who have received it prior to stroke. METHOD Our study encompassed cohort studies that drew a comparison between the severity and diverse outcomes of stroke among individuals with DM who were administered metformin prior to the stroke event and those with DM who did not receive the treatment. RESULTS Ten studies met the eligibility criteria. Prestroke metformin use was associated with a significantly lower National Institutes of Health Stroke Scale score (mean difference = -1.29, 95% confidence interval: -2.11 to -0.47) in ischemic stroke. Metformin pretreatment in ischemic stroke was associated with increased odds of favorable outcome (mRS < 2) at 90 days (odds ratio [OR] = 1.45, 95% confidence interval [CI]: 1.06 to 1.99), but it was not significant at discharge. Metformin was found to be associated with reduced mortality (OR = 0.52, 95% CI: 0.42 to 0.64) in ischemic stroke. In hemorrhagic stroke, the results showed a significantly lower intracranial hemorrhage volume in prestroke metformin use (mean difference = -4.77, 95% CI: -6.56 to -2.98). CONCLUSIONS We found that prestroke metformin use in diabetic patients yielded neuroprotective effects. In ischemic strokes, metformin reduces stroke severity and 90-day mortality; it also improves 90-day functional outcomes. In hemorrhagic strokes, prestroke metformin use can also cause less intracranial hemorrhage volume. Further clinical trials are needed to confirm its efficacy and verify its benefits in stroke management.
Collapse
Affiliation(s)
| | - Sadaf Salehi
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Aynaz Mohammadi
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Mirzaasgari
- Department of Neurology, Firoozgar Hospital, School of Medicine, Iran University of Medical Science, Fasa, Iran
| |
Collapse
|
7
|
Chen J, Xiang J, Zhou M, Huang R, Zhang J, Cui Y, Jiang X, Li Y, Zhou R, Xin H, Li J, Li L, Lam SM, Zhu J, Chen Y, Yang Q, Xie Z, Shui G, Deng F, Zhang Z, Li MD. Dietary timing enhances exercise by modulating fat-muscle crosstalk via adipocyte AMPKα2 signaling. Cell Metab 2025. [DOI: pmid: 40088888 doi: 10.1016/j.cmet.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
|
8
|
Cheng K, Zhou Y, Hao Y, Wu S, Wang N, Zhang P, Wang Y. Magnolol inhibits appetite and causes visceral fat loss through Growth/differentiation factor-15 (GDF-15) by activating transcription factor 4-CCAAT enhancer binding protein γ-mediated endoplasmic reticulum stress responses. Chin J Nat Med 2025; 23:334-345. [PMID: 40122663 DOI: 10.1016/s1875-5364(25)60835-1] [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: 12/23/2023] [Revised: 03/11/2024] [Accepted: 05/07/2024] [Indexed: 03/25/2025]
Abstract
Magnolol, a compound extracted from Magnolia officinalis, demonstrates potential efficacy in addressing metabolic dysfunction and cardiovascular diseases. Its biological activities encompass anti-inflammatory, antioxidant, anticoagulant, and anti-diabetic effects. Growth/differentiation factor-15 (GDF-15), a member of the transforming growth factor β superfamily, is considered a potential therapeutic target for metabolic disorders. This study investigated the impact of magnolol on GDF-15 production and its underlying mechanism. The research examined the pharmacological effect of magnolol on GDF-15 expression in vitro and in vivo, and determined the involvement of endoplasmic reticulum (ER) stress signaling in this process. Luciferase reporter assays, chromatin immunoprecipitation, and in vitro DNA binding assays were employed to examine the regulation of GDF-15 by activating transcription factor 4 (ATF4), CCAAT enhancer binding protein γ (CEBPG), and CCCTC-binding factor (CTCF). The study also investigated the effect of magnolol and ATF4 on the activity of a putative enhancer located in the intron of the GDF-15 gene, as well as the influence of single nucleotide polymorphisms (SNPs) on magnolol and ATF4-induced transcription activity. Results demonstrated that magnolol triggers GDF-15 production in endothelial cells (ECs), hepatoma cell line G2 (HepG2) and hepatoma cell line 3B (Hep3B) cell lines, and primary mouse hepatocytes. The cooperative binding of ATF4 and CEBPG upstream of the GDF-15 gene or the E1944285 enhancer located in the intron led to full-power transcription of the GDF-15 gene. SNP alleles were found to impact the magnolol and ATF4-induced transcription activity of GDF-15. In high-fat diet ApoE-/- mice, administration of magnolol induced GDF-15 production and partially suppressed appetite through GDF-15. These findings suggest that magnolol regulates GDF-15 expression through priming of promoter and enhancer activity, indicating its potential as a drug for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Keru Cheng
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China; Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Yanyun Zhou
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Yilong Hao
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Shengyun Wu
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Nanping Wang
- Health Science Center, East China Normal University, Shanghai 200241, China
| | - Peng Zhang
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China; Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| | - Yinfang Wang
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China; Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| |
Collapse
|
9
|
Ismail E, Chi X, Bhatta M, Hosford J, Bernier A. The Impact of Artificially Sweetened Drinks on Metformin Efficacy. Nutrients 2025; 17:797. [PMID: 40077667 PMCID: PMC11901812 DOI: 10.3390/nu17050797] [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: 01/15/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Artificially sweetened beverages (ASBs) are commonly recommended as a substitute for sugar-sweetened beverages (SSBs) in dietary counseling. Childhood obesity, associated with comorbidities like type 2 diabetes (T2D), has risen alongside increased consumption of both SSBs and ASBs. Metformin, a common treatment for pediatric T2D, affects GDF-15, a hormone involved in weight regulation. This pilot study examines the impact of ASBs on the therapeutic effects of metformin in pediatric patients with obesity and prediabetes, focusing on growth differentiation factor 15 (GDF-15) as a potential mediator. METHODS Forty-six children aged 10-21 years were randomized into two groups: one consuming non-sweetened beverages (USB) and the other consuming ASBs during a 12-week metformin intervention. RESULTS While the USB group showed a greater decrease in the point estimate for mean BMI (-0.55 ± 1.49 USB vs. -0.23 ± 1.60 ASB) and an increase in the point estimate for mean GDF-15 (33.40 ± 58.34 in USB vs. 19.77 ± 85.87 in ASB), these differences were not statistically significant (p = 0.6). As a hypothesis-generating exercise, change in insulin resistance was explored. While again lacking statistical power, we observed that more participants in the USB group showed improvements in insulin resistance. CONCLUSIONS Additional studies are needed to fully elucidate the impact of non-nutritive sweeteners on metabolic health and treatment outcomes in pediatric obesity.
Collapse
Affiliation(s)
- Esraa Ismail
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (E.I.); (M.B.)
- Division of Pediatric Endocrinology, UF Health, Department of Pediatrics, University of Florida, Gainesville, FL 32608, USA
| | - Xiaofei Chi
- Division of Pediatrics Research Hub (PoRCH), UF Health Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA; (X.C.); (J.H.)
| | - Mallika Bhatta
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (E.I.); (M.B.)
| | - Jennifer Hosford
- Division of Pediatrics Research Hub (PoRCH), UF Health Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA; (X.C.); (J.H.)
| | - Angelina Bernier
- College of Medicine, University of Florida, Gainesville, FL 32610, USA; (E.I.); (M.B.)
- Division of Pediatric Endocrinology, UF Health, Department of Pediatrics, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
10
|
Chen Y, Wang H, Yang M, Shen Z, Gao Y. Exploring the Effects of Metformin on the Body via the Urine Proteome. Biomolecules 2025; 15:241. [PMID: 40001544 PMCID: PMC11853151 DOI: 10.3390/biom15020241] [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: 01/05/2025] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Metformin is the first-line medication for treating type 2 diabetes mellitus, with more than 200 million patients taking it daily. Its effects are extensive and play a positive role in multiple areas. Can its effects and potential mechanisms be explored through the urine proteome? In this study, 166 differential proteins were identified following the administration of 150 mg/(kg·d) of metformin to rats for five consecutive days. These included complement component C6, pyruvate kinase, coagulation factor X, growth differentiation factor 15, carboxypeptidase A4, chymotrypsin-like elastase family member 1, and L-lactate dehydrogenase C chain. Several of these proteins have been reported to be directly affected by metformin or associated with its effects. Multiple biological pathways enriched by these differential proteins, or proteins containing differentially modified peptides, have been reported to be associated with metformin, such as the glutathione metabolic process, negative regulation of gluconeogenesis, and the renin-angiotensin system. Additionally, some significantly changed proteins and enriched biological pathways, not yet reported to be associated with metformin's effects, may provide clues for exploring its potential mechanisms. In conclusion, the application of the urine proteome offers a comprehensive and systematic approach to exploring the effects of drugs, providing a new perspective on the study of metformin's mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Youhe Gao
- Gene Engineering Drug and Biotechnology Beijing Key Laboratory, College of Life Sciences, Beijing Normal University, Beijing 100875, China; (Y.C.); (H.W.); (M.Y.); (Z.S.)
| |
Collapse
|
11
|
Reczek CR, Chakrabarty RP, D'Alessandro KB, Sebo ZL, Grant RA, Gao P, Budinger GR, Chandel NS. Metformin targets mitochondrial complex I to lower blood glucose levels. SCIENCE ADVANCES 2024; 10:eads5466. [PMID: 39693440 DOI: 10.1126/sciadv.ads5466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024]
Abstract
Metformin is among the most prescribed antidiabetic drugs, but the primary molecular mechanism by which metformin lowers blood glucose levels is unknown. Previous studies have proposed numerous mechanisms by which acute metformin lowers blood glucose, including the inhibition of mitochondrial complex I of the electron transport chain (ETC). Here, we used transgenic mice that globally express the Saccharomyces cerevisiae internal alternative NADH dehydrogenase (NDI1) protein to determine whether the glucose-lowering effect of acute oral administration of metformin requires inhibition of mitochondrial complex I of the ETC in vivo. NDI1 is a yeast NADH dehydrogenase enzyme that complements the loss of mammalian mitochondrial complex I electron transport function and is insensitive to pharmacologic mitochondrial complex I inhibitors including metformin. We demonstrate that NDI1 expression attenuates metformin's ability to lower blood glucose levels under standard chow and high-fat diet conditions. Our results indicate that acute oral administration of metformin targets mitochondrial complex I to lower blood glucose.
Collapse
Affiliation(s)
- Colleen R Reczek
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ram P Chakrabarty
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karis B D'Alessandro
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zachary L Sebo
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rogan A Grant
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Peng Gao
- Robert H. Lurie Cancer Center Metabolomics Core, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G R Budinger
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Navdeep S Chandel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Chan Zuckerberg Biohub, Chicago, IL, USA
| |
Collapse
|
12
|
Zhou H, Xiao J, Cheng Q, Wang W, Peng H, Lin X, Chen J, Wang X. Metformin inhibits migration and epithelial-to-mesenchymal transition in non-small cell lung cancer cells through AMPK-mediated GDF15 induction. Eur J Pharmacol 2024; 985:177127. [PMID: 39528101 DOI: 10.1016/j.ejphar.2024.177127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/06/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
The growth differentiation factor 15 (GDF15) may serve as a biomarker of metformin, which mediates the bodyweight lowering effect of metformin. However, whether GDF15 also serves as a molecular target of metformin to inhibit carcinogenesis remains largely unknown. This study examined the role and molecular mechanisms of GDF15 in the anticancer effects of metformin in non-small cell lung cancer (NSCLC) cells, which has never been reported before. We found that metformin significantly inhibited the migration of NSCLC A549 and NCI-H460 cells and reduced the expression of epithelial-to-mesenchymal transition (EMT)-related molecules, including neuro-cadherin (N-cadherin), matrix metalloproteinase 2 (MMP2), and the zinc finger transcription factor Snail, but increased epithelial cadherin (E-cadherin) expression. Furthermore, metformin increased GDF15 and its upstream transcription factors activated transcription factor 4 (ATF4) and C/EBP-homologous protein (CHOP) expressions and increased AMP-activated protein kinase (AMPK) phosphorylation in NSCLC cells. GDF15 siRNA partially reverses the inhibitory effect of metformin on NSCLC cell migration. Moreover, metformin-induced increases in GDF15, CHOP, and ATF4 expression and the inhibition of migration were partially reversed by treatment with Compound C, a specific AMPK inhibitor. Meanwhile, metformin significantly inhibited NCI-H460 xenograft tumor growth in nude mice, increased GDF15 expression, and regulated EMT- and migration-related protein expression in xenograft tumors. In conclusion, our results provide novel insights into revealing that GDF15 can serve as a potential molecular target of metformin owing to its anti-cancer effect in NSCLC, which is mediated by AMPK activation.
Collapse
Affiliation(s)
- Hongyu Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Jun Xiao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Qi Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Wen Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - He Peng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Xiaojian Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Jiajun Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Xingya Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China.
| |
Collapse
|
13
|
Jindal J, Hill J, Harte J, Dunachie SJ, Kronsteiner B. Starvation and infection: The role of sickness-associated anorexia in metabolic adaptation during acute infection. Metabolism 2024; 161:156035. [PMID: 39326837 DOI: 10.1016/j.metabol.2024.156035] [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: 05/17/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Sickness-associated anorexia, the reduction in appetite seen during infection, is a widely conserved and well-recognized symptom of acute infection, yet there is very little understanding of its functional role in recovery. Anorexic sickness behaviours can be understood as an evolutionary strategy to increase tolerance to pathogen-mediated illness. In this review we explore the evidence for mechanisms and potential metabolic benefits of sickness-associated anorexia. Energy intake can impact on the immune response, control of inflammation and tissue stress, and on pathogen fitness. Fasting mediators including hormone-sensitive lipase, peroxisome proliferator-activated receptor-alpha (PPAR-α) and ketone bodies are potential facilitators of infection recovery through multiple pathways including suppression of inflammation, adaptation to lipid utilising pathways, and resistance to pathogen-induced cellular stress. However, the effect and benefit of calorie restriction is highly heterogeneous depending on both the infection and the metabolic status of the host, which has implications regarding clinical recommendations for feeding during different infections.
Collapse
Affiliation(s)
- Jessy Jindal
- The Medical School, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Jennifer Hill
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Jodie Harte
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Susanna J Dunachie
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.
| | - Barbara Kronsteiner
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
14
|
Ye J, Yin J. Type 2 diabetes: a sacrifice program handling energy surplus. LIFE METABOLISM 2024; 3:loae033. [PMID: 39873003 PMCID: PMC11748514 DOI: 10.1093/lifemeta/loae033] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/30/2024] [Accepted: 09/12/2024] [Indexed: 01/30/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is closely associated with obesity, while interactions between the two diseases remain to be fully elucidated. To this point, we offer this perspective to introduce a set of new insights into the interpretation of T2DM spanning the etiology, pathogenesis, and treatment approaches. These include a definition of T2DM as an energy surplus-induced diabetes characterized by the gradual decline of β cell insulin secretion function, which ultimately aims to prevent the onset of severe obesity through mechanisms of weight loss. The body employs three adaptive strategies in response to energy surplus: the first one is adipose tissue expansion to store the energy for weight gain under normal weight conditions; the second one is insulin resistance to slow down adipose tissue expansion and weight gain under overweight conditions; and the third one is the onset of T2DM following β cell failure to reverse the weight gain in obese conditions. The primary signaling molecules driving the compensatory responses are adenosine derivatives, such as adenosine triphosphate (ATP), acetyl coenzyme A (acetyl-CoA), and reduced nicotinamide adenine dinucleotide (NADH). These molecules exert their effects through allosteric, post-translational, and transcriptional regulation of metabolic pathways. The insights suggest that insulin resistance and T2DM are protective mechanisms in the defense against excessive adiposity to avert severe obesity. The perspective provides a unified framework explaining the interactions between the two diseases and opens new avenues in the study of T2DM.
Collapse
Affiliation(s)
- Jianping Ye
- Metabolic Disease Research Center, Zhengzhou Key Laboratory of Obesity Research, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450007, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jun Yin
- Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| |
Collapse
|
15
|
Di Pastena F, Pond G, Tsakiridis EE, Gouveia A, Ahmadi E, Biziotis OD, Ali A, Swaminath A, Okawara G, Ellis PM, Abdulkarim B, Ahmed N, Robinson A, Roa W, Valdes M, Kavsak P, Wierzbicki M, Wright J, Steinberg G, Tsakiridis T. Growth differentiation factor 15 (GDF15) predicts relapse free and overall survival in unresected locally advanced non-small cell lung cancer treated with chemoradiotherapy. Radiat Oncol 2024; 19:155. [PMID: 39511611 PMCID: PMC11542377 DOI: 10.1186/s13014-024-02546-y] [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: 04/14/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024] Open
Abstract
INTRODUCTION Growth differentiation factor 15 (GDF15) is a cytokine of the TGFβ family. Here, we analyzed GDF15 levels in patients with locally advanced non-small cell lung cancer (LA-NSCLC) who participated in OCOG-ALMERA (NCT02115464), a phase II randomized clinical trial, that investigated metformin in combination with standard of care concurrent chemoradiotherapy (cCRT). OCOG-ALMERA was not able to demonstrate benefit in the metformin arm. Therefore, biomarker studies are needed to better define stratification parameters for future trials. METHODS Patients were randomized to treatment with platinum-based chemotherapy and concurrent chest radiotherapy (60-66 Gy), with or without metformin (2000 mg/d). The trial collected tumor volume parameters, survival outcomes, and patient blood plasma at baseline, during (weeks 1 and 6) and 6 months after cCRT. Plasma GDF15 levels were assayed with the ELISA method. Statistical analyses explored associations between GDF15, survival outcomes, and radiotherapy tumor volumes. RESULTS Baseline plasma levels of GDF15 were elevated in study patients, they increased during cCRT (p < 0.001), and the addition of metformin was associated with a further increase (week 6, p = 0.033). Baseline GDF15 levels correlated with the radiotherapy gross target volume (GTV, p < 0.01), while week 1 of radiotherapy levels correlated with radiotherapy planned target volume (PTV, p < 0.006). In multivariate analysis, baseline plasma GDF15 was prognostic for poor relapse-free (RFS) and overall survival (OS) (p = 0.005 and p = 0.002, respectively). CONCLUSIONS GDF15 is a plasma marker that responds to the treatment of unresected LA-NSCLC with cCRT and metformin. GDF15 levels correspond with tumor volume and increased GDF15 levels predict for poor RFS and OS. These results require validation in larger clinical trial datasets.
Collapse
Affiliation(s)
- Fiorella Di Pastena
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - Gregory Pond
- Department of Oncology, McMaster University, Hamilton, ON, Canada
- Ontario Clinical Oncology Group, McMaster University, Hamilton, ON, Canada
| | - Evangelia E Tsakiridis
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - Andre Gouveia
- Department of Oncology, McMaster University, Hamilton, ON, Canada
- Radiation Oncology, Juravinski Cancer Center, Hamilton Health Science, Hamilton, ON, Canada
| | - Elham Ahmadi
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada
- Department of Oncology, McMaster University, Hamilton, ON, Canada
- Center for Discovery and Cancer Research, McMaster University, Hamilton, ON, Canada
| | - Olga-Demetra Biziotis
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada
- Department of Oncology, McMaster University, Hamilton, ON, Canada
- Center for Discovery and Cancer Research, McMaster University, Hamilton, ON, Canada
| | - Amr Ali
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada
- Department of Oncology, McMaster University, Hamilton, ON, Canada
- Center for Discovery and Cancer Research, McMaster University, Hamilton, ON, Canada
| | - Anand Swaminath
- Department of Oncology, McMaster University, Hamilton, ON, Canada
- Radiation Oncology, Juravinski Cancer Center, Hamilton Health Science, Hamilton, ON, Canada
| | - Gordon Okawara
- Department of Oncology, McMaster University, Hamilton, ON, Canada
- Radiation Oncology, Juravinski Cancer Center, Hamilton Health Science, Hamilton, ON, Canada
| | - Peter M Ellis
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| | | | | | | | - Wilson Roa
- Cross Cancer Institute, Edmonton, AB, Canada
| | - Mario Valdes
- Grand River Cancer Center, Kitchener, ON, Canada
| | - Peter Kavsak
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Marcin Wierzbicki
- Radiation Physics Program, Juravinski Cancer Centre, Hamilton, ON, Canada
| | - James Wright
- Department of Oncology, McMaster University, Hamilton, ON, Canada
- Ontario Clinical Oncology Group, McMaster University, Hamilton, ON, Canada
- Radiation Oncology, Juravinski Cancer Center, Hamilton Health Science, Hamilton, ON, Canada
| | - Gregory Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Theodoros Tsakiridis
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada.
- Department of Oncology, McMaster University, Hamilton, ON, Canada.
- Ontario Clinical Oncology Group, McMaster University, Hamilton, ON, Canada.
- Radiation Oncology, Juravinski Cancer Center, Hamilton Health Science, Hamilton, ON, Canada.
- Center for Discovery and Cancer Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
16
|
Wang Y, Chen Y, Liang X, Zhu L, Wen X. Network pharmacology and transcriptomics explore the therapeutic effects of Ermiao Wan categorized formulas for diabetes in mice. Sci Rep 2024; 14:27014. [PMID: 39506066 PMCID: PMC11541784 DOI: 10.1038/s41598-024-78364-9] [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: 07/02/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024] Open
Abstract
Ermiao wan (EMW) is a classical traditional Chinese medicine formula, with two modified versions including Sanmiao wan (SMW) and Simiao wan (FMW). These Ermiao wan categorized formulas (ECFs) are traditionally used to treat gouty arthritis and hyperuricemia. However, their potential benefits and mechanisms on diabetes remain to be explored. This study aims to investigate the overall effects and biological differences of ECFs in high fat diet (HFD)-fed mice based on network pharmacology and transcriptomics. ECFs significantly reduced body weight, improved oral glucose tolerance, decreased fat accumulation, and lowered serum insulin and inflammatory cytokine levels in HFD-fed mice. FMW had better efficacy than EMW and SMW. Network pharmacology analysis revealed that ECFs targeted functional modules associated with chronic inflammation, lipid metabolism, and glucose metabolism. Transcriptome results also showed ECFs could inhibit genes associated with inflammation and upregulated some genes in lipid metabolism. Comprehensive analysis and QPCR verification indicated the beneficial effects of ECFs on diabetes might be attributed to the regulation of Ddit3, Ccl2, Esr1, and Cyp7a1. This study provides a theoretical basis for the clinical use of ECFs.
Collapse
Affiliation(s)
- Yuping Wang
- Pukou Hospital of Chinese Medicine affiliated to China Pharmaceutical University, China Pharmaceutical University, 639 Longmian road, Nanjing, China
| | - Yimeng Chen
- Pukou Hospital of Chinese Medicine affiliated to China Pharmaceutical University, China Pharmaceutical University, 639 Longmian road, Nanjing, China
| | - Xinyi Liang
- Pukou Hospital of Chinese Medicine affiliated to China Pharmaceutical University, China Pharmaceutical University, 639 Longmian road, Nanjing, China
| | - Lijuan Zhu
- Pukou Hospital of Chinese Medicine affiliated to China Pharmaceutical University, China Pharmaceutical University, 639 Longmian road, Nanjing, China
| | - Xiaodong Wen
- Pukou Hospital of Chinese Medicine affiliated to China Pharmaceutical University, China Pharmaceutical University, 639 Longmian road, Nanjing, China.
| |
Collapse
|
17
|
Li Y, Zhang J, Chen S, Ke Y, Li Y, Chen Y. Growth differentiation factor 15: Emerging role in liver diseases. Cytokine 2024; 182:156727. [PMID: 39111112 DOI: 10.1016/j.cyto.2024.156727] [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: 06/05/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/25/2024]
Abstract
Growth differentiation factor 15 (GDF15) is a cell stress-response cytokine within the transforming growth factor-β (TGFβ) superfamily. It is known to exert diverse effects on many metabolic pathways through its receptor GFRAL, which is expressed in the hindbrain, and transduces signals through the downstream receptor tyrosine kinase Ret. Since the liver is the core organ of metabolism, summarizing the functions of GDF15 is highly important. In this review, we assessed the relevant literature regarding the main metabolic, inflammatory, fibrogenic, tumorigenic and other effects of GDF15 on different liver diseases, including Metabolic dysfunction-associated steatotic liver disease(MASLD), alcohol and drug-induced liver injury, as well as autoimmune and viral hepatitis, with a particular focus on the pathogenesis of MASLD progression from hepatic steatosis to MASH, liver fibrosis and even hepatocellular carcinoma (HCC). Finally, we discuss the prospects of the clinical application potential of GDF15 along with its research and development progress. With better knowledge of GDF15, increasing in-depth research will lead to a new era in the field of liver diseases.
Collapse
Affiliation(s)
- Yu Li
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jie Zhang
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Shurong Chen
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yini Ke
- Department of Rheumatology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Youming Li
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yi Chen
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
18
|
Maya SA, Shumon MAH, Islam MR, Khan JM, Khan SA, Rana S, Hoque MA, Rahman MM. Investigation of the impacts of several diols on the phase behavior and physico-chemical quantities associated with the Triton X-100 and antidiabetic drug mixture. J INDIAN CHEM SOC 2024; 101:101295. [DOI: 10.1016/j.jics.2024.101295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
19
|
Wang Q, Farhadipour M, Thijs T, Ruilova Sosoranga E, Van der Schueren B, Ceulemans LJ, Deleus E, Lannoo M, Tack J, Depoortere I. Bitter-tasting drugs tune GDF15 and GLP-1 expression via bitter taste or motilin receptors in the intestine of patients with obesity. Mol Metab 2024; 88:102002. [PMID: 39111389 PMCID: PMC11380393 DOI: 10.1016/j.molmet.2024.102002] [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: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024] Open
Abstract
OBJECTIVE Growth differentiation factor 15 (GDF15), a stress related cytokine, was recently identified as a novel satiety signal acting via the GFRAL receptor located in the hindbrain. Bitter compounds are known to induce satiety via the release of glucagon-like peptide 1 (GLP-1) through activation of bitter taste receptors (TAS2Rs, 25 subtypes) on enteroendocrine cells in the gut. This study aimed to investigate whether and how bitter compounds induce a stress response in intestinal epithelial cells to affect GDF15 expression in patients with obesity, thereby facilitating satiety signaling from the gut. METHODS The acute effect of oral intake of the bitter-containing medication Plaquenil (hydroxychloroquine sulfate) on plasma GDF15 levels was evaluated in a placebo-controlled, double-blind, randomized, two-visit crossover study in healthy volunteers. Primary crypts isolated from the jejunal mucosa from patients with obesity were stimulated with vehicle or bitter compounds, and the effect on GDF15 expression was evaluated using RT-qPCR or ELISA. Immunofluorescence colocalization studies were performed between GDF15, epithelial cell type markers and TAS2Rs. The role of TAS2Rs was tested by 1) pretreatment with a TAS2R antagonist, GIV3727; 2) determining TAS2R4/43 polymorphisms that affect taste sensitivity to TAS2R4/43 agonists. RESULTS Acute intake of hydroxychloroquine sulfate increased GDF15 plasma levels, which correlated with reduced hunger scores and plasma ghrelin levels in healthy volunteers. This effect was mimicked in primary jejunal cultures from patients with obesity. GDF15 was expressed in enteroendocrine and goblet cells with higher expression levels in patients with obesity. Various bitter-tasting compounds (medicinal, plant extracts, bacterial) either increased or decreased GDF15 expression, with some also affecting GLP-1. The effect was mediated by specific intestinal TAS2R subtypes and the unfolded protein response pathway. The bitter-induced effect on GDF15/GLP-1 expression was influenced by the existence of TAS2R4 amino acid polymorphisms and TAS2R43 deletion polymorphisms that may predict patient's therapeutic responsiveness. However, the effect of the bitter-tasting antibiotic azithromycin on GDF15 release was mediated via the motilin receptor, possibly explaining some of its aversive side effects. CONCLUSIONS Bitter chemosensory and pharmacological receptors regulate the release of GDF15 from human gut epithelial cells and represent potential targets for modulating metabolic disorders or cachexia.
Collapse
Affiliation(s)
- Qian Wang
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Mona Farhadipour
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Theo Thijs
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | | | - Bart Van der Schueren
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Clinical and Experimental Endocrinology, University of Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Leuven Intestinal Failure and Transplantation (LIFT) Center, University Hospitals Leuven, Leuven, Belgium
| | - Ellen Deleus
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Matthias Lannoo
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Inge Depoortere
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium.
| |
Collapse
|
20
|
Magalhães FMV, Pestana RMC, Ferreira CN, Silva IFO, Candido AL, Oliveira FR, Reis FM, Gomes KB. GDF-15 levels in patients with polycystic ovary syndrome treated with metformin: a combined clinical and in silico pathway analysis. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e230416. [PMID: 39420932 PMCID: PMC11460967 DOI: 10.20945/2359-4292-2023-0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/01/2024] [Indexed: 10/19/2024]
Abstract
Objective Polycystic ovary syndrome (PCOS) is an endocrine disease characterized by metabolic, reproductive, and psychological manifestations. Growth and differentiation factor 15 (GDF-15) is a cytokine associated with metabolic and inflammatory disorders. Metformin is commonly used for the treatment of PCOS. We investigated the relationship between GDF-15 levels and PCOS, the effect of metformin on GDF-15 levels, and potential biologic pathways related to GDF-15. Subjects and methods The study included 35 women with PCOS and 32 women without PCOS (controls). Both groups were compared in terms of GDF-15 levels. Additional analysis was conducted on samples from 22 women with PCOS who were treated with either metformin (n = 7) or placebo (n = 15), retrieved from a previous randomized, controlled trial. Levels of GDF-15 were measured using MILLIPLEX. The biologic pathways related to GDF-15 were evaluated using the databases STRING, SIGNOR, and Pathway Commons. The statistical analysis was conducted using the software SPSS. Results Levels of GDF-15 were higher in the PCOS group compared with the non-PCOS group (p = 0.039). Among women with PCOS, GDF-15 levels were higher in those treated with metformin compared with placebo (p = 0.007). The proteins related to GDF-15 overlapped between the databases, and a significant interaction was found between GDF-15 and proteins related to PCOS and its complications, including those related to estrogen response, oxidative stress, ovarian infertility, interleukin (IL)-18, IL-4, the ratio of advanced glycation end products to their receptor (AGE/RAGE), leptin, transforming growth factor beta (TGF-β), adipogenesis, and insulin. Conclusion The findings of the present study suggest a relationship between GDF-15 and PCOS and a potential increase in GDF-15 levels with metformin treatment. An additional finding was that GDF-15 could be involved in biologic pathways related to PCOS complications.
Collapse
Affiliation(s)
- Fernanda M V Magalhães
- Faculdade de Farmácia Universidade Federal de Minas Gerais Belo HorizonteMG Brasil Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Rodrigo M C Pestana
- Faculdade de Medicina Universidade Federal de Minas Gerais Belo HorizonteMG Brasil Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Cláudia N Ferreira
- Colégio Técnico Universidade Federal de Minas Gerais Belo HorizonteMG Brasil Colégio Técnico, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Ieda F O Silva
- Faculdade de Farmácia Universidade Federal de Minas Gerais Belo HorizonteMG Brasil Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Ana L Candido
- Faculdade de Medicina Universidade Federal de Minas Gerais Belo HorizonteMG Brasil Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Flávia R Oliveira
- Faculdade de Medicina Universidade Federal de Minas Gerais Belo HorizonteMG Brasil Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Fernando M Reis
- Faculdade de Medicina Universidade Federal de Minas Gerais Belo HorizonteMG Brasil Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Karina B Gomes
- Faculdade de Farmácia Universidade Federal de Minas Gerais Belo HorizonteMG Brasil Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| |
Collapse
|
21
|
Sirtori CR, Castiglione S, Pavanello C. METFORMIN: FROM DIABETES TO CANCER TO PROLONGATION OF LIFE. Pharmacol Res 2024; 208:107367. [PMID: 39191336 DOI: 10.1016/j.phrs.2024.107367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
The metformin molecule dates back to over a century, but its clinical use started in the '50s. Since then, its use in diabetics has grown constantly, with over 150 million users today. The therapeutic profile also expanded, with improved understanding of novel mechanisms. Metformin has a major activity on insulin resistance, by acting on the insulin receptors and mitochondria, most likely by activation of the adenosine monophosphate-activated kinase. These and associated mechanisms lead to significant lipid lowering and body weight loss. An anti-cancer action has come up in recent years, with mechanisms partly dependent on the mitochondrial activity and also on phosphatidylinositol 3-kinase resistance occurring in some malignant tumors. The potential of metformin to raise life-length is the object of large ongoing studies and of several basic and clinical investigations. The present review article will attempt to investigate the basic mechanisms behind these diverse activities and the potential clinical benefits. Metformin may act on transcriptional activity by histone modification, DNA methylation and miRNAs. An activity on age-associated inflammation (inflammaging) may occur via activation of the nuclear factor erythroid 2 related factor and changes in gut microbiota. A senolytic activity, leading to reduction of cells with the senescent associated secretory phenotype, may be crucial in lifespan prolongation as well as in ancillary properties in age-associated diseases, such as Parkinson's disease. Telomere prolongation may be related to the activity on mitochondrial respiratory factor 1 and on peroxisome gamma proliferator coactivator 1-alpha. Very recent observations on the potential to act on the most severe neurological disorders, such as amyotrophic lateral sclerosis and frontotemporal dementia, have raised considerable hope.
Collapse
Affiliation(s)
- Cesare R Sirtori
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Sofia Castiglione
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Pavanello
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
22
|
Hurtado-Carneiro V, LeBaut-Ayuso Y, Velázquez E, Flores-Lamas C, Fernández-de la Rosa R, García-García L, Gómez-Oliver F, Ruiz-Albusac JM, Pozo MÁ. Effects of chronic treatment with metformin on brain glucose hypometabolism and central insulin actions in transgenic mice with tauopathy. Heliyon 2024; 10:e35752. [PMID: 39170185 PMCID: PMC11337050 DOI: 10.1016/j.heliyon.2024.e35752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Brain glucose hypometabolism and insulin alterations are common features of many neurological diseases. Herein we sought to corroborate the brain glucose hypometabolism that develops with ageing in 12-months old Tau-VLW transgenic mice, a model of tauopathy, as well as to determine whether this model showed signs of altered peripheral glucose metabolism. Our results demonstrated that 12-old months Tau mice exhibited brain glucose hypometabolism as well as basal hyperglycemia, impaired glucose tolerance, hyperinsulinemia, and signs of insulin resistance. Then, we further studied the effect of chronic metformin treatment (9 months) in Tau-VLW mice from 9 to 18 months of age. Longitudinal PET neuroimaging studies revealed that chronic metformin altered the temporal profile in the progression of brain glucose hypometabolism associated with ageing. Besides, metformin altered the content and/or phosphorylation of key components of the insulin signal transduction pathway in the frontal cortex leading to significant changes in the content of the active forms. Thus, metformin increased the expression of pAKT-Y474 while reducing pmTOR-S2448 and pGSK3β. These changes might be related, at least partially, to a slow progression of ageing, neurological damage, and cognitive decline. Metformin also improved the peripheral glucose tolerance and the ability of the Tau-VLW mice to maintain their body weight through ageing. Altogether our study shows that the tau-VLW mice could be a useful model to study the potential interrelationship between tauopathy and central and peripheral glucose metabolism alterations. More importantly our results suggest that chronic metformin treatment may have direct beneficial central effects by post-transcriptional modulation of key components of the insulin signal transduction pathway.
Collapse
Affiliation(s)
| | - Yannick LeBaut-Ayuso
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Esther Velázquez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Cinthya Flores-Lamas
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | | | - Luis García-García
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University, Madrid, Spain
| | - Francisca Gómez-Oliver
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University, Madrid, Spain
| | - Juan Miguel Ruiz-Albusac
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Miguel Ángel Pozo
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
- Pluridisciplinary Institute, Complutense University, IdISSC, Madrid, Spain
| |
Collapse
|
23
|
Cui R, Wei Y, Liu J, Wang G. Hepatic insulin resistance affects the efficacy of metformin in patients with newly diagnosed type 2 diabetes: A Sub-Analysis of the MARCH trial. Diabetes Res Clin Pract 2024; 214:111788. [PMID: 39032659 DOI: 10.1016/j.diabres.2024.111788] [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: 02/05/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
AIM Our study aimed to analyze how hepatic insulin resistance (IR) influences the efficacy of 48 weeks of metformin treatment in newly diagnosed type 2 diabetes patients. METHODS We chose 291 participants who were allocated to a 48-week metformin treatment in the "Metformin and Acarbose in Chinese as initial Hypoglycemic treatment" (MARCH) trial and calculated their hepatic insulin resistance indexes (HIRI). We equally grouped the subjects into tertiles: low, medium, and high HIRI groups based on baseline HIRI; Low, medium, and high ΔHIRI groups based on the decreasing extent of HIRI after a 48-week metformin treatment. RESULTS Multiple linear regression showed that baseline HIRI was positively associated with the rising degree of Matsuda index and the falling range of fasting blood glucose, fasting insulin, homeostasis model assessment of insulin resistance (HOMA-IR), and HIRI. Furthermore, baseline fasting insulin, homeostatic model assessment of β cell function (HOMA-β), HOMA-IR, and HIRI were positively associated with the decreasing extent of HIRI, while baseline Matsuda index had a negative association with the falling extent of HIRI. CONCLUSIONS Patients with higher levels of hepatic IR obtained better curative effects from metformin in terms of glycemic control, insulin saving, insulin sensitivity enhancement, and IR improvement. Higher fasting blood glucose, fasting insulin, HOMA-β, IR, and lower Matsuda index were indicators of better hepatic IR improvement.
Collapse
Affiliation(s)
- Ruixiang Cui
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China
| | - Ying Wei
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China
| | - Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China.
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China.
| |
Collapse
|
24
|
Bruce K, Garrido AN, Zhang SY, Lam TKT. Regulation of Energy and Glucose Homeostasis by the Nucleus of the Solitary Tract and the Area Postrema. Endocrinol Metab (Seoul) 2024; 39:559-568. [PMID: 39086274 PMCID: PMC11377841 DOI: 10.3803/enm.2024.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/07/2024] [Indexed: 08/02/2024] Open
Abstract
The central nervous system regulates feeding, weight and glucose homeostasis in rodents and humans, but the site-specific mechanisms remain unclear. The dorsal vagal complex in the brainstem that contains the nucleus of the solitary tract (NTS) and area postrema (AP) emerges as a regulatory center that impacts energy and glucose balance by monitoring hormonal and nutrient changes. However, the specific mechanistic metabolic roles of the NTS and AP remain elusive. This mini-review highlights methods to study their distinct roles and recent findings on their metabolic differences and similarities of growth differentiation factor 15 (GDF15) action and glucose sensing in the NTS and AP. In summary, future research aims to characterize hormonal and glucose sensing mechanisms in the AP and/or NTS carries potential to unveil novel targets that lower weight and glucose levels in obesity and diabetes.
Collapse
Affiliation(s)
- Kyla Bruce
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada
| | - Ameth N Garrido
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Song-Yang Zhang
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada
| | - Tony K T Lam
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, Medicine, University of Toronto, Toronto, ON, Canada
- Banting and Best Diabetes Center, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
Petrie JR. Metformin beyond type 2 diabetes: Emerging and potential new indications. Diabetes Obes Metab 2024; 26 Suppl 3:31-41. [PMID: 38965738 DOI: 10.1111/dom.15756] [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: 04/05/2024] [Revised: 06/16/2024] [Accepted: 06/16/2024] [Indexed: 07/06/2024]
Abstract
Metformin is best known as a foundational therapy for type 2 diabetes but is also used in other contexts in clinical medicine with a number of emerging and potential indications. Many of its beneficial effects may be mediated by modest effects on weight loss and insulin sensitivity, but it has multiple other known mechanisms of action. Current clinical uses beyond type 2 diabetes include: polycystic ovarian syndrome; diabetes in pregnancy/gestational diabetes; prevention of type 2 diabetes in prediabetes; and adjunct therapy in type 1 diabetes. As metformin has been in clinical use for almost 70 years, much of the underpinning evidence for its use in these conditions is, by definition, based on trials conducted before the advent of contemporary evidence-based medicine. As a result, some of the above-established uses are 'off-label' in many regulatory territories and their use varies accordingly in different countries. Going forward, several current 'repurposing' investigational uses of metformin are also being investigated: prevention of cancer (including in Li Fraumeni syndrome), renal protection, Alzheimer's disease, metabolic dysfunction-associated steatotic liver disease and promotion of healthy ageing. Despite the longevity of metformin and its important current roles beyond type 2 diabetes in clinical medicine, it has further potential and much research is ongoing.
Collapse
Affiliation(s)
- John R Petrie
- School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| |
Collapse
|
26
|
Hoekx CA, Straat ME, Bizino MB, van Eyk HJ, Lamb HJ, Smit JWA, Jazet IM, de Jager SCA, Boon MR, Martinez‐Tellez B. Growth differentiation factor 15 is not modified after weight loss induced by liraglutide in South Asians and Europids with type 2 diabetes mellitus. Exp Physiol 2024; 109:1292-1304. [PMID: 38965822 PMCID: PMC11291866 DOI: 10.1113/ep091815] [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: 02/21/2024] [Accepted: 05/22/2024] [Indexed: 07/06/2024]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists induce weight loss in patients with type 2 diabetes mellitus (T2DM), but the underlying mechanism is unclear. Recently, the mechanism by which metformin induces weight loss could be explained by an increase in growth differentiation factor 15 (GDF15), which suppresses appetite. Therefore, we aimed to investigate whether the GLP-1R agonist liraglutide modifies plasma GDF15 levels in patients with T2DM. GDF15 levels were measured in plasma samples obtained from Dutch Europids and Dutch South Asians with T2DM before and after 26 weeks of treatment with daily liraglutide (n = 44) or placebo (n = 50) added to standard care. At baseline, circulating GDF15 levels did not differ between South Asians and Europids with T2DM. Treatment with liraglutide, compared to placebo, decreased body weight, but did not modify plasma GDF15 levels in all patients, or when data were split by ethnicity. Also, the change in plasma GDF15 levels after treatment with liraglutide did not correlate with changes in body weight or HbA1c levels. In addition, the dose of metformin used did not correlate with baseline plasma GDF15 levels. Compared to placebo, liraglutide treatment for 26 weeks does not modify plasma GDF15 levels in Dutch Europid or South Asian patients with T2DM. Thus, the weight loss induced by liraglutide is likely explained by other mechanisms beyond the GDF15 pathway. HIGHLIGHTS: What is the central question of this study? Growth differentiation factor 15 (GDF15) suppresses appetite and is increased by metformin: does the GLP-1R agonist liraglutide modify plasma GDF15 levels in patients with type 2 diabetes mellitus (T2DM)? What is the main finding and its importance? Plasma GDF15 levels did not differ between South Asians and Europids with T2DM and were not modified by 26 weeks of liraglutide in either ethnicity. Moreover, there was no correlation between the changes in plasma GDF15 levels and dosage of metformin administered, changes in body weight or HbA1c levels. The appetite-suppressing effect of liraglutide is likely exerted via pathways other than GDF15.
Collapse
Affiliation(s)
- Carlijn A. Hoekx
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Maaike E. Straat
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Maurice B. Bizino
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Huub J. van Eyk
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | | | - Johannes W. A. Smit
- Department of MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Ingrid M. Jazet
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Saskia C. A. de Jager
- Laboratory of Translational ImmunologyUniversity Medical Centre UtrechtUtrechtThe Netherlands
| | - Mariëtte R. Boon
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Borja Martinez‐Tellez
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of Nursing Physiotherapy and Medicine, SPORT Research Group (CTS‐1024), CERNEP Research CenterUniversity of AlmeríaAlmeríaSpain
- Biomedical Research UnitTorrecárdenas University HospitalAlmeríaSpain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN)Instituto de Salud Carlos IIIGranadaSpain
| |
Collapse
|
27
|
Varghese A, Gusarov I, Gamallo-Lana B, Dolgonos D, Mankan Y, Shamovsky I, Phan M, Jones R, Gomez-Jenkins M, White E, Wang R, Jones D, Papagiannakopoulos T, Pacold ME, Mar AC, Littman DR, Nudler E. Unraveling cysteine deficiency-associated rapid weight loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605703. [PMID: 39131293 PMCID: PMC11312522 DOI: 10.1101/2024.07.30.605703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Forty percent of the US population and 1 in 6 individuals worldwide are obese, and the incidence of this disease is surging globally1,2. Various dietary interventions, including carbohydrate and fat restriction, and more recently amino acid restriction, have been explored to combat this epidemic3-6. We sought to investigate the impact of removing individual amino acids on the weight profiles of mice. Compared to essential amino acid restriction, induction of conditional cysteine restriction resulted in the most dramatic weight loss, amounting to 20% within 3 days and 30% within one week, which was readily reversed. This weight loss occurred despite the presence of substantial cysteine reserves stored in glutathione (GSH) across various tissues7. Further analysis demonstrated that the weight reduction primarily stemmed from an increase in the utilization of fat mass, while locomotion, circadian rhythm and histological appearance of multiple other tissues remained largely unaffected. Cysteine deficiency activated the integrated stress response (ISR) and NRF2-mediated oxidative stress response (OSR), which amplify each other, leading to the induction of GDF15 and FGF21, hormones associated with increased lipolysis, energy homeostasis and food aversion8-10. We additionally observed rapid tissue coenzyme A (CoA) depletion, resulting in energetically inefficient anaerobic glycolysis and TCA cycle, with sustained urinary excretion of pyruvate, orotate, citrate, α-ketoglutarate, nitrogen rich compounds and amino acids. In summary, our investigation highlights that cysteine restriction, by depleting GSH and CoA, exerts a maximal impact on weight loss, metabolism, and stress signaling compared to other amino acid restrictions. These findings may pave the way for innovative strategies for addressing a range of metabolic diseases and the growing obesity crisis.
Collapse
Affiliation(s)
- Alan Varghese
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ivan Gusarov
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Begoña Gamallo-Lana
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU School of Medicine, New York, NY 10016, USA
| | - Daria Dolgonos
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Yatin Mankan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ilya Shamovsky
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Mydia Phan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Rebecca Jones
- Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Maria Gomez-Jenkins
- Rutgers Cancer Institute, Rutgers University, New Brunswick, NJ 08901, USA and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Eileen White
- Rutgers Cancer Institute, Rutgers University, New Brunswick, NJ 08901, USA and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Rui Wang
- Department of Biology, York University, Toronto, Ontario, M3J 1P3, Canada
| | - Drew Jones
- Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Michael E Pacold
- Department of Radiation Oncology and Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, NY 10016, USA
| | - Adam C Mar
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU School of Medicine, New York, NY 10016, USA
| | - Dan R Littman
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| |
Collapse
|
28
|
Silva-Bermudez LS, Klüter H, Kzhyshkowska JG. Macrophages as a Source and Target of GDF-15. Int J Mol Sci 2024; 25:7313. [PMID: 39000420 PMCID: PMC11242731 DOI: 10.3390/ijms25137313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024] Open
Abstract
Growth differentiation factor 15 (GDF-15) is a multifunctional cytokine that belongs to the transforming growth factor-beta (TGF-β) superfamily. GDF-15 is involved in immune tolerance and is elevated in several acute and chronic stress conditions, often correlating with disease severity and patient prognosis in cancer172 and metabolic and cardiovascular disorders. Despite these clinical associations, the molecular mechanisms orchestrating its effects remain to be elucidated. The effects of GDF-15 are pleiotropic but cell-specific and dependent on the microenvironment. While GDF-15 expression can be stimulated by inflammatory mediators, its predominant effects were reported as anti-inflammatory and pro-fibrotic. The role of GDF-15 in the macrophage system has been increasingly investigated in recent years. Macrophages produce high levels of GDF-15 during oxidative and lysosomal stress, which can lead to fibrogenesis and angiogenesis at the tissue level. At the same time, macrophages can respond to GDF-15 by switching their phenotype to a tolerogenic one. Several GDF-15-based therapies are under development, including GDF-15 analogs/mimetics and GDF-15-targeting monoclonal antibodies. In this review, we summarize the major physiological and pathological contexts in which GDF-15 interacts with macrophages. We also discuss the major challenges and future perspectives in the therapeutic translation of GDF-15.
Collapse
Affiliation(s)
- Lina Susana Silva-Bermudez
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (L.S.S.-B.); (H.K.)
- German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany
| | - Harald Klüter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (L.S.S.-B.); (H.K.)
- German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany
| | - Julia G. Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (L.S.S.-B.); (H.K.)
- German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany
| |
Collapse
|
29
|
Li J, Hu X, Xie Z, Li J, Huang C, Huang Y. Overview of growth differentiation factor 15 (GDF15) in metabolic diseases. Biomed Pharmacother 2024; 176:116809. [PMID: 38810400 DOI: 10.1016/j.biopha.2024.116809] [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: 03/18/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
GDF15 is a stress response cytokine and a distant member of the transforming growth factor beta (TGFβ) superfamily, its levels increase in response to cell stress and certain diseases in the serum. To exert its effects, GDF15 binds to glial-derived neurotrophic factor (GDNF) receptor alpha-like (GFRAL), which was firstly identified in 2017 and highly expressed in the brain stem. Many studies have demonstrated that elevated serum GDF15 is associated with anorexia and weight loss. Herein, we focus on the biology of GDF15, specifically how this circulating protein regulates appetite and metabolism in influencing energy homeostasis through its actions on hindbrain neurons to shed light on its impact on diseases such as obesity and anorexia/cachexia syndromes. It works as an endocrine factor and transmits metabolic signals leading to weight reduction effects by directly reducing appetite and indirectly affecting food intake through complex mechanisms, which could be a promising target for the treatment of energy-intake disorders.
Collapse
Affiliation(s)
- Jian Li
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, China
| | - Xiangjun Hu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Zichuan Xie
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Jiajin Li
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Chen Huang
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Huang
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
30
|
Tang Y, Yao T, Tian X, Xia X, Huang X, Qin Z, Shen Z, Zhao L, Zhao Y, Diao B, Ping Y, Zheng X, Xu Y, Chen H, Qian T, Ma T, Zhou B, Xu S, Zhou Q, Liu Y, Shao M, Chen W, Shan B, Wu Y. Hepatic IRE1α-XBP1 signaling promotes GDF15-mediated anorexia and body weight loss in chemotherapy. J Exp Med 2024; 221:e20231395. [PMID: 38695876 PMCID: PMC11070642 DOI: 10.1084/jem.20231395] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 02/26/2024] [Accepted: 04/02/2024] [Indexed: 05/08/2024] Open
Abstract
Platinum-based chemotherapy drugs can lead to the development of anorexia, a detrimental effect on the overall health of cancer patients. However, managing chemotherapy-induced anorexia and subsequent weight loss remains challenging due to limited effective therapeutic strategies. Growth differentiation factor 15 (GDF15) has recently gained significant attention in the context of chemotherapy-induced anorexia. Here, we report that hepatic GDF15 plays a crucial role in regulating body weight in response to chemo drugs cisplatin and doxorubicin. Cisplatin and doxorubicin treatments induce hepatic Gdf15 expression and elevate circulating GDF15 levels, leading to hunger suppression and subsequent weight loss. Mechanistically, selective activation by chemotherapy of hepatic IRE1α-XBP1 pathway of the unfolded protein response (UPR) upregulates Gdf15 expression. Genetic and pharmacological inactivation of IRE1α is sufficient to ameliorate chemotherapy-induced anorexia and body weight loss. These results identify hepatic IRE1α as a molecular driver of GDF15-mediated anorexia and suggest that blocking IRE1α RNase activity offers a therapeutic strategy to alleviate the adverse anorexia effects in chemotherapy.
Collapse
Affiliation(s)
- Yuexiao Tang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Tao Yao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Tian
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xintong Xia
- Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xingxiao Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhewen Qin
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhong Shen
- Department of Coloproctology, Hangzhou Third People’s Hospital, Hangzhou, China
| | - Lin Zhao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yaping Zhao
- Division of Life Sciences and Medicine, Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Bowen Diao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yan Ping
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoxiao Zheng
- Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yonghao Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Hui Chen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Tao Qian
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Ma
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ben Zhou
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Suowen Xu
- Division of Life Sciences and Medicine, Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Qimin Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Mengle Shao
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Wei Chen
- Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Shan
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Ying Wu
- Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
31
|
Richter MM, Kemp IM, Heebøll S, Winther-Sørensen M, Kjeldsen SAS, Jensen NJ, Nybing JD, Linden FH, Høgh-Schmidt E, Boesen MP, Madsbad S, Schiødt FV, Nørgaard K, Schmidt S, Gluud LL, Haugaard SB, Holst JJ, Nielsen S, Rungby J, Wewer Albrechtsen NJ. Glucagon augments the secretion of FGF21 and GDF15 in MASLD by indirect mechanisms. Metabolism 2024; 156:155915. [PMID: 38631460 DOI: 10.1016/j.metabol.2024.155915] [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: 12/28/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION Glucagon receptor agonism is currently explored for the treatment of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD). The metabolic effects of glucagon receptor agonism may in part be mediated by increases in circulating levels of Fibroblast Growth Factor 21 (FGF21) and Growth Differentiation Factor 15 (GDF15). The effect of glucagon agonism on FGF21 and GDF15 levels remains uncertain, especially in the context of elevated insulin levels commonly observed in metabolic diseases. METHODS We investigated the effect of a single bolus of glucagon and a continuous infusion of glucagon on plasma concentrations of FGF21 and GDF15 in conditions of endogenous low or high insulin levels. The studies included individuals with overweight with and without MASLD, healthy controls (CON) and individuals with type 1 diabetes (T1D). The direct effect of glucagon on FGF21 and GDF15 was evaluated using our in-house developed isolated perfused mouse liver model. RESULTS FGF21 and GDF15 correlated with plasma levels of insulin, but not glucagon, and their secretion was highly increased in MASLD compared with CON and T1D. Furthermore, FGF21 levels in individuals with overweight with or without MASLD did not increase after glucagon stimulation when insulin levels were kept constant. FGF21 and GDF15 levels were unaffected by direct stimulation with glucagon in the isolated perfused mouse liver. CONCLUSION The glucagon-induced secretion of FGF21 and GDF15 is augmented in MASLD and may depend on insulin. Thus, glucagon receptor agonism may augment its metabolic benefits in patients with MASLD through enhanced secretion of FGF21 and GDF15.
Collapse
Affiliation(s)
- Michael M Richter
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Ida M Kemp
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sara Heebøll
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus 8200, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Marie Winther-Sørensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sasha A S Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Nicole J Jensen
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Janus D Nybing
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Frederik H Linden
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Erik Høgh-Schmidt
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Mikael P Boesen
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital - Hvidovre, Hvidovre 2650, Denmark
| | - Frank Vinholt Schiødt
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Kirsten Nørgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Signe Schmidt
- Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Lise Lotte Gluud
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Gastro Unit, Copenhagen University Hospital - Hvidovre, Hvidovre 2650, Denmark
| | - Steen B Haugaard
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Søren Nielsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jørgen Rungby
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
32
|
Sabaratnam R, Kristensen JM, Pedersen AJT, Kruse R, Handberg A, Wojtaszewski JFP, Højlund K. Acute Exercise Increases GDF15 and Unfolded Protein Response/Integrated Stress Response in Muscle in Type 2 Diabetes. J Clin Endocrinol Metab 2024; 109:1754-1764. [PMID: 38242693 DOI: 10.1210/clinem/dgae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 12/27/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
CONTEXT Regular exercise is a key prevention strategy for obesity and type 2 diabetes (T2D). Exerkines secreted in response to exercise or recovery may contribute to improved systemic metabolism. Conversely, an impaired exerkine response to exercise and recovery may contribute to cardiometabolic diseases. OBJECTIVE We investigated if the exercise-induced regulation of the exerkine, growth differentiation factor 15 (GDF15) and its putative upstream regulators of the unfolded protein response (UPR)/integrated stress response (ISR) is impaired in skeletal muscle in patients with T2D compared with weight-matched glucose-tolerant men. METHODS Thirteen male patients with T2D and 14 age- and weight-matched overweight/obese glucose-tolerant men exercised at 70% of VO2max for 1 hour. Blood and skeletal muscle biopsies were sampled before, immediately after, and 3 hours into recovery. Serum and muscle transcript levels of GDF15 and key markers of UPR/ISR were determined. Additionally, protein/phosphorylation levels of key regulators in UPR/ISR were investigated. RESULTS Acute exercise increased muscle gene expression and serum GDF15 levels in both groups. In recovery, muscle expression of GDF15 decreased toward baseline, whereas serum GDF15 remained elevated. In both groups, acute exercise increased the expression of UPR/ISR markers, including ATF4, CHOP, EIF2K3 (encoding PERK), and PPP1R15A (encoding GADD34), of which only CHOP remained elevated 3 hours into recovery. Downstream molecules of the UPR/ISR including XBP1-U, XBP1-S, and EDEM1 were increased with exercise and 3 hours into recovery in both groups. The phosphorylation levels of eIF2α-Ser51, a common marker of unfolded protein response (UPR) and ISR, increased immediately after exercise in controls, but decreased 3 hours into recovery in both groups. CONCLUSION In conclusion, exercise-induced regulation of GDF15 and key markers of UPR/ISR are not compromised in patients with T2D compared with weight-matched controls.
Collapse
Affiliation(s)
- Rugivan Sabaratnam
- Department of Clinical Research & Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense M, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Jonas M Kristensen
- Department of Clinical Research & Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense M, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Andreas J T Pedersen
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Rikke Kruse
- Department of Clinical Research & Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense M, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, North Denmark Region, DK-9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark
| | - Jørgen F P Wojtaszewski
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Kurt Højlund
- Department of Clinical Research & Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense M, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark
| |
Collapse
|
33
|
Yau K, Kuah R, Cherney DZI, Lam TKT. Obesity and the kidney: mechanistic links and therapeutic advances. Nat Rev Endocrinol 2024; 20:321-335. [PMID: 38351406 DOI: 10.1038/s41574-024-00951-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/02/2024] [Indexed: 02/19/2024]
Abstract
Obesity is strongly associated with the development of diabetes mellitus and chronic kidney disease (CKD), but there is evidence for a bidirectional relationship wherein the kidney also acts as a key regulator of body weight. In this Review, we highlight the mechanisms implicated in obesity-related CKD, and outline how the kidney might modulate feeding and body weight through a growth differentiation factor 15-dependent kidney-brain axis. The favourable effects of bariatric surgery on kidney function are discussed, and medical therapies designed for the treatment of diabetes mellitus that lower body weight and preserve kidney function independent of glycaemic lowering, including sodium-glucose cotransporter 2 inhibitors, incretin-based therapies and metformin, are also reviewed. In summary, we propose that kidney function and body weight are related in a bidirectional fashion, and that this interrelationship affects human health and disease.
Collapse
Affiliation(s)
- Kevin Yau
- Division of Nephrology, Department of Medicine, Toronto General Hospital, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Kuah
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - David Z I Cherney
- Division of Nephrology, Department of Medicine, Toronto General Hospital, Toronto, Ontario, Canada.
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada.
| | - Tony K T Lam
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada.
| |
Collapse
|
34
|
Nunes JRC, O'Dwyer C, Ghorbani P, Smith TKT, Chauhan S, Robert-Gostlin V, Girouard MD, Viollet B, Foretz M, Fullerton MD. Myeloid AMPK signaling restricts fibrosis but is not required for metformin improvements during CDAHFD-induced NASH in mice. J Lipid Res 2024; 65:100564. [PMID: 38762124 PMCID: PMC11222943 DOI: 10.1016/j.jlr.2024.100564] [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: 08/08/2023] [Revised: 05/07/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024] Open
Abstract
Metabolic programming underpins inflammation and liver macrophage activation in the setting of chronic liver disease. Here, we sought to identify the role of an important metabolic regulator, AMP-activated protein kinase (AMPK), specifically within myeloid cells during the progression of non-alcoholic steatohepatitis (NASH) and whether treatment with metformin, a firstline therapy for diabetes and activator of AMPK could stem disease progression. Male and female Prkaa1fl/fl/Prkaa2fl/fl (Flox) control and Flox-LysM-Cre+ (MacKO) mice were fed a low-fat control or a choline-deficient, amino acid defined 45% Kcal high-fat diet (CDAHFD) for 8 weeks, where metformin was introduced in the drinking water (50 or 250 mg/kg/day) for the last 4 weeks. Hepatic steatosis and fibrosis were dramatically increased in response to CDAHFD-feeding compared to low-fat control. While myeloid AMPK signaling had no effect on markers of hepatic steatosis or circulating markers, fibrosis as measured by total liver collagen was significantly elevated in livers from MacKO mice, independent of sex. Although treatment with 50 mg/kg/day metformin had no effect on any parameter, intervention with 250 mg/kg/day metformin completely ameliorated hepatic steatosis and fibrosis in both male and female mice. While the protective effect of metformin was associated with lower final body weight, and decreased expression of lipogenic and Col1a1 transcripts, it was independent of myeloid AMPK signaling. These results suggest that endogenous AMPK signaling in myeloid cells, both liver-resident and infiltrating, acts to restrict fibrogenesis during CDAHFD-induced NASH progression but is not the mechanism by which metformin improves markers of NASH.
Collapse
Affiliation(s)
- Julia R C Nunes
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Conor O'Dwyer
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Peyman Ghorbani
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Tyler K T Smith
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Samarth Chauhan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Victoria Robert-Gostlin
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Madison D Girouard
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Benoit Viollet
- Université Paris cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Marc Foretz
- Université Paris cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Morgan D Fullerton
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Centre for Infection, Immunity and Inflammation, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada; Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
35
|
Lasaad S, Crambert G. GDF15, an Emerging Player in Renal Physiology and Pathophysiology. Int J Mol Sci 2024; 25:5956. [PMID: 38892145 PMCID: PMC11172470 DOI: 10.3390/ijms25115956] [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: 04/05/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
These last years, the growth factor GDF15 has emerged as a key element in many different biological processes. It has been established as being produced in response to many pathological states and is now referred to as a stress-related hormone. Regarding kidney functions, GDF15 has been involved in different pathologies such as chronic kidney disease, diabetic nephropathy, renal cancer, and so on. Interestingly, recent studies also revealed a role of GDF15 in the renal homeostatic mechanisms allowing to maintain constant, as far as possible, the plasma parameters such as pH and K+ values. In this review, we recapitulate the role of GDF15 in physiological and pathological context by focusing our interest on its renal effect.
Collapse
Affiliation(s)
- Samia Lasaad
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Gilles Crambert
- Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Scientifique (INSERM), Sorbonne Université, Université Paris Cité, Laboratoire de Physiologie Rénale et Tubulopathies, F-75006 Paris, France
- Unité Métabolisme et Physiologie Rénale, Centre National de la Recherche Scientifique (CNRS) EMR 8228, F-75006 Paris, France
| |
Collapse
|
36
|
Wang Y, Jia X, Cong B. Advances in the mechanism of metformin with wide-ranging effects on regulation of the intestinal microbiota. Front Microbiol 2024; 15:1396031. [PMID: 38855769 PMCID: PMC11157079 DOI: 10.3389/fmicb.2024.1396031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/29/2024] [Indexed: 06/11/2024] Open
Abstract
Metformin is of great focus because of its high safety, low side effects, and various effects other than lowering blood sugar, such as anti-inflammation, anti-tumor, and anti-aging. Studies have shown that metformin has a modulating effect on the composition and function of the intestinal microbiota other than acting on the liver. However, the composition of microbiota is complex and varies to some extent between species and individuals, and the experimental design of each study is also different. Multiple factors present a major obstacle to better comprehending the effects of metformin on the gut microbiota. This paper reviews the regulatory effects of metformin on the gut microbiota, such as increasing the abundance of genus Akkermansia, enriching short-chain fatty acids (SCFAs)-producing bacterial genus, and regulating gene expression of certain genera. The intestinal microbiota is a large and vital ecosystem in the human body and is considered to be the equivalent of an "organ" of the human body, which is highly relevant to human health and disease status. There are a lot of evidences that the gut microbiota is responsible for metformin's widespread effects. However, there are only a few systematic studies on this mechanism, and the specific mechanism is still unclear. This paper aims to summarize the possible mechanism of metformin in relation to gut microbiota.
Collapse
Affiliation(s)
- Yue Wang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Beijing, China
| | - Xianxian Jia
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Beijing, China
- Department of Pathogen Biology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Bin Cong
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
37
|
Maya SA, Khan JM, Islam MR, Shumon MAH, Banjare MK, Rana S, Hoque MA. Phase behaviour and thermodynamics of triton X-100 + metformin hydrochloride mixture: influences of several polyols and polymer media. Mol Phys 2024. [DOI: 10.1080/00268976.2024.2349195] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/24/2024] [Indexed: 01/12/2025]
Affiliation(s)
| | - Javed Masood Khan
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Md. Rafikul Islam
- Department of Chemistry, Dhaka University of Engineering & Technology, Gazipur, Bangladesh
| | - Md Anamul Haque Shumon
- Department of Chemistry, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | | | - Shahed Rana
- Department of Chemistry, Jahangirnagar University, Savar, Bangladesh
| | - Md. Anamul Hoque
- Department of Chemistry, Jahangirnagar University, Savar, Bangladesh
| |
Collapse
|
38
|
Yew MJ, Heywood SE, Ng J, West OM, Pal M, Kueh A, Lancaster GI, Myers S, Yang C, Liu Y, Reibe S, Mellett NA, Meikle PJ, Febbraio MA, Greening DW, Drew BG, Henstridge DC. ACAD10 is not required for metformin's metabolic actions or for maintenance of whole-body metabolism in C57BL/6J mice. Diabetes Obes Metab 2024; 26:1731-1745. [PMID: 38351663 DOI: 10.1111/dom.15484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/08/2024] [Accepted: 01/18/2024] [Indexed: 04/09/2024]
Abstract
AIM Acyl-coenzyme A dehydrogenase family member 10 (ACAD10) is a mitochondrial protein purported to be involved in the fatty acid oxidation pathway. Metformin is the most prescribed therapy for type 2 diabetes; however, its precise mechanisms of action(s) are still being uncovered. Upregulation of ACAD10 is a requirement for metformin's ability to inhibit growth in cancer cells and extend lifespan in Caenorhabditis elegans. However, it is unknown whether ACAD10 plays a role in metformin's metabolic actions. MATERIALS AND METHODS We assessed the role for ACAD10 on whole-body metabolism and metformin action by generating ACAD10KO mice on a C57BL/6J background via CRISPR-Cas9 technology. In-depth metabolic phenotyping was conducted in both sexes on a normal chow and high fat-high sucrose diet. RESULTS Compared with wildtype mice, we detected no difference in body composition, energy expenditure or glucose tolerance in male or female ACAD10KO mice, on a chow diet or high-fat, high-sucrose diet (p ≥ .05). Hepatic mitochondrial function and insulin signalling was not different between genotypes under basal or insulin-stimulated conditions (p ≥ .05). Glucose excursions following acute administration of metformin before a glucose tolerance test were not different between genotypes nor was body composition or energy expenditure altered after 4 weeks of daily metformin treatment (p ≥ .05). Despite the lack of a metabolic phenotype, liver lipidomic analysis suggests ACAD10 depletion influences the abundance of specific ceramide species containing very long chain fatty acids, while metformin treatment altered clusters of cholesterol ester, plasmalogen, phosphatidylcholine and ceramide species. CONCLUSIONS Loss of ACAD10 does not alter whole-body metabolism or impact the acute or chronic metabolic actions of metformin in this model.
Collapse
Affiliation(s)
- Michael J Yew
- School of Health Sciences, The University of Tasmania, Launceston, Tasmania, Australia
| | - Sarah E Heywood
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Joe Ng
- School of Health Sciences, The University of Tasmania, Launceston, Tasmania, Australia
| | - Olivia M West
- School of Health Sciences, The University of Tasmania, Launceston, Tasmania, Australia
| | - Martin Pal
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Andrew Kueh
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - Stephen Myers
- School of Health Sciences, The University of Tasmania, Launceston, Tasmania, Australia
| | - Christine Yang
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yingying Liu
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Saskia Reibe
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- University of Oxford, Oxford, UK
| | | | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia
| | - Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - David W Greening
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Darren C Henstridge
- School of Health Sciences, The University of Tasmania, Launceston, Tasmania, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| |
Collapse
|
39
|
Engström Ruud L, Font-Gironès F, Zajdel J, Kern L, Teixidor-Deulofeu J, Mannerås-Holm L, Carreras A, Becattini B, Björefeldt A, Hanse E, Fenselau H, Solinas G, Brüning JC, Wunderlich TF, Bäckhed F, Ruud J. Activation of GFRAL + neurons induces hypothermia and glucoregulatory responses associated with nausea and torpor. Cell Rep 2024; 43:113960. [PMID: 38507407 DOI: 10.1016/j.celrep.2024.113960] [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: 09/21/2023] [Revised: 01/11/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
GFRAL-expressing neurons actuate aversion and nausea, are targets for obesity treatment, and may mediate metformin effects by long-term GDF15-GFRAL agonism. Whether GFRAL+ neurons acutely regulate glucose and energy homeostasis is, however, underexplored. Here, we report that cell-specific activation of GFRAL+ neurons using a variety of techniques causes a torpor-like state, including hypothermia, the release of stress hormones, a shift from glucose to lipid oxidation, and impaired insulin sensitivity, glucose tolerance, and skeletal muscle glucose uptake but augmented glucose uptake in visceral fat. Metabolomic analysis of blood and transcriptomics of muscle and fat indicate alterations in ketogenesis, insulin signaling, adipose tissue differentiation and mitogenesis, and energy fluxes. Our findings indicate that acute GFRAL+ neuron activation induces endocrine and gluco- and thermoregulatory responses associated with nausea and torpor. While chronic activation of GFRAL signaling promotes weight loss in obesity, these results show that acute activation of GFRAL+ neurons causes hypothermia and hyperglycemia.
Collapse
Affiliation(s)
- Linda Engström Ruud
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ferran Font-Gironès
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joanna Zajdel
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lara Kern
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Júlia Teixidor-Deulofeu
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise Mannerås-Holm
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Alba Carreras
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Barbara Becattini
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Andreas Björefeldt
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eric Hanse
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Giovanni Solinas
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | | | - Fredrik Bäckhed
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Ruud
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
40
|
Jensterle M, Ferjan S, Janez A. The maintenance of long-term weight loss after semaglutide withdrawal in obese women with PCOS treated with metformin: a 2-year observational study. Front Endocrinol (Lausanne) 2024; 15:1366940. [PMID: 38665260 PMCID: PMC11043580 DOI: 10.3389/fendo.2024.1366940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Background Withdrawal of semaglutide is frequently followed by weight regain due to compensatory biological changes that prevent the maintenance of long-term weight loss. There are some studies implying that metformin might attenuate weight regain. The weight trajectory after discontinuation of short-term semaglutide treatment in obese women with PCOS who continued metformin treatment has not yet been evaluated. Aims We explored changes in body weight, cardiometabolic and endocrine parameters in obese women with PCOS who continued treatment with metformin 2 years after discontinuation of short-term intervention with semaglutide. Methods 25 women with PCOS and obesity, aged 33.7 ± 5.3 years (mean ± SD), were treated with once-weekly subcutaneous semaglutide 1.0 mg as an adjunct to metformin 2000 mg/day and lifestyle intervention for 16 weeks. At week 16, semaglutide was discontinued. Treatment with metformin 2000 mg/day and promotion of lifestyle intervention were continued during the 2-year follow-up period. Weight change, cardiometabolic, and endocrine parameters were assessed 2 years after semaglutide discontinuation. Results During semaglutide treatment phase, weight decreased from 101 (90-106.8) kg to 92 (83.3-100.8) kg. Two years after semaglutide withdrawal, weight was 95 (77-104) kg. The net weight loss 2 years after discontinuation of semaglutide remained significant when compared to baseline (p=0.003). At the end of the study, 21 out of 25 subjects had lower body weight compared to baseline. Improvements in cardiometabolic parameters including decrease in total and LDL cholesterol, fasting glucose, and glucose after OGTT that had been seen during semaglutide-treatment phase reverted towards baseline two years after semaglutide cessation. Free testosterone levels significantly decreased during semaglutide treatment from 6.16 (4.07-9.71) to 4.12 (2.98-6.93) nmol/l, (p= 0.012) and did not significantly deteriorate after semaglutide discontinuation. Conclusion Two years after semaglutide withdrawal, women with PCOS who continued with metformin regained about one-third of the semaglutide-induced weight loss. At the end of the follow up, 84% of women had a lower body weight than at baseline.
Collapse
Affiliation(s)
- Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Simona Ferjan
- Department of Endocrinology, Diabetes and Metabolic Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Andrej Janez
- Department of Endocrinology, Diabetes and Metabolic Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
41
|
Allard C, Miralpeix C, López-Gambero AJ, Cota D. mTORC1 in energy expenditure: consequences for obesity. Nat Rev Endocrinol 2024; 20:239-251. [PMID: 38225400 DOI: 10.1038/s41574-023-00934-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2023] [Indexed: 01/17/2024]
Abstract
In eukaryotic cells, the mammalian target of rapamycin complex 1 (sometimes referred to as the mechanistic target of rapamycin complex 1; mTORC1) orchestrates cellular metabolism in response to environmental energy availability. As a result, at the organismal level, mTORC1 signalling regulates the intake, storage and use of energy by acting as a hub for the actions of nutrients and hormones, such as leptin and insulin, in different cell types. It is therefore unsurprising that deregulated mTORC1 signalling is associated with obesity. Strategies that increase energy expenditure offer therapeutic promise for the treatment of obesity. Here we review current evidence illustrating the critical role of mTORC1 signalling in the regulation of energy expenditure and adaptive thermogenesis through its various effects in neuronal circuits, adipose tissue and skeletal muscle. Understanding how mTORC1 signalling in one organ and cell type affects responses in other organs and cell types could be key to developing better, safer treatments targeting this pathway in obesity.
Collapse
Affiliation(s)
- Camille Allard
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | | | | | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France.
| |
Collapse
|
42
|
Scott B, Day EA, O'Brien KL, Scanlan J, Cromwell G, Scannail AN, McDonnell ME, Finlay DK, Lynch L. Metformin and feeding increase levels of the appetite-suppressing metabolite Lac-Phe in humans. Nat Metab 2024; 6:651-658. [PMID: 38499765 PMCID: PMC11052712 DOI: 10.1038/s42255-024-01018-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/22/2024] [Indexed: 03/20/2024]
Abstract
Metformin, a widely used first-line treatment for type 2 diabetes (T2D), is known to reduce blood glucose levels and suppress appetite. Here we report a significant elevation of the appetite-suppressing metabolite N-lactoyl phenylalanine (Lac-Phe) in the blood of individuals treated with metformin across seven observational and interventional studies. Furthermore, Lac-Phe levels were found to rise in response to acute metformin administration and post-prandially in patients with T2D or in metabolically healthy volunteers.
Collapse
Affiliation(s)
- Barry Scott
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Emily A Day
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Katie L O'Brien
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - John Scanlan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Grace Cromwell
- Division of Endocrinology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Aine Ni Scannail
- Division of Endocrinology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marie E McDonnell
- Division of Endocrinology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David K Finlay
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
- Division of Endocrinology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
43
|
Xiao S, Li VL, Lyu X, Chen X, Wei W, Abbasi F, Knowles JW, Tung ASH, Deng S, Tiwari G, Shi X, Zheng S, Farrell L, Chen ZZ, Taylor KD, Guo X, Goodarzi MO, Wood AC, Chen YDI, Lange LA, Rich SS, Rotter JI, Clish CB, Tahir UA, Gerszten RE, Benson MD, Long JZ. Lac-Phe mediates the effects of metformin on food intake and body weight. Nat Metab 2024; 6:659-669. [PMID: 38499766 PMCID: PMC11062621 DOI: 10.1038/s42255-024-00999-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 01/30/2024] [Indexed: 03/20/2024]
Abstract
Metformin is a widely prescribed anti-diabetic medicine that also reduces body weight. There is ongoing debate about the mechanisms that mediate metformin's effects on energy balance. Here, we show that metformin is a powerful pharmacological inducer of the anorexigenic metabolite N-lactoyl-phenylalanine (Lac-Phe) in cells, in mice and two independent human cohorts. Metformin drives Lac-Phe biosynthesis through the inhibition of complex I, increased glycolytic flux and intracellular lactate mass action. Intestinal epithelial CNDP2+ cells, not macrophages, are the principal in vivo source of basal and metformin-inducible Lac-Phe. Genetic ablation of Lac-Phe biosynthesis in male mice renders animals resistant to the effects of metformin on food intake and body weight. Lastly, mediation analyses support a role for Lac-Phe as a downstream effector of metformin's effects on body mass index in participants of a large population-based observational cohort, the Multi-Ethnic Study of Atherosclerosis. Together, these data establish Lac-Phe as a critical mediator of the body weight-lowering effects of metformin.
Collapse
Affiliation(s)
- Shuke Xiao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Veronica L Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Xuchao Lyu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Xudong Chen
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Wei Wei
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Fahim Abbasi
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua W Knowles
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan Sheng-Hwa Tung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Shuliang Deng
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Gaurav Tiwari
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Xu Shi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Shuning Zheng
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Laurie Farrell
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Zsu-Zsu Chen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alexis C Wood
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Clary B Clish
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Usman A Tahir
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mark D Benson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA.
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
44
|
Cheng M, Ren L, Jia X, Wang J, Cong B. Understanding the action mechanisms of metformin in the gastrointestinal tract. Front Pharmacol 2024; 15:1347047. [PMID: 38617792 PMCID: PMC11010946 DOI: 10.3389/fphar.2024.1347047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/15/2024] [Indexed: 04/16/2024] Open
Abstract
Metformin is the initial medication recommended for the treatment of type 2 diabetes mellitus (T2DM). In addition to diabetes treatment, the function of metformin also can be anti-aging, antiviral, and anti-inflammatory. Nevertheless, further exploration is required to fully understand its mode of operation. Historically, the liver has been acknowledged as the main location where metformin reduces glucose levels, however, there is increasing evidence suggesting that the gastrointestinal tract also plays a significant role in its action. In the gastrointestinal tract, metformin effects glucose uptake and absorption, increases glucagon-like peptide-1 (GLP-1) secretion, alters the composition and structure of the gut microbiota, and modulates the immune response. However, the side effects of it cannot be ignored such as gastrointestinal distress in patients. This review outlines the impact of metformin on the digestive system and explores potential explanations for variations in metformin effectiveness and adverse effects like gastrointestinal discomfort.
Collapse
Affiliation(s)
- Meihui Cheng
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lili Ren
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianxian Jia
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Pathogen Biology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Jianwei Wang
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Cong
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
45
|
Zhang SY, Danaei Z, Bruce K, Chiu JFM, Lam TKT. Acute Activation of GFRAL in the Area Postrema Contributes to Glucose Regulation Independent of Weight. Diabetes 2024; 73:426-433. [PMID: 38064571 DOI: 10.2337/db23-0705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/03/2023] [Indexed: 02/22/2024]
Abstract
GDF15 regulates energy balance and glucose homeostasis in rodents by activating its receptor GFRAL, expressed in the area postrema of the brain. However, whether GDF15-GFRAL signaling in the area postrema regulates glucose tolerance independent of changes in food intake and weight and contributes to the glucose-lowering effect of metformin remain unknown. Herein, we report that direct, acute GDF15 infusion into the area postrema of rats fed a high-fat diet increased intravenous glucose tolerance and insulin sensitivity to lower hepatic glucose production independent of changes in food intake, weight, and plasma insulin levels under conscious, unrestrained, and nonstressed conditions. In parallel, metformin infusion concurrently increased plasma GDF15 levels and glucose tolerance. Finally, a knockdown of GFRAL expression in the area postrema negated administration of GDF15, as well as metformin, to increase glucose tolerance independent of changes in food intake, weight, and plasma insulin levels. In summary, activation of GFRAL in the area postrema contributes to glucose regulation of GDF15 and metformin in vivo. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Song-Yang Zhang
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Zahra Danaei
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Kyla Bruce
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer F M Chiu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Tony K T Lam
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Ellibishy F, Tarek M, Abd-Elsalam MM, Elgayar N, El Bakly W. Metformin improves d-galactose induced premature ovarian insufficiency through PI3K-Akt-FOXO3a pathway. Adv Med Sci 2024; 69:70-80. [PMID: 38387407 DOI: 10.1016/j.advms.2024.02.004] [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: 10/17/2023] [Revised: 12/23/2023] [Accepted: 02/12/2024] [Indexed: 02/24/2024]
Abstract
PURPOSE Metformin (MET), a first-line treatment for type 2 diabetes mellitus, restores ovarian function in women with polycystic ovary syndrome. MET has been shown to increase the rate of success for in vitro fertilization when utilized in assisted reproductive technologies. This study was designed to examine the impact of MET on ovarian function and fertility in a mouse model of galactose-induced premature ovarian insufficiency (POI). We further investigated the underlying mechanisms. MATERIALS AND METHODS Female mice were divided into 4 groups: saline, d-galactose, d-galactose + MET, and MET. Body weight, ovarian index, and fertility were assessed. The hormonal profile was done. Advanced glycation end products (AGEPs), receptor for advanced glycation end products (RAGE), phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), forkhead box O3a (FOXO3a) expression were measured. Ovarian follicle counting and morphology were analyzed. Immunohistochemistry of cleaved caspase-3 expression was performed. RESULTS Our findings demonstrated that MET reversed irregularities in the estrus cycle, enhanced the ovarian index, and improved the abnormal levels of hormones and AGEs induced by d-galactose. Furthermore, the expression levels of PI3K, Akt, FOXO3a, and RAGE were upregulated with d-galactose. However, MET attenuated their expression levels. The primordial follicles ratio was improved, whereas atretic follicles and apoptotic-related cleaved caspase-3 expression were decreased in the d-galactose + MET group compared to the d-galactose group. CONCLUSION This study demonstrates that MET partially rescued ovarian dysfunction and apoptosis induced by d-galactose via a mechanism involving PI3K-Akt-FOXO3a pathway. Our finding proposed that MET may be a promising alternative treatment for POI.
Collapse
Affiliation(s)
- Fatima Ellibishy
- Clinical Pharmacology Department, Faculty of Medicine, Kafr Elsheikh University, Kafr Elsheikh, Egypt.
| | - Maha Tarek
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Marwa M Abd-Elsalam
- Histology & Cell Biology Department, Faculty of Medicine, Kafr Elsheikh University, Kafr Elsheikh, Egypt
| | - Nesreen Elgayar
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Wesam El Bakly
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
47
|
Jurado-Aguilar J, Barroso E, Bernard M, Zhang M, Peyman M, Rada P, Valverde ÁM, Wahli W, Palomer X, Vázquez-Carrera M. GDF15 activates AMPK and inhibits gluconeogenesis and fibrosis in the liver by attenuating the TGF-β1/SMAD3 pathway. Metabolism 2024; 152:155772. [PMID: 38176644 DOI: 10.1016/j.metabol.2023.155772] [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: 07/20/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
INTRODUCTION The levels of the cellular energy sensor AMP-activated protein kinase (AMPK) have been reported to be decreased via unknown mechanisms in the liver of mice deficient in growth differentiation factor 15 (GDF15). This stress response cytokine regulates energy metabolism mainly by reducing food intake through its hindbrain receptor GFRAL. OBJECTIVE To examine how GDF15 regulates AMPK. METHODS Wild-type and Gdf15-/- mice, mouse primary hepatocytes and the human hepatic cell line Huh-7 were used. RESULTS Gdf15-/- mice showed glucose intolerance, reduced hepatic phosphorylated AMPK levels, increased levels of phosphorylated mothers against decapentaplegic homolog 3 (SMAD3; a mediator of the fibrotic response), elevated serum levels of transforming growth factor (TGF)-β1, as well as upregulated gluconeogenesis and fibrosis. In line with these observations, recombinant (r)GDF15 promoted AMPK activation and reduced the levels of phosphorylated SMAD3 and the markers of gluconeogenesis and fibrosis in the liver of mice and in mouse primary hepatocytes, suggesting that these effects may be independent of GFRAL. Pharmacological inhibition of SMAD3 phosphorylation in Gdf15-/- mice prevented glucose intolerance, the deactivation of AMPK and the increase in the levels of proteins involved in gluconeogenesis and fibrosis, suggesting that overactivation of the TGF-β1/SMAD3 pathway is responsible for the metabolic alterations in Gdf15-/- mice. CONCLUSIONS Overall, these findings indicate that GDF15 activates AMPK and inhibits gluconeogenesis and fibrosis by lowering the activity of the TGF-β1/SMAD3 pathway.
Collapse
Affiliation(s)
- Javier Jurado-Aguilar
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Maribel Bernard
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Meijian Zhang
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Mona Peyman
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Patricia Rada
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigaciones Biomédicas Alberto Sols (CSIC/UAM), Madrid, Spain
| | - Ángela M Valverde
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigaciones Biomédicas Alberto Sols (CSIC/UAM), Madrid, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland; Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232; ToxAlim (Research Center in Food Toxicology), INRAE, UMR1331, F-31300 Toulouse Cedex, France
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain.
| |
Collapse
|
48
|
Richter MM, Thomsen MN, Skytte MJ, Kjeldsen SAS, Samkani A, Frystyk J, Magkos F, Holst JJ, Madsbad S, Krarup T, Haugaard SB, Wewer Albrechtsen NJ. Effect of a 6-Week Carbohydrate-Reduced High-Protein Diet on Levels of FGF21 and GDF15 in People With Type 2 Diabetes. J Endocr Soc 2024; 8:bvae008. [PMID: 38379856 PMCID: PMC10875725 DOI: 10.1210/jendso/bvae008] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Indexed: 02/22/2024] Open
Abstract
Context Fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15) are increased in type 2 diabetes and are potential regulators of metabolism. The effect of changes in caloric intake and macronutrient composition on their circulating levels in patients with type 2 diabetes are unknown. Objective To explore the effects of a carbohydrate-reduced high-protein diet with and without a clinically significant weight loss on circulating levels of FGF21 and GDF15 in patients with type 2 diabetes. Methods We measured circulating FGF21 and GDF15 in patients with type 2 diabetes who completed 2 previously published diet interventions. Study 1 randomized 28 subjects to an isocaloric diet in a 6 + 6-week crossover trial consisting of, in random order, a carbohydrate-reduced high-protein (CRHP) or a conventional diabetes (CD) diet. Study 2 randomized 72 subjects to a 6-week hypocaloric diet aiming at a ∼6% weight loss induced by either a CRHP or a CD diet. Fasting plasma FGF21 and GDF15 were measured before and after the interventions in a subset of samples (n = 24 in study 1, n = 66 in study 2). Results Plasma levels of FGF21 were reduced by 54% in the isocaloric study (P < .05) and 18% in the hypocaloric study (P < .05) in CRHP-treated individuals only. Circulating GDF15 levels increased by 18% (P < .05) following weight loss in combination with a CRHP diet but only in those treated with metformin. Conclusion The CRHP diet significantly reduced FGF21 in people with type 2 diabetes independent of weight loss, supporting the role of FGF21 as a "nutrient sensor." Combining metformin treatment with carbohydrate restriction and weight loss may provide additional metabolic improvements due to the rise in circulating GDF15.
Collapse
Affiliation(s)
- Michael M Richter
- Department of Clinical Biochemistry, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Mads N Thomsen
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
| | - Mads J Skytte
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Department of Forensic Medicine, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Sasha A S Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Amirsalar Samkani
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
| | - Jan Frystyk
- Department of Endocrinology, Odense University Hospital, Odense, 5000, Denmark
| | - Faidon Magkos
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital—Hvidovre, Hvidovre, 2650, Denmark
| | - Thure Krarup
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Steen B Haugaard
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| |
Collapse
|
49
|
Guo X, Asthana P, Zhai L, Cheng KW, Gurung S, Huang J, Wu J, Zhang Y, Mahato AK, Saarma M, Ustav M, Kwan HY, Lyu A, Chan KM, Xu P, Bian ZX, Wong HLX. Artesunate treats obesity in male mice and non-human primates through GDF15/GFRAL signalling axis. Nat Commun 2024; 15:1034. [PMID: 38310105 PMCID: PMC10838268 DOI: 10.1038/s41467-024-45452-3] [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: 12/06/2022] [Accepted: 01/23/2024] [Indexed: 02/05/2024] Open
Abstract
Obesity, a global health challenge, is a major risk factor for multiple life-threatening diseases, including diabetes, fatty liver, and cancer. There is an ongoing need to identify safe and tolerable therapeutics for obesity management. Herein, we show that treatment with artesunate, an artemisinin derivative approved by the FDA for the treatment of severe malaria, effectively reduces body weight and improves metabolic profiles in preclinical models of obesity, including male mice with overnutrition-induced obesity and male cynomolgus macaques with spontaneous obesity, without inducing nausea and malaise. Artesunate promotes weight loss and reduces food intake in obese mice and cynomolgus macaques by increasing circulating levels of Growth Differentiation Factor 15 (GDF15), an appetite-regulating hormone with a brainstem-restricted receptor, the GDNF family receptor α-like (GFRAL). Mechanistically, artesunate induces the expression of GDF15 in multiple organs, especially the liver, in mice through a C/EBP homologous protein (CHOP)-directed integrated stress response. Inhibition of GDF15/GFRAL signalling by genetic ablation of GFRAL or tissue-specific knockdown of GDF15 abrogates the anti-obesity effect of artesunate in mice with diet-induced obesity, suggesting that artesunate controls bodyweight and appetite in a GDF15/GFRAL signalling-dependent manner. These data highlight the therapeutic benefits of artesunate in the treatment of obesity and related comorbidities.
Collapse
Affiliation(s)
- Xuanming Guo
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Pallavi Asthana
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| | - Lixiang Zhai
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ka Wing Cheng
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Centre for Chinese Herbal Medicine Drug Development Limited, Hong Kong Baptist University, Hong Kong SAR, China
| | - Susma Gurung
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jiangang Huang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Jiayan Wu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yijing Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Arun Kumar Mahato
- Institute of Biotechnology-HILIFE, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology-HILIFE, University of Helsinki, Helsinki, Finland
| | | | - Hiu Yee Kwan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Aiping Lyu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Pingyi Xu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhao-Xiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Centre for Chinese Herbal Medicine Drug Development Limited, Hong Kong Baptist University, Hong Kong SAR, China.
| | | |
Collapse
|
50
|
Asundi J, Zhang C, Donnelly‐Roberts D, Solorio JZ, Challagundla M, Connelly C, Boch C, Chen J, Richter M, Maneshi MM, Swensen AM, Lebon L, Schiffmann R, Sanyal S, Sidrauski C, Kolumam G, Baruch A. GDF15 is a dynamic biomarker of the integrated stress response in the central nervous system. CNS Neurosci Ther 2024; 30:e14600. [PMID: 38357857 PMCID: PMC10867791 DOI: 10.1111/cns.14600] [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: 06/22/2023] [Revised: 12/11/2023] [Accepted: 12/28/2023] [Indexed: 02/16/2024] Open
Abstract
AIM Characterize Growth Differentiation Factor 15 (GDF15) as a secreted biomarker of the integrated stress response (ISR) within the central nervous system (CNS). METHODS We determined GDF15 levels utilizing in vitro and in vivo neuronal systems wherein the ISR was activated. Primarily, we used the murine model of vanishing white matter disease (VWMD), a neurological disease driven by persistent ISR in the CNS, to establish a link between levels of GDF15 in the cerebrospinal fluid (CSF) and ISR gene expression signature in the CNS. GDF15 was also determined in the CSF of VWM patients. RESULTS GDF15 expression was increased concomitant to ISR activation in stress-induced primary astrocytes as well as in retinal ganglion cells following optic nerve crush, while treatment with 2Bact, a specific eIF2B activator, suppressed both the ISR and GDF15. In the VWMD model, CSF GDF15 levels corresponded with the magnitude of the ISR and were reduced by 2BAct. In VWM patients, mean CSF GDF15 was elevated >20-fold as compared to healthy controls, whereas plasma GDF15 was undifferentiated. CONCLUSIONS These data suggest that CSF GDF15 is a dynamic marker of ISR activation in the CNS and may serve as a pharmacodynamic biomarker for ISR-modulating therapies.
Collapse
Affiliation(s)
- Jyoti Asundi
- Calico Life Sciences LLCSouth San FranciscoCaliforniaUSA
| | - Chunlian Zhang
- Calico Life Sciences LLCSouth San FranciscoCaliforniaUSA
| | | | | | | | | | | | | | | | | | | | - Lauren Lebon
- Calico Life Sciences LLCSouth San FranciscoCaliforniaUSA
| | | | | | | | - Ganesh Kolumam
- Calico Life Sciences LLCSouth San FranciscoCaliforniaUSA
| | - Amos Baruch
- Calico Life Sciences LLCSouth San FranciscoCaliforniaUSA
| |
Collapse
|