251
|
Lu D, Zhang W, Li R, Tan S, Zhang Y. Targeting necroptosis in Alzheimer's disease: can exercise modulate neuronal death? Front Aging Neurosci 2025; 17:1499871. [PMID: 40161268 PMCID: PMC11950841 DOI: 10.3389/fnagi.2025.1499871] [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: 09/22/2024] [Accepted: 01/15/2025] [Indexed: 04/02/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and neuronal degeneration. Emerging evidence implicates necroptosis in AD pathogenesis, driven by the RIPK1-RIPK3-MLKL pathway, which promotes neuronal damage, inflammation, and disease progression. Exercise, as a non-pharmacological intervention, can modulate key inflammatory mediators such as TNF-α, HMGB1, and IL-1β, thereby inhibiting necroptotic signaling. Additionally, exercise enhances O-GlcNAc glycosylation, preventing Tau hyperphosphorylation and stabilizing neuronal integrity. This review explores how exercise mitigates necroptosis and neuroinflammation, offering novel therapeutic perspectives for AD prevention and management.
Collapse
Affiliation(s)
- Donglei Lu
- Tianjin Key Laboratory of Sports and Health Integration and Health Promotion, Tianjin, China
| | - Wenyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruiyu Li
- Beijing University of Chinese Medicine Shenzhen Hospital (Longgang), Shenzhen, China
| | - Sijie Tan
- Tianjin Key Laboratory of Sports and Health Integration and Health Promotion, Tianjin, China
| | - Yan Zhang
- Tianjin Shengzhi Sports Technology Co., Ltd., Tianjin, China
| |
Collapse
|
252
|
Dai J, Zhao Y, Chen Y, Jiang Y, Sun R, Tang X, Cui Y, Mao H, Peng XG. Irisin reverses high-fat diet-induced metabolic dysfunction via activation of brown adipose tissue in mice. Int J Obes (Lond) 2025:10.1038/s41366-025-01739-z. [PMID: 40082597 DOI: 10.1038/s41366-025-01739-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 02/01/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND High-fat diet (HFD) induces negative effects on the activity of interscapular brown adipose tissue (iBAT) and systemic energy metabolism. Irisin, a small hormonal agent known to modulate metabolism has been used for intervening HFD-induced obesity. However, its mechanism of action on iBAT function remains to be fully elucidated. This study sought to investigate whether irisin intervention could restore the thermogenic function of iBAT in mice with HFD-induced obesity, thereby regulating systemic metabolism. METHODS Magnetic resonance imaging (MRI) and positron emission tomography/computed tomography (PET/CT) were used to monitor changes of thermogenic capacity of iBAT and systemic metabolism in mice with HFD-induced obesity and iBAT deficiency during 2-week or 4-week irisin intervention. Pathological and molecular biology analyses were performed on tissue and blood samples. RESULTS Prolonged HFD feeding in mice induced obesity and impaired the thermogenic capacity of iBAT. MRI results showed that irisin intervention for 4-week reduced lipid content in iBAT, increased uncoupling protein 1 (UCP 1) expression and enhanced glucose analogue uptake capacity. These improvements of functions in iBAT activity were accompanied by an improvement in systemic metabolism. The positive effects of irisin appears to be dependent on the length of intervention time. When iBAT was removed, the beneficial effects of irisin were partially suppressed, suggesting that irisin regulates metabolism through the restoration of the thermogenic function of iBAT. CONCLUSIONS HFD results in reduced thermogenic capacity of iBAT, while irisin intervention can effectively restore iBAT function, leading to improvement in overall glucose and lipid metabolism.
Collapse
Affiliation(s)
- Jingyue Dai
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yufei Zhao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yue Chen
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yang Jiang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Rui Sun
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215002, China
| | - Xingzhe Tang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ying Cui
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Xin-Gui Peng
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
- Department of Radiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 211200, China.
| |
Collapse
|
253
|
Yenmis G, Kallenci I, Dokur M, Koc S, Yalinkilic SB, Atak E, Demirbilek M, Arkan H. The Distribution of Sport Performance Gene Variations Through COVID-19 Disease Severity. Diagnostics (Basel) 2025; 15:701. [PMID: 40150043 PMCID: PMC11941099 DOI: 10.3390/diagnostics15060701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/23/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Since its emergence in 2020, researchers worldwide have been collaborating to better understand the SARS-CoV-2 disease's pathophysiology. Disease severity can vary based on several factors, including comorbidities and genetic variations. Notably, recent studies have highlighted the role of genes associated with athletic performance, such as ACE, ACTN3, and PPARGC1A, in influencing muscle function, cardiovascular health, and the body's metabolic response. Given that these genes also impact oxidative metabolism, inflammation, and respiratory efficiency, we hypothesized that they might play a critical role in the host's response to SARS-CoV-2 infection. This study aimed to investigate the association between disease severity and genetic polymorphisms in these sport performance-related genes, specifically ACE rs4646994, ACTN3 rs1815739, and PPARGC1A rs8192678. Methods: A total of 422 COVID-19-positive patients were included in this study. The participants were divided into three groups: a severe group (77 patients) requiring intensive care unit (ICU) admission, a mild group (300 patients) exhibiting at least one symptom, and an asymptomatic control group. Genotyping was performed using restriction fragment length polymorphism PCR. Results: The D allele and DD genotype of ACE and the T allele and TT genotype of ACTN3 were found to confer protective effects against severe SARS-CoV-2 infection. Conversely, the PPARGC1A TC genotype and the ACE-PPARGC1A ins/ins + TC combined genotype were associated with increased disease severity (p < 0.05). Conclusions: Although vaccination has reduced the severity of SARS-CoV-2, the virus continues to impact human health. Inter-individual differences due to these genetic variations will broaden the horizon of knowledge on the pathophysiology of the disease.
Collapse
Affiliation(s)
- Guven Yenmis
- Department of Medical Biology, Tayfur Ata Sokmen School of Medicine, Hatay Mustafa Kemal University, Hatay 31060, Turkey
| | - Ilayda Kallenci
- Department of Molecular Biology and Genetics, Faculty of Natural Sciences and Engineering, Biruni University, Istanbul 34015, Turkey; (I.K.); (S.B.Y.)
| | - Mehmet Dokur
- Department of Emergency Medicine, Faculty of Medicine, Bilecik Seyh Edebali University, Bilecik 11230, Turkey;
| | - Suna Koc
- Department of Anesthesia and Reanimation, School of Medicine, Biruni University, Istanbul 34015, Turkey;
| | - Sila Basak Yalinkilic
- Department of Molecular Biology and Genetics, Faculty of Natural Sciences and Engineering, Biruni University, Istanbul 34015, Turkey; (I.K.); (S.B.Y.)
| | - Evren Atak
- Department of Bioinformatics and System Biology, Institute of Natural and Applied Sciences, Gebze Technical University, Kocaeli 41400, Turkey;
| | - Mahmut Demirbilek
- Department of Emergency Medicine, School of Medicine, Biruni University, Istanbul 34015, Turkey;
| | - Hulya Arkan
- Department of Biotechnology, Institute of Science, Yildiz Technical University, Istanbul 34210, Turkey;
| |
Collapse
|
254
|
Zhang N, Sun Q, Zhang J, Zhang R, Liu S, Zhao X, Ma J, Li X. Intrapancreatic adipocytes and beta cell dedifferentiation in human type 2 diabetes. Diabetologia 2025:10.1007/s00125-025-06392-9. [PMID: 40072535 DOI: 10.1007/s00125-025-06392-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/20/2025] [Indexed: 03/14/2025]
Abstract
AIMS/HYPOTHESIS Fat deposition in the pancreas is implicated in beta cell dysfunction and the progress of type 2 diabetes. However, there is limited evidence to confirm the correlation and explore how pancreatic fat links with beta cell dysfunction in human type 2 diabetes. This study aimed to examine the spatial relationship between pancreatic fat and islets in human pancreases. METHODS Histological analysis of pancreatic specimens from 50 organ donors (15 with type 2 diabetes, 35 without) assessed pancreatic fat content variation among individuals with diabetes and its correlation with estimated beta cell mass and cell distribution within islets. Bioinformatic analysis of single-cell RNA-seq of 11 type 2 diabetic donors (from the Human Pancreatic Analysis Project database) explored the impact of high pancreatic fat content on beta cell gene expression and cell fate. Validation of bioinformatic results was performed with the above diabetic pancreases. RESULTS Pancreatic fat content was higher in individuals with type 2 diabetes (10.24% [3.29-13.89%] vs 0.74% [0.34-5.11%], p<0.001), negatively correlated with estimated beta cell mass (r=-0.675, p=0.006) and positively with alpha-to-beta cell ratio (r=0.608, p=0.016). Enrichment analysis indicated that in diabetic donors with higher pancreatic fat content, the expression of ALDH1A3, beta cell dedifferentiation marker, in both alpha and beta cells was significantly increased, and in beta cells, the expression of NPY decreased. Pseudotime analysis revealed beta cell dedifferentiation and transdifferentiation towards alpha cells in diabetic donors with higher pancreatic fat content, with decreased expression of genes related to beta cell maturation and function, including INSM1, MafA and NPY. Concurrently, pathways related to inflammation and immune response were activated. Histologically, pancreatic fat content correlated positively with the percentage of beta cells positive for aldehyde dehydrogenase 1 family member A3 (ALDH1A3) within the islets (r=0.594, p=0.020) and the ALDH1A3 positivity rate in beta cells (r=0.615, p=0.015). And the number of T cells adjacent to adipocytes was related to the distribution pattern of adipocytes and the dedifferentiation phenotype in islets. CONCLUSIONS/INTERPRETATION Higher pancreatic fat content was accompanied by increased beta cell dedifferentiation in the individuals with diabetes. Clusters of adipocytes significantly contribute to higher pancreatic fat content and immune cell recruitment. Overall, the interactions among adipocytes, immune cells and beta cells in the pancreas microenvironment might contribute to beta cell failure and dedifferentiation in type 2 diabetes.
Collapse
Affiliation(s)
- Na Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiman Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaxin Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ruonan Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Siyi Liu
- Fudan University, Shanghai, China
| | - Xuelian Zhao
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Ma
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaomu Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
255
|
Szukiewicz D. Potential Therapeutic Exploitation of G Protein-Coupled Receptor 120 (GPR120/FFAR4) Signaling in Obesity-Related Metabolic Disorders. Int J Mol Sci 2025; 26:2501. [PMID: 40141148 PMCID: PMC11941992 DOI: 10.3390/ijms26062501] [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: 02/16/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The increasing prevalence of overweight and obesity not only in adults but also among children and adolescents has become one of the most alarming health problems worldwide. Metabolic disorders accompanying fat accumulation during pathological weight gain induce chronic low-grade inflammation, which, in a vicious cycle, increases the immune response through pro-inflammatory changes in the cytokine (adipokine) profile. Obesity decreases life expectancy, largely because obese individuals are at an increased risk of many medical complications, often referred to as metabolic syndrome, which refers to the co-occurrence of insulin resistance (IR), impaired glucose tolerance, type 2 diabetes (T2D), atherogenic dyslipidemia, hypertension, and premature ischemic heart disease. Metabotropic G protein-coupled receptors (GPCRs) constitute the most numerous and diverse group of cell surface transmembrane receptors in eukaryotes. Among the GPCRs, researchers are focusing on the connection of G protein-coupled receptor 120 (GPR120), also known as free fatty acid receptor 4 (FFAR4), with signaling pathways regulating the inflammatory response and insulin sensitivity. This review presents the current state of knowledge concerning the involvement of GPR120 in anti-inflammatory and metabolic signaling. Since both inflammation in adipose tissue and insulin resistance are key problems in obesity, there is a rationale for the development of novel, GPR120-based therapies for overweight and obese individuals. The main problems associated with introducing this type of treatment into clinical practice are also discussed.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
256
|
Giacomello E, Nicoletti C, Canato M, Toniolo L. Exercise Mimetics in Aging: Suggestions from a Systematic Review. Nutrients 2025; 17:969. [PMID: 40289996 PMCID: PMC11944853 DOI: 10.3390/nu17060969] [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: 02/17/2025] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Growth in the aging world population is accompanied by an increase in comorbidities, profoundly impacting the quality of life of older people. This development has motivated a large effort to investigate the mechanisms underlying aging and the search for countermeasures. The most investigated strategies envisage the control of diet and physical exercise, which exploit both common and distinct mechanisms to promote health. Since the application of nutritional and exercise protocols to aged persons introduces several issues due to their disabled state, some strategies have been developed. The nutritional approach exploits a wide range of compounds, including calorie restriction mimetics, supplements, antioxidants, and others. In the context of exercise, in recent years, molecules able to provide similar effects to exercise, the so-called exercise mimetics, have been developed. Methods: To have a better perspective on exercise mimetics and their connection with nutrition, we performed a systematic search of the PubMed and Scopus databases using the term "exercise mimetics". Results: In total, 97 research articles were selected and discussed. The present review provides evidence of the presence of multiple exercise-mimetic compounds and physical strategies that can target metabolic pathways, oxidative stress defense mechanisms, or myokine modulation. Conclusions: Interestingly, this review highlights that an important number of exercise mimetics are represented by products of natural origin and supplements assimilable with diet. This evidence provides a further link between exercise and nutrition and confers a central role on nutrition in the context of exercise mimetics.
Collapse
Affiliation(s)
- Emiliana Giacomello
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Claudio Nicoletti
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy;
| | - Marta Canato
- Laboratory of Muscle Biophysics, Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy;
| | - Luana Toniolo
- Laboratory of Muscle Biophysics, Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy;
| |
Collapse
|
257
|
Wu R, Zhao XJ, Du Y, Dong Y, Song X, Zhu Y. Lipid metabolic disorders and their impact on cartilage endplate and nucleus pulposus function in intervertebral disk degeneration. Front Nutr 2025; 12:1533264. [PMID: 40129665 PMCID: PMC11931516 DOI: 10.3389/fnut.2025.1533264] [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: 11/23/2024] [Accepted: 02/06/2025] [Indexed: 03/26/2025] Open
Abstract
Lipid metabolism encompasses the processes of digestion, absorption, synthesis, and degradation of fats within biological systems, playing a crucial role in sustaining normal physiological functions. Disorders of lipid metabolism, characterized by abnormal blood lipid levels and dysregulated fatty acid metabolism, have emerged as significant contributors to intervertebral disk degeneration (IDD). The pathogenesis of IDD is multifaceted, encompassing genetic predispositions, nutritional and metabolic factors, mechanical stressors, trauma, and inflammatory responses, which collectively facilitate the progression of IDD. Although the precise mechanisms underlying IDD remain incompletely elucidated, there is substantial consensus regarding the close association between lipid metabolism disorders and its development. Intervertebral disks are essential for maintaining spinal alignment. Their primary functions encompass shock absorption, preservation of physiological curvature, facilitation of movement, and provision of stability. The elasticity and thickness of these disks effectively absorb daily impacts, safeguard the spine, uphold its natural curvature and flexibility, while also creating space for nerve roots to prevent compression and ensure normal transmission of nerve signals. Research indicates that such metabolic disturbances may compromise the functionality of cartilaginous endplates (CEP) and nucleus pulposus (NP), thereby facilitating IDD's onset and progression. The CEP is integral to internal material exchange and shock absorption while mitigating NP herniation under mechanical load conditions. As the central component of intervertebral disks, NP is essential for maintaining disk height and providing shock-absorbing capabilities; thus, damage to these critical structures accelerates IDD progression. Furthermore, lipid metabolism disorders contribute to IDD through mechanisms including activation of endoplasmic reticulum stress pathways, enhancement of oxidative stress levels, induction of cellular pyroptosis alongside inhibition of autophagy processes-coupled with the promotion of inflammation-induced fibrosis and fibroblast proliferation leading to calcification within intervertebral disks. This review delineates the intricate interplay between lipid metabolism disorders and IDD; it is anticipated that advancing our understanding of this pathogenesis will pave the way for more effective preventive measures and therapeutic strategies against IDD in future research.
Collapse
Affiliation(s)
- Ruixia Wu
- Inner Mongolia Medical University, Hohhot, China
| | - Xiao Juan Zhao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yaxin Du
- Inner Mongolia Medical University, Hohhot, China
| | - Yizhi Dong
- Inner Mongolia Medical University, Hohhot, China
| | - Xinyue Song
- Inner Mongolia Medical University, Hohhot, China
| | - Yong Zhu
- Peking University Cancer Hospital Inner Mongolia Hospital, Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
258
|
Rivera-Alvarez I, Vázquez-Lizárraga R, Mendoza-Viveros L, Sotelo-Rivera I, Viveros-Ruiz TL, Morales-Maza J, Orozco L, Romano MC, Noriega LG, Tovar AR, Aguilar-Arnal L, Cruz-Bautista I, Aguilar-Salinas C, Orozco-Solis R. Transcriptional dynamics in type 2 diabetes progression is linked with circadian, thermogenic, and cellular stress in human adipose tissue. Commun Biol 2025; 8:398. [PMID: 40057615 PMCID: PMC11890630 DOI: 10.1038/s42003-025-07709-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/10/2025] [Indexed: 04/03/2025] Open
Abstract
The prevalence of type 2 diabetes (T2D) has increased significantly over the past three decades, with an estimated 30-40% of cases remaining undiagnosed. Brown and beige adipose tissues are known for their remarkable catabolic capacity, and their ability to diminish blood glucose plasma concentration. Beige adipose tissue can be differentiated from adipose-derived stem cells or through transdifferentiation from white adipocytes. However, the impact of T2D progression on beige adipocytes' functional capacity remains unclear. Transcriptomic profiling of subcutaneous adipose tissue biopsies from healthy normal-weight, obese, prediabetic obese, and obese subjects diagnosed with T2D, reveals a progressive alteration in cellular processes associated with catabolic metabolism, circadian rhythms, thermogenesis-related signaling pathways, cellular stress, and inflammation. MAX is a potential transcription factor that links inflammation with the circadian clock and thermogenesis during the progression of T2D. This study unveils an unrecognized transcriptional circuit that increasingly disrupts subcutaneous adipose tissue oxidative capacity during the progression of T2D. These findings could open new research venues for developing chrono-pharmaceutical strategies to treat and prevent T2D.
Collapse
Affiliation(s)
| | - Rosa Vázquez-Lizárraga
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), México City, México
| | - Lucía Mendoza-Viveros
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, San Luis Potosi, S.L.P., México
| | | | - Tannia L Viveros-Ruiz
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Jesús Morales-Maza
- Departamento de Cirugía Endocrina, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Lorena Orozco
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
| | - Marta C Romano
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y Estudios Avanzados (CINVESTAV), México City, México
| | - Lilia G Noriega
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Lorena Aguilar-Arnal
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), México City, México
| | - Ivette Cruz-Bautista
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Carlos Aguilar-Salinas
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, México City, México
| | - Ricardo Orozco-Solis
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México.
- Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), México City, México.
| |
Collapse
|
259
|
Wang X, Wang J, Zhao X, Zhang J, Zhang Y. The adipokines in oral cancer pathogenesis and its potential as a new therapeutic approach. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03939-w. [PMID: 40056203 DOI: 10.1007/s00210-025-03939-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 02/16/2025] [Indexed: 03/10/2025]
Abstract
The involvement of adipose tissue in the development of cancer is currently the subject of an increasing number of research due to the growing relevance of lipid metabolism in tumor growth. Obesity influences the tumor immune microenvironment (TME) in oral cancer. Visceral white adipose tissue (WAT) consists of adipocytes, connective tissue, immune cells, and stromovascular cells. The metabolic processes of immune cells within the adipose tissue of individuals with obesity predominantly depend on oxidative phosphorylation (intrinsically) and are characterized by elevated levels of M2 macrophages, Treg cells, Th2 cells, and eosinophils from an extrinsic perspective. The adipokines secreted by adipocytes facilitate communication with adjacent tissues to regulate glucose and lipid metabolism. Obesity influences cancer progression through the dysregulation of adipocytokines, characterized by an augmented synthesis of the oncogenic adipokine leptin, coupled with a reduced secretion of adiponectin. Under standard physiological settings, these adipokines fulfill essential roles in sustaining homeostasis. This review analyzed the influence of adipocytes on oral cancer by detailing the mediators released by adipocytes. Comprehending the molecular foundations of the protumor roles of adipokines in oral cancers might provide novel treatment targets.
Collapse
Affiliation(s)
- Xue Wang
- Department of Stomatology, Jilin Province FAW General Hospital, Jilin, 130000, China
| | - Jiapeng Wang
- Department of Orthopedics, Jilin Province FAW General Hospital, Jilin, 130000, China.
| | - Xuemei Zhao
- Department of Stomatology, Jilin Province FAW General Hospital, Jilin, 130000, China
| | - Jiayin Zhang
- Department of Stomatology, Jilin Province FAW General Hospital, Jilin, 130000, China
| | - Yan Zhang
- Medical Department, Changchun Sci-Tech University, Changchun, 130000, China
| |
Collapse
|
260
|
Khamis MM, Moselhy SS, Rihan S. Role of trans-resveratrol in ameliorating biochemical and molecular alterations in obese rats induced by a high fructose/fat diet. Sci Rep 2025; 15:7879. [PMID: 40050385 PMCID: PMC11885455 DOI: 10.1038/s41598-025-91027-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
We evaluated the effect of trans-resveratrol (RSV) in ameliorating biochemical and molecular alterations in obese Wister male rats fed on high-fat/high-fructose-fed. Male Wister rats were divided into eight groups and fed with either a standard diet (control), high fructose (HF), high fat (HFAT), or a high- fructose high- fat (HF/HFAT) diet and supplemented with RSV (30 mg/kg/day) for 4 weeks. The food intake, body weight, glycemic parameters, lipid profile, oxidative stress were assessed. SIRT1 gene expression, PGC-1α, cyto-c and GLUT-4 were evaluated by qRT-PCR in adipose tissue of normal and obese rats. The body weight gain, serum fasting glucose, insulin, and HOMA-IR values were significantly higher in the HF and HF/HFAT groups than in the HFAT and control groups. Hyperlipidemia was observed in high calorie diets fed rats compared to control group. The levels of total cholesterol, triglycerides and LDL-c were significantly elevated while HDL- c was significantly decreased in HF & HF/HFAT groups compared to HFAT group. The levels of serum malondialdhyde (MDA) and superoxide dismutase (SOD) activity in adipose tissue were elevated in all groups compared to control group, particularly in the groups that were kept on a high fructose diets (HF, HF/HFAT). SIRT-1, PGC-1α, Cyto-c, and GLUT-4 genes levels were significantly down regulated in HF, HFAT & HF/HFAT groups compared to control group. Supplementation of T-RSV restored the alteration in carbohydrates-lipid metabolism as well as oxidative stress and upregulation of SIRT-1, PGC-1α, Cyto-c, and GLUT-4 genes. RSV is a promising treatment in the management of pathologic consequences of obesity from high-calorie diet consumption via molecular alteration of target genes.
Collapse
Affiliation(s)
- Marwa Maher Khamis
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Said Salama Moselhy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Shaimaa Rihan
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
261
|
Zhu M, Peng Q, Li S, Zhang G, Zhang Z. Irisin promotes autophagy and attenuates NLRP3 inflammasome activation in Parkinson's disease. Int Immunopharmacol 2025; 149:114201. [PMID: 39914281 DOI: 10.1016/j.intimp.2025.114201] [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: 12/18/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/22/2025]
Abstract
Parkinson's disease (PD) is characterized by the aggregation and prion-like propagation of α-synuclein (α-syn). Irisin is an exercise-induced myokine that regulates energy metabolism and exerts protective effects in PD by reducing α-syn pathology. However, the molecular mechanisms underlying the role of irisin are not fully understood. Here, we show that irisin inhibits NLRP3 inflammasome activation and promotes autophagy in cultured cells. Additionally, irisin alleviates oxidative stress and reduces cell apoptosis induced by α-syn fibrils. In a PD mouse model induced by intrastriatal injection of α-syn fibrils, irisin mitigated α-syn aggregation, neuroinflammation and neurodegeneration. These observations suggest that irisin functions as a protective mediator against α-syn pathology in PD and that irisin may serve as a potential therapeutic target for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Min Zhu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qinyu Peng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sheng Li
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China.
| |
Collapse
|
262
|
Dixon ED, Claudel T, Trauner M. Reply to "Pharmacological inhibition of ATGL as therapeutic approach for MASH: Far beyond the lipase suppressing role of ATGL inhibitors". J Hepatol 2025:S0168-8278(25)00131-X. [PMID: 40054634 DOI: 10.1016/j.jhep.2025.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 04/07/2025]
Affiliation(s)
- Emmanuel Dauda Dixon
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria.
| |
Collapse
|
263
|
Majeed S, Moin H, Waseem M, Khalid Z, Wajid Abbasi S, Rasool K. Identification of candidate nsSNPs of the human FNDC5 gene and their structural and functional consequences using in silico analysis. Sci Rep 2025; 15:7681. [PMID: 40044721 PMCID: PMC11882896 DOI: 10.1038/s41598-024-83254-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/12/2024] [Indexed: 03/09/2025] Open
Abstract
Fibronectin type-III domain containing protein-5 (FNDC5), predominantly expressed in skeletal muscles, encodes FNDC5 transmembrane-protein. A segment of this protein is cleaved and secreted into blood as irisin, which promotes browning of white adipose tissue, leading to energy expenditure. It functions synergistically with fibroblast growth factor-21 (FGF21). Irisin is considered as a potential target for treating obesity-related disorders. Likewise, FNDC5 variations can contribute to development of such disorders. This study aimed to identify putative non-synonymous single nucleotide polymorphisms (nsSNPs) of human FNDC5, potentially impacting FNDC5-FGF21 interaction. Sequence and structure based computational tools were used to identify nsSNPs of FNDC5, which revealed eight nsSNPs as being most deleterious (N39K, R78H, R209H, T124I, L150P, L156V, V83M, and T86I). Molecular-docking was performed to analyze the impact of FNDC5 mutations on wild-type and mutant FNDC5-FGF21 complexes, revealing that T124I (rs185141197) and L150P (rs377741902) showed higher buried surface area (BSA) than wild-type. Following this, molecular dynamic (MD) simulation further affirmed the findings and revealed that T124I induced conformational changes in the irisin domain of FNDC5, which may significantly affect its binding with protein FGF21, potentially impairing synergistic effects of FNDC5 and FGF21 on adipocyte browning and increasing risk for developing obesity and related disorders.
Collapse
Affiliation(s)
- Sadaf Majeed
- Department of Biomedical Sciences, Dubai Medical College for Girls, Dubai, United Arab Emirates
| | - Hira Moin
- Department of Physiology, NUST School of Health Sciences, National University of Sciences and Technology, Islamabad, 44000, Pakistan.
| | - Maaz Waseem
- National University of Sciences and Technology, Islamabad, 44000, Pakistan
- School of Biological Sciences, University of the Punjab, Lahore, 05422, Pakistan
| | - Zoya Khalid
- Department of Biosciences, COMSATS University, Islamabad, 44000, Pakistan.
| | - Sumra Wajid Abbasi
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, 44000, Pakistan
| | - Kashaf Rasool
- National University of Sciences and Technology, Islamabad, 44000, Pakistan
| |
Collapse
|
264
|
Craveiro V, Severo M, Ramos E. Association between adiposity distribution and low-grade systemic inflammation: Tackling multicollinearity. Nutrition 2025; 135:112737. [PMID: 40188501 DOI: 10.1016/j.nut.2025.112737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 04/08/2025]
Abstract
OBJECTIVES To examine the association between adiposity distribution and low-grade systemic inflammation, testing how the statistical approach can be used to tackle the multicollinearity between variables of adiposity. METHODS Cross-sectional analysis of 987 adults (27 years old) from the EPITeen cohort (Porto, Portugal). Adiposity distribution was assessed based on the fat distribution of the participants, measured by whole-body dual-energy X-ray absorptiometry (DXA). Low-grade systemic inflammation was determined based on the serum high-sensitivity C-reactive protein (hsCRP) of the participants. The associations were estimated by linear regression models, stratified by sex, and multicollinearity between variables of adiposity was considered. RESULTS The association (standardized β [95%CI]) between total body fat and hsCRP (ln) was 0.332 (0.252, 0.411) in women and 0.256 (0.175, 0.337) in men. Finally, in women, the association (standardized β [95%CI]) with hsCRP (ln) was -0.014 (-0.094, 0.065) for limb fat and 0.019 (-0.061, 0.099) for trunk fat, and, in men it was -0.136 (-0.216, -0.056) for limb fat and 0.130 (0.050, 0.211) for trunk fat. CONCLUSIONS After a comprehensive study of fat, its overall proinflammatory effect was acknowledged. Trunk fat was more proinflammatory than limb fat, even though the results were only statistically significant in men. Statistical analysis should consider multicollinearity between variables of adiposity, though simpler models could be used to address this problem.
Collapse
Affiliation(s)
- Vanda Craveiro
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal; Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal.
| | - Milton Severo
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal; Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal; Departamento de Ensino Médico Pré-Graduado, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Elisabete Ramos
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal; Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal; Departamento de Ciências da Saúde Pública e Forenses, e Educação Médica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
265
|
Reinisch I, Ghosh A, Noé F, Sun W, Dong H, Leary P, Dietrich A, Hoffmann A, Blüher M, Wolfrum C. Unveiling adipose populations linked to metabolic health in obesity. Cell Metab 2025; 37:640-655.e4. [PMID: 39694039 DOI: 10.1016/j.cmet.2024.11.006] [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: 02/09/2024] [Revised: 08/06/2024] [Accepted: 11/10/2024] [Indexed: 12/20/2024]
Abstract
Precision medicine is still not considered as a standard of care in obesity treatment, despite a large heterogeneity in the metabolic phenotype of individuals with obesity. One of the strongest factors influencing the variability in metabolic disease risk is adipose tissue (AT) dysfunction; however, there is little understanding of the link between distinct cell populations, cell-type-specific transcriptional programs, and disease severity. Here, we generated a comprehensive cellular map of subcutaneous and visceral AT of individuals with metabolically healthy and unhealthy obesity. By combining single-nucleus RNA-sequencing data with bulk transcriptomics and clinical parameters, we identified that mesothelial cells, adipocytes, and adipocyte-progenitor cells exhibit the strongest correlation with metabolic disease. Furthermore, we uncovered cell-specific transcriptional programs, such as the transitioning of mesothelial cells to a mesenchymal phenotype, that are involved in uncoupling obesity from metabolic disease. Together, these findings provide valuable insights by revealing biological drivers of clinical endpoints.
Collapse
Affiliation(s)
- Isabel Reinisch
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Adhideb Ghosh
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland; Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Falko Noé
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland; Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Wenfei Sun
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Hua Dong
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland; Stem Cell Bio Regenerative Med Institute, Stanford University, Stanford, CA, USA
| | - Peter Leary
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Arne Dietrich
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany; Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
266
|
Slattery JD, Rambousek JR, Tsui E, Honeycutt MK, Goldberg M, Graham JL, Wietecha TA, Wolden-Hanson T, Williams AL, O’Brien KD, Havel PJ, Blevins JE. Effects of systemic oxytocin and beta-3 receptor agonist (CL 316243) treatment on body weight and adiposity in male diet-induced obese rats. Front Endocrinol (Lausanne) 2025; 16:1503096. [PMID: 40104132 PMCID: PMC11913664 DOI: 10.3389/fendo.2025.1503096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/31/2025] [Indexed: 03/20/2025] Open
Abstract
Previous studies have implicated hindbrain oxytocin (OT) receptors in the control of food intake and brown adipose tissue (BAT) thermogenesis. We recently demonstrated that hindbrain [fourth ventricle (4V)] administration of oxytocin (OT) could be used as an adjunct to drugs that directly target beta-3 adrenergic receptors (β3-AR) to elicit weight loss in diet-induced obese (DIO) rodents. What remains unclear is whether systemic OT can be used as an adjunct with the β3-AR agonist, CL 316243, to increase BAT thermogenesis and elicit weight loss in DIO rats. We hypothesized that systemic OT and β3-AR agonist (CL 316243) treatment would produce an additive effect to reduce body weight and adiposity in DIO rats by decreasing food intake and stimulating BAT thermogenesis. To test this hypothesis, we determined the effects of systemic (subcutaneous) infusions of OT (50 nmol/day) or vehicle (VEH) when combined with daily systemic (intraperitoneal) injections of CL 316243 (0.5 mg/kg) or VEH on body weight, adiposity, food intake and brown adipose tissue temperature (TIBAT). OT and CL 316243 monotherapy decreased body weight by 8.0 ± 0.9% (P<0.05) and 8.6 ± 0.6% (P<0.05), respectively, but OT in combination with CL 316243 produced more substantial weight loss (14.9 ± 1.0%; P<0.05) compared to either treatment alone. These effects were associated with decreased adiposity, energy intake and elevated TIBAT during the treatment period. The findings from the current study suggest that the effects of systemic OT and CL 316243 to elicit weight loss are additive and appear to be driven primarily by OT-elicited changes in food intake and CL 316243-elicited increases in BAT thermogenesis.
Collapse
Affiliation(s)
- Jared D. Slattery
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - June R. Rambousek
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Edison Tsui
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Mackenzie K. Honeycutt
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Matvey Goldberg
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - James L. Graham
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Tomasz A. Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington (UW) School of Medicine, Seattle, WA, United States
- University of Washington (UW) Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA, United States
| | - Tami Wolden-Hanson
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Amber L. Williams
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
| | - Kevin D. O’Brien
- University of Washington (UW) Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA, United States
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Peter J. Havel
- Department of Nutrition, University of California, Davis, Davis, CA, United States
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - James E. Blevins
- Veterans Affairs (VA) Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington (UW) School of Medicine, Seattle, WA, United States
| |
Collapse
|
267
|
Dumesic PA, Wilensky SE, Bose S, Van Vranken JG, Gygi SP, Spiegelman BM. RBM43 controls PGC1α translation and a PGC1α-STING signaling axis. Cell Metab 2025; 37:742-757.e8. [PMID: 39965564 PMCID: PMC11885043 DOI: 10.1016/j.cmet.2025.01.013] [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: 01/30/2024] [Revised: 10/17/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025]
Abstract
Obesity is associated with systemic inflammation that impairs mitochondrial function. This disruption curtails oxidative metabolism, limiting adipocyte lipid metabolism and thermogenesis, a metabolically beneficial program that dissipates chemical energy as heat. Here, we show that PGC1α, a key governor of mitochondrial biogenesis, is negatively regulated at the level of its mRNA translation by the RNA-binding protein RBM43. RBM43 is induced by inflammatory cytokines and suppresses mitochondrial biogenesis in a PGC1α-dependent manner. In mice, adipocyte-selective Rbm43 disruption elevates PGC1α translation and oxidative metabolism. In obesity, Rbm43 loss improves glucose tolerance, reduces adipose inflammation, and suppresses activation of the innate immune sensor cGAS-STING in adipocytes. We further identify a role for PGC1α in safeguarding against cytoplasmic accumulation of mitochondrial DNA, a cGAS ligand. The action of RBM43 defines a translational regulatory axis by which inflammatory signals dictate cellular energy metabolism and contribute to metabolic disease pathogenesis.
Collapse
Affiliation(s)
- Phillip A Dumesic
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah E Wilensky
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Symanthika Bose
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
268
|
Shimura H, Yamamoto S, Shiiba I, Oikawa M, Uchinomiya S, Ojida A, Yanagi S, Kadowaki H, Nishitoh H, Fukuda T, Nagashima S, Yamaguchi T. Etomoxir suppresses the expression of PPARγ2 and inhibits the thermogenic gene induction of brown adipocytes through pathways other than β-oxidation inhibition. J Biochem 2025; 177:203-212. [PMID: 39727324 DOI: 10.1093/jb/mvae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024] Open
Abstract
Brown adipocytes are characterized by a high abundance of mitochondria, allowing them to consume fatty acids for heat production. Increasing the number of brown adipocytes is considered a promising strategy for combating obesity. However, the molecular mechanisms underlying their differentiation remain poorly understood. In this study, we demonstrate that etomoxir, an inhibitor of Carnitine Palmitoyltransferase 1 (CPT1), inhibits their differentiation through mechanisms independent of β-oxidation inhibition. In the presence of etomoxir during brown adipocyte differentiation, reduced expression of the thermogenic gene UCP1 and decreased lipid droplets formation were observed. Furthermore, a transient reduction in the expression of PPARγ2, a critical factor in adipocyte differentiation, was also observed in the presence of etomoxir. These findings suggest the presence of a regulatory mechanism that specifically enhances PPARγ2 expression during brown adipocyte differentiation, thereby modulating thermogenic gene expression.
Collapse
Affiliation(s)
- Hiroki Shimura
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Sota Yamamoto
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Isshin Shiiba
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima, Tokyo 171-8588, Japan
| | - Mami Oikawa
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Shohei Uchinomiya
- Medical Chemistry and Chemical Biology, Department of Medicinal Sciences, Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka, Fukuoka 812-8582, Japan
| | - Akio Ojida
- Medical Chemistry and Chemical Biology, Department of Medicinal Sciences, Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka, Fukuoka 812-8582, Japan
| | - Shigeru Yanagi
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima, Tokyo 171-8588, Japan
| | - Hisae Kadowaki
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Sciences, University of Miyazaki, Miyazaki, Miyazaki 889-1692, Japan
| | - Hideki Nishitoh
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Sciences, University of Miyazaki, Miyazaki, Miyazaki 889-1692, Japan
- Frontier Science Research Center, University of Miyazaki, Miyazaki, Miyazaki 889-1692, Japan
| | - Toshifumi Fukuda
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Shun Nagashima
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Tomoyuki Yamaguchi
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
269
|
Wang Y, Ma Z, Jiang L, Bojan N, Sha Y, Huang B, Ming L, Shen J, Pang W. Specific muscle targeted delivery of miR-130a loaded lipid nanoparticles: a novel approach to inhibit lipid accumulation in skeletal muscle and obesity. J Nanobiotechnology 2025; 23:159. [PMID: 40033366 DOI: 10.1186/s12951-025-03225-0] [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: 09/30/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Skeletal muscle lipid deposition is a key manifestation of obesity, often accompanied by decreased exercise capacity and muscle atrophy. Skeletal muscle as the largest organ in the body, makes it challenges for designing targeted drug delivery systems. Lipid nanoparticles (LNPs) are widely used as a safe and efficient delivery carrier, there is limited research on LNPs that specifically target skeletal muscle. RESULTS A LNP designed with five specific receptor complements on its surface, which specifically targets skeletal muscle in vivo in mice, without off-target effects on other tissues and organs. MiR-130a, a regulator of PPARG, which is a key factor in skeletal muscle lipid deposition, was encapsulated with LNP (LNP@miR-130a). In high-fat diet (HFD) mice, LNP@miR-130a effectively reduced skeletal muscle lipid deposition, increased exercise activity and enhanced muscle mass. Interestingly, the myokines in skeletal muscle have also changed which may leading to reduce the adipose tissue weight and liver lipid deposition in HFD mice. CONCLUSIONS These results indicated LNP@miR-130a is a promising inhibitor of skeletal muscle lipid deposition and may help alleviate obesity. This study provides new insights for obesity treatment and lays foundation for the development of targeted skeletal muscle therapeutics.
Collapse
Affiliation(s)
- Yingqian Wang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Shaanxi, China
| | - Zeqiang Ma
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Shaanxi, China
| | - Lehua Jiang
- Center for Metabolic & Gastroenterology, Institute of Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Nataraj Bojan
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Shaanxi, China
| | - Yiwen Sha
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Shaanxi, China
| | - Boyu Huang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Shaanxi, China
| | - Lianxi Ming
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Shaanxi, China
| | - Junnan Shen
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Shaanxi, China
| | - Weijun Pang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Shaanxi, China.
- , No. 22 Xinong Road, Yangling, Shaanxi Province, 712100, China.
| |
Collapse
|
270
|
Bezawork-Geleta A, Devereux CJ, Keenan SN, Lou J, Cho E, Nie S, De Souza DP, Narayana VK, Siddall NA, Rodrigues CHM, Portelli S, Zheng T, Nim HT, Ramialison M, Hime GR, Dodd GT, Hinde E, Ascher DB, Stroud DA, Watt MJ. Proximity proteomics reveals a mechanism of fatty acid transfer at lipid droplet-mitochondria- endoplasmic reticulum contact sites. Nat Commun 2025; 16:2135. [PMID: 40032835 DOI: 10.1038/s41467-025-57405-5] [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: 04/11/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
Membrane contact sites between organelles are critical for the transfer of biomolecules. Lipid droplets store fatty acids and form contacts with mitochondria, which regulate fatty acid oxidation and adenosine triphosphate production. Protein compartmentalization at lipid droplet-mitochondria contact sites and their effects on biological processes are poorly described. Using proximity-dependent biotinylation methods, we identify 71 proteins at lipid droplet-mitochondria contact sites, including a multimeric complex containing extended synaptotagmin (ESYT) 1, ESYT2, and VAMP Associated Protein B and C (VAPB). High resolution imaging confirms localization of this complex at the interface of lipid droplet-mitochondria-endoplasmic reticulum where it likely transfers fatty acids to enable β-oxidation. Deletion of ESYT1, ESYT2 or VAPB limits lipid droplet-derived fatty acid oxidation, resulting in depletion of tricarboxylic acid cycle metabolites, remodeling of the cellular lipidome, and induction of lipotoxic stress. These findings were recapitulated in Esyt1 and Esyt2 deficient mice. Our study uncovers a fundamental mechanism that is required for lipid droplet-derived fatty acid oxidation and cellular lipid homeostasis, with implications for metabolic diseases and survival.
Collapse
Affiliation(s)
| | - Camille J Devereux
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Stacey N Keenan
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Jieqiong Lou
- School of Physics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Ellie Cho
- Biological Optical Microscopy Platform (BOMP), The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility (MMSPF), Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3052, Australia
| | - David P De Souza
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Vinod K Narayana
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Nicole A Siddall
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Carlos H M Rodrigues
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Stephanie Portelli
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Tenghao Zheng
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Hieu T Nim
- Murdoch Children's Research Institute, reNEW Novo Nordisk Foundation for Stem Cell Medicine, Melbourne, VIC, 3052, Australia
| | - Mirana Ramialison
- Murdoch Children's Research Institute, reNEW Novo Nordisk Foundation for Stem Cell Medicine, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Gary R Hime
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Garron T Dodd
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Elizabeth Hinde
- School of Physics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - David B Ascher
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - David A Stroud
- Murdoch Children's Research Institute, reNEW Novo Nordisk Foundation for Stem Cell Medicine, Melbourne, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3052, Australia
- Victorian Clinical Genetics Services, Royal Children's Hospital, Melbourne, VIC, 3052, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
271
|
Schiano ME, Billi C, Grillo G, Tkachuk O, De Caro C, Russo E, Comella F, Meli R, Frecentese F, Santagada V, Cinque P, Albrizio S, Persico M, Sodano F, Fattorusso C, Rimoli MG. Eco-friendly Synthesis and Molecular Modelling of 2-Phenylimidazo[1,2-b]pyridazine Derivatives: In Vitro and In Vivo Studies for Lead Optimization. ChemMedChem 2025; 20:e202400721. [PMID: 39561040 DOI: 10.1002/cmdc.202400721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/31/2024] [Accepted: 11/19/2024] [Indexed: 11/21/2024]
Abstract
7-methyl-2-phenylimidazo[1,2-b]pyridazin-3-carboxylic acid (DM1) and 6-methoxy-2-phenylimidazo[1,2-b]pyridazin-3-carboxylic acid (DM2) have been shown to act as human (h) Cav3.1 voltage-gated calcium channel blockers with promising in vivo anti-absence activity, positioning them as potential antiepileptic drugs. The primary aim of this work was to develop cost-effective and environmentally friendly synthetic procedures for preparing 2-phenylimidazo[1,2-b]pyridazine derivatives. After optimizing the synthesis of this compound class using efficient and green techniques such as microwaves and ultrasound irradiation, we further evaluated the antiepileptic effects of DM1 and DM2 in two animal models: CD-1 ICR mice after pentylenetetrazol administration and DBA/2 mice with seizures induced by audiogenic stimuli. Their neuroprotective effect against oxidative stress were assessed using C6 rat brain glioma cells. DM1 and DM2 exhibited potent anti-seizure effects in both animal models and demonstrated significant in vitro neuroprotective activity by reducing reactive oxygen species release. To lay the groundwork for the future rational optimization of this promising class of compounds, the molecular bases of DM1 and DM2 activity were investigated by modelling their interaction with hCav3.1 channels. The calculated binding modes of DM1 and DM2 to hCav3.1 channels partially mirrored that of the selective Cav3.1 blocker Z944, paving the way for future lead optimization.
Collapse
Affiliation(s)
- Marica Erminia Schiano
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Chiara Billi
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Giorgio Grillo
- Department of Drug Science and Technology, University of Torino, 10125, Torino, Italy
| | - Oleh Tkachuk
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Carmen De Caro
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
- Department of Science of Health, University "Magna Graecia" of Catanzaro, 88100, Catanzaro, Italy
| | - Emilio Russo
- Department of Science of Health, University "Magna Graecia" of Catanzaro, 88100, Catanzaro, Italy
| | - Federica Comella
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Rosaria Meli
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Francesco Frecentese
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Vincenzo Santagada
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Pierfrancesco Cinque
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Stefania Albrizio
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Marco Persico
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Federica Sodano
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Caterina Fattorusso
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Maria Grazia Rimoli
- Department of Pharmacy, "Federico II" University of Napoli, Via Domenico Montesano 49, 80131, Napoli, Italy
| |
Collapse
|
272
|
Zhang J, Shen M, Yin Y, Chen Y, Deng X, Mo J, Zhou X, Lin J, Chen X, Xie X, Wu X, Chen X. Carnosic acid reduces lipid content, enhances gut health, and modulates microbiota composition and metabolism in diet-induced obese mice. Food Funct 2025; 16:1888-1902. [PMID: 39932492 DOI: 10.1039/d4fo04534c] [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: 03/04/2025]
Abstract
Carnosic acid (CA) is a bioactive phenolic diterperne compound found in sage and rosemary. The present study investigated the beneficial effects of CA (50 and 100 mg per kg bw) in diet-induced obese mice and the underlying mechanisms of action. After the intervention, the physiology, lipid metabolism, and tissue morphology, as well as the inflammation, gut microbiota, and metabolomics in the colon were measured. We found that CA improved the composition and metabolism of the gut microbiota in obese mice, with Akkermansia being the dominant bacterium negatively correlated with obesity and various fecal metabolites. Regarding the intestinal barrier function, CA promoted the expression of tight junction proteins and inhibited the TLR4/MyD88/NF-κB signaling pathway in obese mice to alleviate colonic inflammation. These results suggest that CA improved multiple aspects of gut health in diet-induced obesity in mice, providing a scientific basis for future clinical studies in humans.
Collapse
Affiliation(s)
- Jing Zhang
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Mengzhu Shen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yue Yin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yuru Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xianying Deng
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Jingyun Mo
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xiaoling Zhou
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Juanying Lin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinxin Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinwei Xie
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xian Wu
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, Ohio 45056, USA.
| | - Xuexiang Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| |
Collapse
|
273
|
Nikolova D, Kamenov Z, Hristova J, Gateva AT. Levels of DEFA1, Progranulin, and NRG4 in Patients with Autonomic Neuropathy: Potential Biomarkers for Diagnosis and Prognosis. Metabolites 2025; 15:169. [PMID: 40137134 PMCID: PMC11944139 DOI: 10.3390/metabo15030169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Diabetic autonomic neuropathy (DAN) is a severe complication of diabetes that affects the autonomic nervous system, impacting cardiovascular, gastrointestinal, genitourinary, and other systems. This study examines the levels of three potential biomarkers-DEFA1, progranulin, and NRG4-to assess their diagnostic and prognostic value in DAN patients. METHODS This observational, single-center study included 80 patients with type 2 diabetes. Clinical data and laboratory results were collected, and serum levels of DEFA1, progranulin, and NRG4 were measured using ELISA. The presence of DAN was assessed using Ewing's tests. Statistical analyses included t-tests, Pearson's correlations, and ROC analysis to explore associations and the predictive values of the biomarkers. RESULTS Progranulin levels were significantly elevated in patients with DAN compared to those without (p < 0.05), showing a positive correlation with diabetes duration (r = 0.375; p = 0.01) and a significant predictive value for DAN (AUC = 0.666; p = 0.013). DEFA1 and NRG4 levels did not differ significantly between the groups. Progranulin was also higher in patients who were treated with sulfonylureas and GLP-1 receptor agonists and in those with coronary artery disease. CONCLUSIONS Progranulin emerges as a potential biomarker for the presence and severity of DAN, correlating with disease duration and autonomic dysfunction. While DEFA1 and NRG4 showed no significant association, the findings underscore the importance of further exploring the inflammatory pathways in DAN. Progranulin measurement could enhance early diagnosis and personalized management of autonomic neuropathy in diabetes.
Collapse
Affiliation(s)
- Diana Nikolova
- Department of Internal Medicine, Aleksandrovska University Hospital, Medical University of Sofia, 1431 Sofia, Bulgaria; (Z.K.); (A.T.G.)
| | - Zdravko Kamenov
- Department of Internal Medicine, Aleksandrovska University Hospital, Medical University of Sofia, 1431 Sofia, Bulgaria; (Z.K.); (A.T.G.)
| | - Julieta Hristova
- Department of Clinical Laboratory, Aleksandrovska University Hospital, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Antoaneta Trifonova Gateva
- Department of Internal Medicine, Aleksandrovska University Hospital, Medical University of Sofia, 1431 Sofia, Bulgaria; (Z.K.); (A.T.G.)
| |
Collapse
|
274
|
Ma C, Guo Y, Tu T, Cui S, Zhong J, Zhang Y, Song N, Liu H. ELF5 gene promotes milk lipid synthesis in goat mammary epithelial cells by transcriptomic analysis. Genomics 2025; 117:111023. [PMID: 40015575 DOI: 10.1016/j.ygeno.2025.111023] [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: 12/08/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
E74-like factor 5 (ELF5) is an Ets transcription factor of epithelial development, while the function of ELF5 gene in goat milk fat synthesis remains to be elucidated. In goat mammary epithelial cells, we performed RNA sequencing and analyzed differentially expressed genes (DEGs) after ELF5 gene overexpression. ELF5 gene significantly up-regulated the synthesis of triglyceride, total cholesterol, free fatty acid, and lipid droplets. We obtained 929 DEGs after ELF5 gene overexpression in GMECs. Among the DEGs, SPP1, ELOVL1, PNPLA2, FOXO1, PTGS2, SEMA6A, ACSL5, and GPNMB genes that are related to lipid metabolism were identified. Enrichment analysis showed MAPK and FoxO signaling pathways were up-regulated by ELF5 gene overexpression in GMECs. These findings offer evidence that ELF5 gene could be a candidate gene for the regulation of milk lipid synthesis in goats, and provide molecular targets for the breeding of goats with high milk fat.
Collapse
Affiliation(s)
- Cunxia Ma
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Germplasm Innovation, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yuzhu Guo
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Germplasm Innovation, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Tongtong Tu
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Germplasm Innovation, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Shuangshuang Cui
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Germplasm Innovation, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Jintao Zhong
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Germplasm Innovation, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yunhai Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Germplasm Innovation, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Ning Song
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Germplasm Innovation, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.
| | - Hongyu Liu
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Germplasm Innovation, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
275
|
Herouvi D, Vlachopapadopoulou EA, Vakaki M, Gouriotis D, Marmarinos A, Kalpia C, Kossiva L, Tsolia M, Karavanaki K. Relation of serum irisin levels with adiposity, components of metabolic syndrome and carotid intima media thickness in prepubertal children with obesity: a cross-sectional study. Endocrine 2025; 87:1031-1040. [PMID: 39725768 DOI: 10.1007/s12020-024-04139-x] [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: 10/14/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
AIM Irisin, a newly discovered adipomyokine, has pleiotropic effects in metabolic and energy homeostasis, insulin resistance (IR), and browning of white adipose tissue. The aim of this study was to evaluate irisin levels in children with obesity and also to elucidate possible relationships between irisin with anthropometric obesity indices, parameters of metabolic syndrome (MetS), and intima media thickness (IMT). METHODS A total of 77 prepubertal children, 4-12 years old, were enrolled in this study, including 44 children with obesity (BMI ≥ 95th percentile) and 33 normal weight controls of matched age and gender. Detailed clinical examination, anthropometric parameters, laboratory data, including serum irisin levels, using ELISA technique and cIMT measurement were carried out in all subjects. RESULTS Children with obesity had significantly higher values of irisin compared to controls (p = 0.003) independently of age, gender, or IR status. Irisin levels were positively correlated with weight z-score, Body Mass Index (BMI), BMI z-score, % Body Fat, waist circumference (WC), triglycerides (TG), and HOMA-IR (p = 0.016, p = 0.025, p = 0.028, p = 0.035, p = 0.019, p = 0.049, p = 0.007 respectively) and inversely correlated with HDL (p = 0.037). In multiple regression analysis irisin levels were strongly associated with excess adiposity (p < 0.001) and uric acid (p = 0.054). CONCLUSION Children with obesity showed an unfavorable cardiometabolic profile and higher levels of IMT and irisin. Moreover, irisin was correlated with metabolic parameters, suggesting that irisin can serve as a prognostic index for future development of MetS in children with obesity.
Collapse
Affiliation(s)
- D Herouvi
- Department of Endocrinology-Growth and Development, "P. & A. Kyriakou" Children's Hospital, Athens, Greece.
| | - E A Vlachopapadopoulou
- Department of Endocrinology-Growth and Development, "P. & A. Kyriakou" Children's Hospital, Athens, Greece
| | - M Vakaki
- Department of Radiology, "P. & A. Kyriakou" Children's Hospital, Athens, Greece
| | - D Gouriotis
- Biochemical Laboratory, 2nd Department of Pediatrics, "P. & A. Kyriakou" Children's Hospital, Athens, Greece
| | - A Marmarinos
- Biochemical Laboratory, 2nd Department of Pediatrics, "P. & A. Kyriakou" Children's Hospital, Athens, Greece
| | - C Kalpia
- Department of Endocrinology-Growth and Development, "P. & A. Kyriakou" Children's Hospital, Athens, Greece
| | - L Kossiva
- 2nd Department of Pediatrics, "P. & A. Kyriakou" Children's Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - M Tsolia
- 2nd Department of Pediatrics, "P. & A. Kyriakou" Children's Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - K Karavanaki
- Diabetes & Metabolism Clinic, 2nd Department of Pediatrics, "P. & A. Kyriakou" Children's Hospital, Athens, Greece
| |
Collapse
|
276
|
Dragoni F, Garofalo M, Di Gerlando R, Rizzo B, Bordoni M, Scarian E, Viola C, Bettoni V, Fiamingo G, Tornabene D, Scanu L, Pansarasa O, Diamanti L, Gagliardi S. Whole transcriptome analysis of unmutated sporadic ALS patients' peripheral blood reveals phenotype-specific gene expression signature. Neurobiol Dis 2025; 206:106823. [PMID: 39904421 DOI: 10.1016/j.nbd.2025.106823] [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: 10/02/2024] [Revised: 01/29/2025] [Accepted: 01/29/2025] [Indexed: 02/06/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult neurodegenerative disorder. According to clinical criteria, ALS patients can be classified into eight subgroups: classic, bulbar, pyramidal, pure lower motor neuron, flail arm, pure upper motor neuron, flail leg, and respiratory. There are no well-established molecular biomarkers for early diagnosis, prognosis, and progression monitoring of this fatal disease. Classification based on clinical phenotypes could be associated with peculiar gene expression patterns shaped during lifespan, allowing the identification of specific sporadic ALS (sALS) subtypes with less heterogeneous clinical and biological features. Our objective was to define a phenotype-specific transcriptomic signature of distinct ALS phenotypes, and lay the foundation for biomarkers development. We characterized 48 sALS patients by clinical and paraclinical parameters, and subdivided them in "Classic" (n = 12), "Bulbar" (n = 10), "Flail Arm" (n = 7), "Flail Leg" (n = 10) and "Pyramidal" (n = 9) phenotypes. RNAs extracted from patients' PBMCs and 19 controls were sequenced. Our analysis allowed the visualization of gene expression differential clusters between patients and controls. Interestingly, only one gene (Y3_RNA, a misc_RNA component of the Ro60 ribonucleoprotein involved in cellular response to interferon-alpha) was upregulated at different levels across all phenotypes, whereas other genes appeared phenotype-specific. The work proposed stress the innovative view of ALS as a multi-systemic disorder rather than a pure motor neuron-associated and 'neurocentric' pathology. The possibility to cluster ALS patients based on their molecular signature pave the way for future personalized clinical trials and early diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Camilla Viola
- IRCCS Mondino Foundation, Pavia, Italy; University of Pavia, Pavia, Italy
| | - Veronica Bettoni
- IRCCS Mondino Foundation, Pavia, Italy; University of Pavia, Pavia, Italy
| | - Giuseppe Fiamingo
- IRCCS Mondino Foundation, Pavia, Italy; University of Pavia, Pavia, Italy
| | - Danilo Tornabene
- IRCCS Mondino Foundation, Pavia, Italy; University of Pavia, Pavia, Italy
| | - Lucia Scanu
- IRCCS Mondino Foundation, Pavia, Italy; University of Pavia, Pavia, Italy
| | | | | | | |
Collapse
|
277
|
Ding Y, Lin F, Liu Z, Zhou X, Liang X. Targeting Epicardial/Pericardial Adipose Tissue in Cardiovascular Diseases: A Novel Therapeutic Strategy. Rev Cardiovasc Med 2025; 26:26128. [PMID: 40160564 PMCID: PMC11951288 DOI: 10.31083/rcm26128] [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: 08/14/2024] [Revised: 11/01/2024] [Accepted: 11/14/2024] [Indexed: 04/02/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain a global health concern, prompting ongoing research into novel contributors to their pathogenesis. Due to the proximity of the coronary arteries and the myocardium in epicardial adipose tissue (EAT) and pericardial adipose tissue (PAT), these tissues have emerged as key areas of interest for their potential influence on cardiac function and vascular health. This review synthesizes current research on the physiological and biological characteristics of EAT and PAT, exploring their composition and clinical measurement approaches. The roles of EAT and PAT in coronary artery disease (CAD), atrial fibrillation, and heart failure are discussed, and the contributions of EAT and PAT to these cardiovascular conditions are highlighted alongside their potential as therapeutic targets.
Collapse
Affiliation(s)
- Yue Ding
- Department of Organ Transplantation, Changzheng Hospital, Second Military Medical University, 200003 Shanghai, China
| | - Fang Lin
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Zhongmin Liu
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Xiaohui Zhou
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Xiaoting Liang
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| |
Collapse
|
278
|
Zeng X, Wang Y, Farias K, Rappa A, Darko C, Sauve A, Huang Q, Alonso LC, Yang Y. NRH, a potent NAD + enhancer, improves glucose homeostasis and lipid metabolism in diet-induced obese mice through an active adenosine kinase pathway. Metabolism 2025; 164:156110. [PMID: 39710001 PMCID: PMC11788054 DOI: 10.1016/j.metabol.2024.156110] [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: 08/12/2024] [Revised: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
AIMS NAD+ deficiency underlies obesity-induced metabolic disturbances. This study evaluated dihydronicotinamide riboside (NRH), a potent NAD+ enhancer, in lean and obese mice and explored whether NRH operates through a unique mechanism involving adenosine kinase (ADK), an enzyme critical for NRH-driven NAD+ synthesis. METHODS Pharmacokinetic and pharmacodynamic analyses were performed following a single 250 mg/kg intraperitoneal injection of NRH in healthy mice. In long-term studies, lean and high-fat diet-induced obese mice were treated with 250 mg/kg NRH thrice weekly for 7 weeks. Blood NAD+ levels, body composition, energy expenditure, and glucose and lipid metabolism were monitored. To test ADK's role, the ADK inhibitor ABT702 was co-administered with NRH in obese mice. RESULTS NRH entered tissues unassisted and was rapidly metabolized for NAD+ biosynthesis, while ADK inhibition blocked its phosphorylation, leading to NRH accumulation in all examined tissues and possible release back into circulation. The 7-week NRH administration was well-tolerated in both lean and obese mice. In obese mice, NRH improved glucose homeostasis by boosting insulin secretion, enhancing muscle insulin signaling, and reducing hepatic gluconeogenesis. It also lowered fat mass, decreased serum lipids, and improved white adipose function. These benefits were linked to elevated tissue NAD+ levels, enhanced Sirtuin activities, and increased mitochondrial antioxidant defenses. ADK inhibition abolished these effects, confirming that NRH's direct entry into tissues and subsequent phosphorylation is essential for its full benefits. CONCLUSION This study establishes NRH as a promising therapeutic agent for obesity-induced metabolic dysfunction, correcting glucose intolerance and hyperlipidemia through ADK-dependent NAD+ enhancement.
Collapse
Affiliation(s)
- Xinliu Zeng
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America; Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yongjie Wang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America; Department of Animal Sciences, North Carolina A&T State University, Greensboro, NC 27411, United States of America
| | - Karina Farias
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Andrew Rappa
- Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Christine Darko
- Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Anthony Sauve
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Qingxia Huang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America; Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Laura C Alonso
- Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Yue Yang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America
| |
Collapse
|
279
|
Piana K, Ziomber-Lisiak A, Ruszczycki B, Bugajski A, Szczerbowska-Boruchowska M. Effects of high-calorie diet-induced obesity on molecular structures of lipids and proteins - A multi-organ study using FTIR spectroscopy. Arch Biochem Biophys 2025; 765:110325. [PMID: 39894381 DOI: 10.1016/j.abb.2025.110325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/19/2025] [Accepted: 01/31/2025] [Indexed: 02/04/2025]
Abstract
In the presented study, we evaluated changes in the molecular structures of lipids and proteins in organs/tissues at the early stage of obesity induced by a high-calorie diet (HCD), using animal models. We examined several different molecular parameters and the organs most affected by obesity. Fourier transform infrared (FTIR) spectroscopy combined with Principal Component Analysis (PCA) and Receiver Operating Characteristic (ROC) analysis were used to evaluate molecular changes in tissues taken from HCD-induced obese Wistar rats and their lean counterparts. We observed that at the early stage of obesity, changes occurred mainly in lipid structures, primarily affecting white epididymal adipose tissue (WAT) and the liver (Lr). No changes in protein molecular structures were observed in any of the examined organs. PCA showed distinctly different organ/tissue compositions, in terms of molecular parameters, for both groups. In turn, ROC analysis indicated that fatty acid chain length (FACL), lipid unsaturation (L_Unsat), and carbonyl/lipid ratio (Carb/L) for WAT, and FACL and lipid/protein ratio (L/P) for Lr, were the molecular parameters, whose levels differentiated the most between both groups. We demonstrated that studies using FTIR spectroscopy combined with advanced data mining methods could deepen the current knowledge about obesity and the biochemical changes occurring in the organs affected by this disease. Thus, they can help in the future with better and faster diagnosis and prevention of obesity and its complications.
Collapse
Affiliation(s)
- Kaja Piana
- AGH University of Krakow, Faculty of Physics and Applied Computer Science, Al. A. Mickiewicza 30, 30-059, Krakow, Poland
| | - Agata Ziomber-Lisiak
- Chair of Pathophysiology, Faculty of Medicine, Jagiellonian University Medical College, ul. Czysta 18, 31-121, Krakow, Poland
| | - Blazej Ruszczycki
- AGH University of Krakow, Faculty of Physics and Applied Computer Science, Al. A. Mickiewicza 30, 30-059, Krakow, Poland
| | - Andrzej Bugajski
- Chair of Pathophysiology, Faculty of Medicine, Jagiellonian University Medical College, ul. Czysta 18, 31-121, Krakow, Poland
| | | |
Collapse
|
280
|
Gao Z, Yu Y, Eckel‐Mahan K, Kolonin MG. Caloric Restriction and Telomere Preservation in TERT Knockout Adipocyte Progenitors Does Not Rescue Mice From Metabolic Dysfunction due to a TERT Function in Adipocyte Mitochondria. Aging Cell 2025; 24:e14499. [PMID: 39932851 PMCID: PMC11896407 DOI: 10.1111/acel.14499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/20/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
Inactivation of telomerase (TERT) in adipocyte progenitor cells (APC) expedites telomere attrition, and the onset of diabetes in mice fed high-fat diet (HFD), which promotes APC over-proliferation and replicative senescence. Here, we show that time-restricted feeding or caloric restriction in the postnatal development of mice subsequently subjected to HFD prevents telomere attrition but not glucose intolerance. This metabolic effect of dietary intervention was not observed for mice with TERT KO in endothelial or myeloid cells. To characterize the telomere-independent effects of TERT in the APC lineage, we analyzed mice with TERT knockout in mature adipocytes (AD-TERT-KO), which do not proliferate and avoid telomere attrition. Analysis of adipocytes from AD-TERT-KO mice indicated reliance on glycolysis and decreased mitochondrial oxidative metabolism. We show that AD-TERT-KO mice have reduced cold tolerance and metabolism abnormality indicating a defect in adaptive thermogenesis, characteristic of aging. Conversely, ectopic TERT expression in brown adipocytes-induced mitochondrial oxidation and thermogenic gene expression. We conclude that TERT plays an important non-canonical function in the mitochondria of adipocytes.
Collapse
Affiliation(s)
- Zhanguo Gao
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Kristin Eckel‐Mahan
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| |
Collapse
|
281
|
Cuesta‐Gomez N, Castro C, Rosko M, Seeberger K, Korbutt GS. Sex Differences in Maturation and Function of Neonatal Porcine Islets Upon Transplantation in Mice. Xenotransplantation 2025; 32:e70039. [PMID: 40243327 PMCID: PMC12005065 DOI: 10.1111/xen.70039] [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: 01/13/2025] [Revised: 03/08/2025] [Accepted: 03/14/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Neonatal porcine islets (NPIs) can mature into a mixed population of endocrine cells that can restore glucose control in mice, pigs, and non-human primates, representing a potential alternative islet source for clinical beta cell replacement therapy. However, it remains unclear how conditions in the recipient influence the maturation and function of these cells. Here, we investigated the impact of host sex on NPIs implanted under the kidney capsule of male and female B6.129S7-Rag1tm1Mom (B6/Rag-/-) mice. METHODS Diabetic mice were transplanted with 3000 NPIs under the kidney capsule. All mice were monitored for reversal of hyperglycemia and glucose clearance at 8- and 20-weeks post-transplant. Grafts were assessed for cell composition and insulin content. RESULTS Female mice demonstrated improved glucose clearance at 8- and 20-weeks post-transplant compared to their male counterparts. Improved glucose clearance correlated with accelerated diabetes reversal in females (8 weeks vs. 12 weeks in males) and increased rates of euglycemic achievement (17/18 in females vs. 14/19 in males). However, grafts collected from male mice exhibited an increased percentage of insulin-positive cells as well as increased insulin content. CONCLUSION The sex of the host influences the outcomes of NPI transplantation, showcasing the relevance of understanding the role of sex as a biological variable in islet transplantation.
Collapse
Affiliation(s)
- Nerea Cuesta‐Gomez
- Department of SurgeryUniversity of AlbertaEdmontonAlbertaCanada
- Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Chelsea Castro
- Department of SurgeryUniversity of AlbertaEdmontonAlbertaCanada
- Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Mandy Rosko
- Department of SurgeryUniversity of AlbertaEdmontonAlbertaCanada
- Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Karen Seeberger
- Department of SurgeryUniversity of AlbertaEdmontonAlbertaCanada
- Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Gregory S. Korbutt
- Department of SurgeryUniversity of AlbertaEdmontonAlbertaCanada
- Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
282
|
Cinti F, Mezza T, Severi I, Moffa S, Giuseppe GD, Capece U, Ciccarelli G, Soldovieri L, Brunetti M, Morciano C, Gugliandolo S, Senzacqua M, Avolio A, Quero G, Tondolo V, Nista EC, Moroni R, Cinti S, Alfieri S, Gasbarrini A, Pontecorvi A, Giaccari A. In humans increase in intrapancreatic adipose tissue predicts beta-cell dedifferentiation score before diabetes onset: A pilot study. Diabetes Res Clin Pract 2025; 221:112029. [PMID: 39938572 DOI: 10.1016/j.diabres.2025.112029] [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: 11/14/2024] [Revised: 01/18/2025] [Accepted: 01/29/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND The role of intrapancreatic fat (WAT) in the development of T2D remains debated. In T2D, β-cell dedifferentiation is one of the mechanisms responsible for β-cell failure but its role in prediabetes is unknown. We aimed to investigate the relation between WAT and β-cell dedifferentiation prior to diabetes onset. METHODS We evaluated pancreatic samples from patients without history of diabetes, who had previously undergone an oral glucose tolerance test and hyperglycemic clamp. Subjects were divided into 3 glucose tolerance groups: normal (NGT), altered (IGT) or newly diagnosed diabetes (nDM). Dedifferentiation and WAT% were morphologically assessed. RESULTS WAT was higher in nDM patients compared to NGT and IGT (WAT nDM 43.79 ± 20.83 %, IGT 10.67 ± 8.5 %, NGT 4.43 ± 4.37 %). We observed a progressive increase in dedifferentiation score, in parallel with worsening glucose tolerance (from NGT to IGT to nDM; 4.8 ± 3.8; 32.37 ± 7.4; 40.38 ± 19 respectively). A strong linear regression established that WAT could statistically significantly predict dedifferentiated β-cells (R = 0.86, p = 0.005), and that the predicted increase in dedifferentiated β-cells was 1.25 points for every extra one-point change in WAT. Interestingly, the WAT and dedifferentiation score variable pair were significantly related to 1-hour post-load glycemia. CONCLUSIONS The accumulation of WAT might be responsible for dedifferentiation, making it a potential new target to curb diabetes onset.
Collapse
Affiliation(s)
- Francesca Cinti
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Teresa Mezza
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy; Pancreas Unit CEMAD Centro Malattie dell'Apparato Digerente Medicina Interna e Gastroenterologia Fondazione Policlinico Universitario Gemelli IRCCS Università cattolica del Sacro Cuore Rome Italy
| | - Ilenia Severi
- Pancreas Unit CEMAD Centro Malattie dell'Apparato Digerente Medicina Interna e Gastroenterologia Fondazione Policlinico Universitario Gemelli IRCCS Università cattolica del Sacro Cuore Rome Italy
| | - Simona Moffa
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Gianfranco Di Giuseppe
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Umberto Capece
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Gea Ciccarelli
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Laura Soldovieri
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Michela Brunetti
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Cassandra Morciano
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy; Dipartimento di Scienze Cliniche e Sperimentali, Medicina Interna - Università degli Studi di Brescia Brescia BS Italy
| | - Shawn Gugliandolo
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Martina Senzacqua
- Department of Clinical and Experimental Medicine Center of Obesity Università Politecnica delle Marche Ancona Italy
| | - Adriana Avolio
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Giuseppe Quero
- Chirurgia Digestiva, Fondazione Policlinico Universitario Agostino Gemelli IRCCS Roma Italy
| | - Vincenzo Tondolo
- Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Enrico Celestino Nista
- Pancreas Unit CEMAD Centro Malattie dell'Apparato Digerente Medicina Interna e Gastroenterologia Fondazione Policlinico Universitario Gemelli IRCCS Università cattolica del Sacro Cuore Rome Italy
| | | | - Saverio Cinti
- Department of Clinical and Experimental Medicine Center of Obesity Università Politecnica delle Marche Ancona Italy
| | - Sergio Alfieri
- Chirurgia Digestiva, Fondazione Policlinico Universitario Agostino Gemelli IRCCS Roma Italy
| | - Antonio Gasbarrini
- Pancreas Unit CEMAD Centro Malattie dell'Apparato Digerente Medicina Interna e Gastroenterologia Fondazione Policlinico Universitario Gemelli IRCCS Università cattolica del Sacro Cuore Rome Italy
| | - Alfredo Pontecorvi
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy
| | - Andrea Giaccari
- Endocrinologia e Diabetologia Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy; Dipartimento di Medicina e Chirurgia Traslazionale Università Cattolica del Sacro Cuore Rome Italy.
| |
Collapse
|
283
|
Algin O, Güneş YC, Cankurtaran RE, Corabay S, Yurekli OT. The Relationship Between Intramural Fat Accumulation and Sarcopenia on MR Enterography Exams in Patients with Crohn's Disease. ROFO-FORTSCHR RONTG 2025; 197:298-307. [PMID: 38977011 DOI: 10.1055/a-2330-8148] [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: 07/10/2024]
Abstract
Research on magnetic resonance enterography (MRE) and sarcopenia for assessing Crohn's disease (CD) is growing. Our study examined the connections between the presence of sarcopenia, intramural fat accumulation (IFA), and clinical, laboratory, and MRE findings.This retrospective study was conducted on 112 patients with suspected or diagnosed CD who underwent 3-tesla MRE. The study examined the correlation between sarcopenia-related parameters and MRE findings. Results of MRE exams and clinical and laboratory results were statistically analyzed. The Kruskal-Wallis, Pearson chi-square, and Fisher-Freeman-Halton tests were used for comparison.It was determined that patients with active inflammation on a chronic basis had more IFA than the others (p<0.001). There were positive relationships between IFA and intramural edema (p<0.001). There were positive correlations between IFA and high b-values and negative correlations with apparent diffusion coefficient values (p<0.05). Positively significant relationships were found between IFA and wall thickness, affected segment length, disease duration, and sedimentation values (p<0.05). Strong correlations were found between sarcopenia and the CD activity index as well as wall thickness (p<0.001/p=0.003). There was no significant relationship between steroid usage and other variables.The presence of IFA is associated with chronic inflammation. There was no clear relationship between steroid use and IFA. Our findings support the idea that sarcopenia is related to the activity of CD. Further comprehensive research is required on these subjects. · The usage of MR enterography for the management of CD is increasing day by day due to its advantages.. · There is a paucity of evidence regarding the relationship between sarcopenia and MR enterography findings in patients with CD.. · Intramural fat accumulation (IFA) is a sign of chronicity in patients with CD.. · The presence of IFA seems to be associated with active inflammation on a chronic basis.. · There was no clear relationship between steroid use and IFA.. · Algin O, Güneş YC, Cankurtaran RE et al. The Relationship Between Intramural Fat Accumulation and Sarcopenia on MR Enterography Exams in Patients with Crohn's Disease. Rofo 2025; DOI 10.1055/a-2330-8148.
Collapse
Affiliation(s)
- Oktay Algin
- Radiology Department, Ankara University, Ankara, Türkiye
| | - Yasin Celal Güneş
- Radiology Department, Kırıkkale Yuksek Ihtısas Hospital, Kırıkkale, Türkiye
| | - Rasim Eren Cankurtaran
- Gastroenterology Department, Ministry of Health Ankara Etlik City Hospital, Ankara, Türkiye
| | | | - Oyku Tayfur Yurekli
- Gastroenterology Department, Yildirim Beyazit University Faculty of Medicine, Ankara, Türkiye
| |
Collapse
|
284
|
Ozer F, Baygutalp F, Celik M, Kılıc Baygutalp N. Evaluation of adipokines, oxidative stress, and inflammatory markers in gout patients. Arch Rheumatol 2025; 40:112-122. [PMID: 40264479 PMCID: PMC12010260 DOI: 10.46497/archrheumatol.2025.11001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/31/2025] [Indexed: 04/24/2025] Open
Abstract
Objectives This study aims to investigate the role of adipokines, oxidative stress, and inflammatory markers in the pathogenesis of gout. Patients and methods Between September 2021 and December 2022, a total of 88 volunteers including 44 patients (37 males, 7 females; mean age: 56.5±11.6 years; range, 30 to 77 years) with gout disease and 44 age- and sex-matched healthy controls (37 males, 7 females; mean age: 54.3±12.1 years; range, 30 to 78 years) were retrospectively analyzed. Serum interleukin-10 (IL-10), tumor necrosis factor-alpha (TNF-α), irisin, resistin, total oxidant status (TOS), and total antioxidant status (TAS) levels were analyzed. Results The mean serum irisin levels were significantly higher in patients with gout than in the healthy controls (405.6±97.3 and 316.0±80.8 pg/mL, respectively). The mean IL-10 levels were significantly lower in patients with gout than in healthy controls (90.8±71.9 and 172.7±128.6 pg/mL, respectively). There was no statistically significant difference in the serum resistin, TNF-α, TOS, or TAS levels between the groups (p>0.05). Conclusion Our study results show that serum irisin and IL-10 seem to be candidate biomarkers of diagnosis of gout.
Collapse
Affiliation(s)
- Fatma Ozer
- Department of Physical Medicine and Rehabilitation, Atatürk University Faculty of Medicine, Erzurum, Türkiye
| | - Fatih Baygutalp
- Department of Physical Medicine and Rehabilitation, Atatürk University Faculty of Medicine, Erzurum, Türkiye
| | - Muhammet Celik
- Department of Medical Biochemistry, Atatürk University Faculty of Medicine, Erzurum, Türkiye
| | | |
Collapse
|
285
|
Maniglio M, Loisay L, de Haro D, Antoniadis A, Hügle T, Geurts J. Subchondral bone marrow adipose tissue lipolysis regulates bone formation in hand osteoarthritis. Osteoarthritis Cartilage 2025; 33:322-329. [PMID: 39725154 DOI: 10.1016/j.joca.2024.12.005] [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: 09/27/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Bone marrow adipose tissue (BMAT) is emerging as an important regulator of bone formation and energy metabolism. Lipolysis of BMAT releases glycerol and fatty acid substrates that are catabolized by osteoblasts. Here, we investigated whether BMAT lipolysis is involved in subchondral bone formation in hand osteoarthritis (OA). METHODS Subchondral BMAT lipolysis and bone marrow adipocyte (BMAd) morphology were studied in clinical specimens of carpometacarpal (CMC-1) and distal interphalangeal joint OA. BMAd size, osteoblast numbers and expression of lipolysis enzymes (ATGL, phospho-HSL, MGLL) were compared between regions of low and high bone formation. Free fatty acids, glycerol and bone biomarkers were measured in osteochondral explants. RESULTS Subchondral BMAd size was positively correlated with BMI (r = 0.60, [0.082,0.87]) and reduced in regions of high bone formation (-1149 µm2, [-1977,-726.2]). Osteoblast numbers were negatively correlated with BMAd size (r = -0.48, [-0.73,-0.12]). All lipolysis enzymes were expressed in both in BMAds and activated osteoblasts and the area percentages of ATGL (+2.26% [0.19,3.47]), phospho-HSL (+1.57% [0.31,6.48]) and MGLL (+4.04% [1.09,5.69]) were increased in regions of high bone formation. Secreted glycerol levels, but not free fatty acids, were correlated with bone formation markers pro-collagen type I (rho = 0.90) and alkaline phosphatase (rho = 0.78). CONCLUSION Our findings reveal a previously unrecognized role of BMAT lipolysis in regulating bone formation in hand OA, which may be modulated by BMI.
Collapse
Affiliation(s)
- Mauro Maniglio
- Plastic and Hand Surgery, Department of Musculoskeletal Medicine, University Hospital Lausanne and University of Lausanne (CHUV-UNIL), Lausanne, Switzerland; Department of Hand Surgery, Balgrist University Clinic, Zürich, Switzerland
| | - Léa Loisay
- Rheumatology, Department of Musculoskeletal Medicine, University Hospital Lausanne and University of Lausanne (CHUV-UNIL), Lausanne, Switzerland
| | - Diego de Haro
- Rheumatology, Department of Musculoskeletal Medicine, University Hospital Lausanne and University of Lausanne (CHUV-UNIL), Lausanne, Switzerland
| | - Alexander Antoniadis
- Orthopaedics, Department of Musculoskeletal Medicine, University Hospital Lausanne and University of Lausanne (CHUV-UNIL), Lausanne, Switzerland
| | - Thomas Hügle
- Rheumatology, Department of Musculoskeletal Medicine, University Hospital Lausanne and University of Lausanne (CHUV-UNIL), Lausanne, Switzerland
| | - Jeroen Geurts
- Rheumatology, Department of Musculoskeletal Medicine, University Hospital Lausanne and University of Lausanne (CHUV-UNIL), Lausanne, Switzerland.
| |
Collapse
|
286
|
Plata VTG, de Jesus Simão J, de Sousa Bispo AF, Alonso-Vale MI, Armelin-Correa L. Impact of fish oil on epigenetic regulation in perirenal adipose tissue of obese mice. Obes Res Clin Pract 2025; 19:122-129. [PMID: 40246605 DOI: 10.1016/j.orcp.2025.03.006] [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: 10/21/2024] [Revised: 03/10/2025] [Accepted: 03/27/2025] [Indexed: 04/19/2025]
Abstract
It has been demonstrated that fish oil (FO), a source of omega-3 polyunsaturated fatty acids (n-3 PUFA), offers partial protection to mice from the adverse effects of a high-fat diet (HFD) by altering the expression of genes involved in adipogenesis and adipocyte metabolism. Histone 3 lysine 27 (H3K27) modifiers, namely Ezh2, Kdm6a, Kdm6b, Crebbp and Ep300, are vital for the appropriate differentiation and metabolism of adipocytes, as they can either silence or activate transcription. The expansion of perirenal adipose tissue (AT) in obesity is associated with a number of complications, including hypertension and kidney disease. The aim of this study was to assess the expression of H3K27 modifiers and genes involved in adipogenesis and adipocyte metabolism in perirenal AT of HFD-fed and FO-treated (5DHA:1EPA) mice using real-time PCR. This study demonstrates, for the first time, that a high-fat diet (HFD) increases the expression of Kdm6b (H3K27 demethylase) in perirenal AT, and that treatment with FO can completely reverse this effect. Conversely, the expression of the Acly gene, which encodes an enzyme that provides a substrate for histone acetylases, was found to be reduced in HFD-fed mice and this was not reversed by FO treatment. Additionally, transcription factor genes, such as Tbx1, exhibited diminished expression in perirenal AT of mice fed an HFD. These observations suggest that a HFD affects the expression of chromatin modifiers, transcription factors, and metabolic genes in perirenal AT, and that FO can reverse some of these effects, offering a promising avenue for the treatment of obesity.
Collapse
Affiliation(s)
- Victor Tadeu Gonçalves Plata
- Post-graduation Program in Chemical Biology Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil
| | - Jussara de Jesus Simão
- Post-graduation Program in Chemical Biology Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil
| | - Andressa França de Sousa Bispo
- Post-graduation Program in Chemical Biology Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil
| | - Maria Isabel Alonso-Vale
- Post-graduation Program in Chemical Biology Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil; Department of Biological Sciences, Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil
| | - Lucia Armelin-Correa
- Post-graduation Program in Chemical Biology Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil; Department of Biological Sciences, Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil.
| |
Collapse
|
287
|
Zhang Y, Chen Y, Zhuang C, Qi J, Zhao RC, Wang J. Lipid droplets in the nervous system: involvement in cell metabolic homeostasis. Neural Regen Res 2025; 20:740-750. [PMID: 38886939 PMCID: PMC11433920 DOI: 10.4103/nrr.nrr-d-23-01401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/05/2023] [Accepted: 01/20/2024] [Indexed: 06/20/2024] Open
Abstract
Lipid droplets serve as primary storage organelles for neutral lipids in neurons, glial cells, and other cells in the nervous system. Lipid droplet formation begins with the synthesis of neutral lipids in the endoplasmic reticulum. Previously, lipid droplets were recognized for their role in maintaining lipid metabolism and energy homeostasis; however, recent research has shown that lipid droplets are highly adaptive organelles with diverse functions in the nervous system. In addition to their role in regulating cell metabolism, lipid droplets play a protective role in various cellular stress responses. Furthermore, lipid droplets exhibit specific functions in neurons and glial cells. Dysregulation of lipid droplet formation leads to cellular dysfunction, metabolic abnormalities, and nervous system diseases. This review aims to provide an overview of the role of lipid droplets in the nervous system, covering topics such as biogenesis, cellular specificity, and functions. Additionally, it will explore the association between lipid droplets and neurodegenerative disorders. Understanding the involvement of lipid droplets in cell metabolic homeostasis related to the nervous system is crucial to determine the underlying causes and in exploring potential therapeutic approaches for these diseases.
Collapse
Affiliation(s)
- Yuchen Zhang
- School of Life Sciences, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Yiqing Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Cheng Zhuang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Jingxuan Qi
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Robert Chunhua Zhao
- School of Life Sciences, Shanghai University, Shanghai, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy, Beijing, China
| | - Jiao Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
288
|
Lin S, Wang CJ, Yang PK, Li B, Wu Y, Yu KW. Enriched environment improves memory function by promoting synaptic remodeling in vascular dementia rats. Brain Res Bull 2025; 222:111262. [PMID: 39978738 DOI: 10.1016/j.brainresbull.2025.111262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/12/2025] [Accepted: 02/15/2025] [Indexed: 02/22/2025]
Abstract
Vascular dementia (VaD), attributed to cerebrovascular pathology, is a leading cause of cognitive decline, characterized by memory loss, bradyphrenia, and affective lability, with memory deficits being particularly pronounced. The potential of enriched environment (EE) to ameliorate cognitive impairments by enhancing hippocampal synaptic plasticity, neurogenesis, and white matter remodeling has garnered considerable interest. In this study, we used a rat model for VaD through the procedure of bilateral common carotid artery ligation (BCCAO). We randomly assigned male Sprague-Dawley (SD) rats to three groups: the control sham-operated group (Sham group), the surgery-induced dementia group (BCCAO group), and the surgery-induced dementia group with enriched environment (EE group). The Sham and BCCAO groups were kept under standard lab conditions, whereas the EE group was housed in an enriched setting. Employing a behavioral assay battery, we observed that EE intervention significantly improved the spatial learning and memory performance in the Morris water maze. Subsequent neuromorphological assessments utilizing transmission electron microscopy disclosed an increase in synaptic density and postsynaptic density (PSD) thickness within the hippocampal CA1 region, indicative of structural synaptic modulation. Further probing into the molecular underpinnings revealed that EE upregulated the expression of PSD95, corroborating its role in enhancing cognitive faculties. Additionally, our investigation into the PGC-1α/FNDC5/BDNF pathway demonstrated that EE intervention elevated the expression of these neurotrophic factors, suggesting a mechanistic link to synaptic and cognitive restoration. In summation, our findings elucidate the neurorestorative potential of EE in a preclinical VaD model, presenting a non-pharmacological intervention that modulates synaptic architecture and activates neuroprotective pathways. The observed correlations between synaptic remodeling and cognitive enhancement underscore the therapeutic relevance of EE in VaD, warranting further investigation for clinical applications.
Collapse
Affiliation(s)
- Shuang Lin
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuan-Jie Wang
- Department of Rehabilitation Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Peng-Kun Yang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Bing Li
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Yi Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| | - Ke-Wei Yu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
289
|
Sayharman S, Belviranlı M, Okudan N. The association between circulating irisin, osteocalcin and FGF21 levels with anthropometric characteristics and blood lipid profile in young obese male subjects. Adv Med Sci 2025; 70:117-123. [PMID: 39894443 DOI: 10.1016/j.advms.2025.01.010] [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: 07/23/2024] [Revised: 10/11/2024] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE Myokines secreted from skeletal muscle such as irisin, osteokines secreted from bone such as osteocalcin, and hepatokines secreted from the liver such as fibroblast growth factor 21 (FGF21) play a role in the regulation of metabolic homeostasis. However, the changes that occur in obesity and the interaction between them have not been fully explained. Therefore, this study aimed to compare irisin, osteocalcin and FGF21 levels in young obese males against individuals with normal body weight and to reveal the possible relationship between them and with anthropometric measurements and blood lipid profile. MATERIALS AND METHODS This single-center study included 28 Turkish young males aged 20-29 years: 14 obese participants with a body mass index (BMI) between 30.0 and 34.9 and 14 healthy controls with a BMI between 18.5 and 24.9. Anthropometric, and body composition parameters, blood lipid profile, and irisin, osteocalcin and FGF21 levels of groups were measured. Correlation analyses were performed between irisin, osteocalcin, and FGF21 and other measured parameters. RESULTS Circulating irisin, osteocalcin and FGF21 levels were significantly higher in the obese group than in the control group (p < 0.05). Correlation analysis showed that irisin was positively correlated with total cholesterol, triglycerides and low density lipoprotein-cholesterol (LDL-C) and FGF21 was positively correlated with total cholesterol and LDL-C (p < 0.05). Positive correlation between irisin and osteocalcin, FGF21 and osteocalcin and FGF21 and irisin was observed (p < 0.05). CONCLUSIONS Irisin, osteocalcin, and FGF21 have a potential role in the pathophysiology of obesity and related metabolic diseases due to their interactions.
Collapse
Affiliation(s)
- Sema Sayharman
- Selçuk University, School of Medicine, Department of Physiology, Konya, Turkey
| | - Muaz Belviranlı
- Selçuk University, School of Medicine, Department of Physiology, Konya, Turkey.
| | - Nilsel Okudan
- Selçuk University, School of Medicine, Department of Physiology, Konya, Turkey
| |
Collapse
|
290
|
Pahlavani M, Pham K, Kalupahana NS, Morovati A, Ramalingam L, Abidi H, Kiridana V, Moustaid-Moussa N. Thermogenic adipose tissues: Promising therapeutic targets for metabolic diseases. J Nutr Biochem 2025; 137:109832. [PMID: 39653156 DOI: 10.1016/j.jnutbio.2024.109832] [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: 05/02/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025]
Abstract
The ongoing increase in the prevalence of obesity and its comorbidities such as cardiovascular disease, type 2 diabetes (T2D) and dyslipidemia warrants discovery of novel therapeutic options for these metabolic diseases. Obesity is characterized by white adipose tissue expansion due to chronic positive energy balance as a result of excessive energy intake and/or reduced energy expenditure. Despite various efforts to prevent or reduce obesity including lifestyle and behavioral interventions, surgical weight reduction approaches and pharmacological methods, there has been limited success in significantly reducing obesity prevalence. Recent research has shown that thermogenic adipocyte (brown and beige) activation or formation, respectively, could potentially act as a therapeutic strategy to ameliorate obesity and its related disorders. This can be achieved through the ability of these thermogenic cells to enhance energy expenditure and regulate circulating levels of glucose and lipids. Thus, unraveling the molecular mechanisms behind the formation and activation of brown and beige adipocytes holds the potential for probable therapeutic paths to combat obesity. In this review, we provide a comprehensive update on the development and regulation of different adipose tissue types. We also emphasize recent interventions in harnessing therapeutic potential of thermogenic adipocytes by bioactive compounds and new pharmacological anti-obesity agents.
Collapse
Affiliation(s)
- Mandana Pahlavani
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Department of Nutrition and Food Sciences, Texas Woman's University, Dallas, Texas, USA
| | - Kenneth Pham
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Ashti Morovati
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Department of Nutrition and Food Studies, Syracuse University, Syracuse, New York, USA
| | - Hussain Abidi
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Vasana Kiridana
- Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Institute for One Health Innovation, Texas Tech University and Texas Tech Health Sciences Center, Lubbock, Texas, USA.
| |
Collapse
|
291
|
Hanada S, Ishikawa K, Shirai T, Takemasa T, Nakada K. Endurance swimming exacerbates mitochondrial myopathy in mice with high mtDNA deletions. Mitochondrion 2025; 81:102010. [PMID: 39956167 DOI: 10.1016/j.mito.2025.102010] [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: 12/09/2024] [Revised: 01/25/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Recent studies have reported that endurance exercise enhances mitochondrial function, facilitating discussions of its potential as a therapeutic strategy for mitochondrial diseases caused by the accumulation of mutant mitochondrial DNA (mtDNA). In this study, we assessed the effects of endurance exercise on muscle pathology in a mitochondrial disease mouse model (mito-miceΔ) that is characterized by severe clinical phenotypes owing to the predominant accumulation of mtDNA with a large-scale deletion (ΔmtDNA). Contrary to expectations that endurance exercise may enhance mitochondrial function, endurance exercise exacerbated muscle pathology in mito-miceΔ. Therefore, exercise interventions should be potentially avoided in patients with severe mitochondrial diseases.
Collapse
Affiliation(s)
- Sho Hanada
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan
| | - Kaori Ishikawa
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan.
| | - Takanaga Shirai
- Institute of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan; Research Fellow of Japan Society for Promotion Science, Chiyoda-ku, Tokyo 102-0083, Japan; Department of Human Sciences, Kanagawa University, 3-27-1 Rokkakubashi, Kanagawa-ku, Yokohama-shi, Kanagawa 221-8686, Japan
| | - Tohru Takemasa
- Institute of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan
| | - Kazuto Nakada
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan.
| |
Collapse
|
292
|
Peng L, Li S, Huang Q, Sun Y, Sun J, Luo T, Wang Y, Hu Z, Lai W, Peng H. Irisin-mediated muscle-renal crosstalk as a protective mechanism against contrast-induced acute kidney injury via cGAS-STING signalling inhibition. Clin Transl Med 2025; 15:e70235. [PMID: 40008481 PMCID: PMC11862893 DOI: 10.1002/ctm2.70235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/17/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Contrast-induced acute kidney injury (CI-AKI) continues to pose a pressing clinical challenge during invasive cardiovascular procedures due to the limited availability of preventative strategies. We aimed to demonstrate that irisin, a myokine induced by exercise, protects against CI-AKI by inhibiting the cGAS-STING inflammatory pathway. METHODS AND RESULTS We explored the relationship between serum irisin levels and CI-AKI incidence in patients administered the contrast media iohexol. Notably, lower serum irisin levels were strongly associated with an increased incidence of CI-AKI following contrast media administration. To establish a causal link between serum irisin levels and CI-AKI, we utilised a mouse model that simulates exercise by overexpressing muscle-specific PGC-1α. This approach showed a significant reduction in tubular injury and mitochondrial dysfunction induced by iohexol via cGAS/STING suppression, thereby diminishing inflammation. Mechanistically, irisin was found to inhibit the activation of cGAS/STING, preventing double stranded DNA (dsDNA) leakage and reducing inflammation in tubular epithelial cells (TECs). Pharmacological inhibition of STING further corroborated these observations. Moreover, we identified integrin complex αV/β5 as the irisin receptor on TECs, which is essential for irisin-mediated suppression of cGAS-STING signalling and resolution of inflammation. CONCLUSIONS Our data position irisin as a crucial factor in muscle‒kidney crosstalk, inhibiting cGAS-STING signalling and preventing dsDNA leakage via integrin αV/β5 in TECs, thus mitigating tubular injury and inflammation. These data underscore the potential of irisin as both a predictive biomarker for CI-AKI and a promising candidate for preventative strategies against CI-AKI. HIGHLIGHTS Irisin mediated muscle-kidney crosstalk mitigated tubular injury and inflammation. Irisin inhibited the cGAS-STING signalling activation via integrin αV/β5 in tubular epithelial cells. Irisin was a predictive biomarker and a promising candidate for CI-AKI.
Collapse
Affiliation(s)
- Long Peng
- Division of Cardiovascular Medicine, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Suhua Li
- Division of Cardiovascular Medicine, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Qiang Huang
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yuxiang Sun
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Juan Sun
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Ting Luo
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yanlin Wang
- Division of Nephrology, Department of MedicineUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Zhaoyong Hu
- Division of Nephrology, Department of MedicineBaylor College of MedicineHoustonTexasUSA
| | - Weiyan Lai
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Hui Peng
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
293
|
Kielbowski K, Bratborska AW, Bakinowska E, Pawlik A. Sirtuins as therapeutic targets in diabetes. Expert Opin Ther Targets 2025; 29:117-135. [PMID: 40116767 DOI: 10.1080/14728222.2025.2482563] [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: 08/28/2024] [Revised: 03/01/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
INTRODUCTION Sirtuins (SIRTs) are NAD+-dependent deacetylases that mediate post-translational modifications of proteins. Seven members of the SIRT family have been identified in mammals. Importantly, SIRTs interact with numerous metabolic and inflammatory pathways. Thus, researchers have investigated their role in metabolic and inflammatory disorders. AREAS COVERED In this review, we comprehensively discuss the involvement of SIRTs in the processes of pancreatic β-cell dysfunction, glucose tolerance, insulin secretion, lipid metabolism, and adipocyte functions. In addition, we describe the current evidence regarding modulation of the expression and activity of SIRTs in diabetes, diabetic complications, and obesity. EXPERT OPINION The development of specific SIRT activators and inhibitors that exhibit high selectivity toward specific SIRT isoforms remains a major challenge. This involves the need to elucidate the physiological pathways involving SIRTs, as well as their important role in the development of metabolic disorders. Molecular modeling techniques will be helpful to develop new compounds that modulate the activity of SIRTs, which may contribute to the preparation of new drugs that selectively target specific SIRTs. SIRTs hold promise as potential targets in metabolic disease, but there is much to learn about specific modulators and the final answers will await clinical trials.
Collapse
Affiliation(s)
- Kajetan Kielbowski
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | | | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
294
|
Hurtado-Carneiro V, Juan-Arevalo Y, Flores CN, Herrero-De-Dios C, Perez-Garcia A, Contreras C, Lopez M, Alvarez E, Sanz C. Enhanced thermogenesis in PAS Kinase-deficient male mice. Biochem Pharmacol 2025; 233:116757. [PMID: 39824466 DOI: 10.1016/j.bcp.2025.116757] [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: 07/31/2024] [Revised: 12/15/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
PAS domain-containing serine/threonine-protein kinase (PASK) is a nutrient and energy sensor regulated by fasting/refeeding conditions in hypothalamic areas involved in controlling energy balance. In this sense, PASK plays a role in coordinating the activation/inactivation of AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) in response to fasting. PASK deficiency protects against the development of diet-induced obesity. This has prompted an investigation into the potential role of PASK on energy expenditure through thermogenesis in adipose tissue. Our results indicate that PASK-deficient male mice exhibited higher brown adipose tissue (BAT) thermogenic activity and heat production. The inhibition of PASK function induces the expression of Uncoupling Protein 1 (UCP1) and the adipogenic marker peroxisome proliferator-activated receptor gamma (PPARγ) in BAT. In addition, PASK deficiency promotes the expression of UCP1 and other browning markers such as PR/SET Domain 16 (PRDM16) in inguinal white adipose tissue (WAT). PASK-deficient mice record an enhanced thermogenic response, even under stimuli such as β-3adrenergic receptor agonist or cold. This evidence reveals PASK as a new mechanism modulating BAT thermogenesis.
Collapse
Affiliation(s)
- Veronica Hurtado-Carneiro
- Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Spain; Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Madrid, Spain.
| | - Yolanda Juan-Arevalo
- Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Spain; Department of Cell Biology, Faculty of Medicine, Complutense University of Madrid, Spain
| | - Cinthya N Flores
- Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Spain; Department of Cell Biology, Faculty of Medicine, Complutense University of Madrid, Spain
| | - Carmen Herrero-De-Dios
- Department of Cell Biology, Faculty of Medicine, Complutense University of Madrid, Spain
| | - Ana Perez-Garcia
- Department of Cell Biology, Faculty of Medicine, Complutense University of Madrid, Spain
| | - Cristina Contreras
- Department of Physiology, Faculty of Pharmacy, Complutense University of Madrid, Spain; NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel Lopez
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Elvira Alvarez
- Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Madrid, Spain
| | - Carmen Sanz
- Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Madrid, Spain; Department of Cell Biology, Faculty of Medicine, Complutense University of Madrid, Spain
| |
Collapse
|
295
|
Katz DH, Lindholm ME, Ashley EA. Charting the Molecular Terrain of Exercise: Energetics, Exerkines, and the Future of Multiomic Mapping. Physiology (Bethesda) 2025; 40:0. [PMID: 39136551 DOI: 10.1152/physiol.00024.2024] [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: 05/07/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 11/21/2024] Open
Abstract
Physical activity plays a fundamental role in human health and disease. Exercise has been shown to improve a wide variety of disease states, and the scientific community is committed to understanding the precise molecular mechanisms that underlie the exquisite benefits. This review provides an overview of molecular responses to acute exercise and chronic training, particularly energy mobilization and generation, structural adaptation, inflammation, and immune regulation. Furthermore, it offers a detailed discussion of known molecular signals and systemic regulators activated during various forms of exercise and their role in orchestrating health benefits. Critically, the increasing use of multiomic technologies is explored with an emphasis on how multiomic and multitissue studies contribute to a more profound understanding of exercise biology. These data inform anticipated future advancement in the field and highlight the prospect of integrating exercise with pharmacology for personalized disease prevention and treatment.
Collapse
Affiliation(s)
- Daniel H Katz
- Division of Cardiovascular MedicineStanford University School of Medicine, Stanford, California, United States
| | - Maléne E Lindholm
- Division of Cardiovascular MedicineStanford University School of Medicine, Stanford, California, United States
| | - Euan A Ashley
- Division of Cardiovascular MedicineStanford University School of Medicine, Stanford, California, United States
| |
Collapse
|
296
|
Wang J, Gao S, Fu S, Li Y, Su L, Li X, Wu G, Jiang J, Zhao Z, Yang C, Wang X, Cui K, Sun X, Qi X, Wang C, Sun H, Shao S, Tian Y, Gong T, Luo J, Zheng J, Cui S, Liao F, Liu F, Wang D, Wong CCL, Yi M, Wan Y. Irisin reprograms microglia through activation of STAT6 and prevents cognitive dysfunction after surgery in mice. Brain Behav Immun 2025; 125:68-91. [PMID: 39701329 DOI: 10.1016/j.bbi.2024.12.019] [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: 05/19/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) is common in the aged population and associated with poor clinical outcomes. Irisin, an endogenous molecule that mediates the beneficial effects of exercise, has shown neuroprotective potential in several models of neurological diseases. Here we show that preoperative serum level of irisin is reduced in dementia patients over the age of 70. Comprehensive proteomics analysis reveals that deletion of irisin affects the nervous and immune systems, and reduces the expression of complement proteins. Systemically administered irisin penetrates the blood-brain barrier in mice, targets the microglial integrin αVβ5 receptor, activates signal transducer and activator of transcription 6 (STAT6), induces microglia reprogramming to the M2 phenotype, and improves immune microenvironment in LPS-induced neuroinflammatory mice. Finally, prophylactic administration of irisin prevents POCD-like behavior, particularly early cognitive dysfunction. Our findings provide new insights into the direct regulation of the immune microenvironment by irisin, and reveal that recombinant irisin holds great promise as a novel therapy for preventing POCD and other neuroinflammatory disorders. SUMMARY: Our findings reveal molecular and cellular mechanisms of irisin on neuroinflammation, and show that prophylactic administration of irisin prevents POCD-like behavior, particularly early cognitive dysfunction.
Collapse
Affiliation(s)
- Jiaxin Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Shuaixin Gao
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China; Human Nutrition Program, Department of Human Sciences & James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Su Fu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Yawei Li
- Department of Anesthesiology, Peking University First Hospital, Beijing 10034, China
| | - Li Su
- Peking University Medical and Health Analysis Center, Peking University, Beijing 10034, China
| | - Xiaoman Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Guanghao Wu
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Jiankuo Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Zifang Zhao
- Department of Pain Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Chaojuan Yang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Xiaoyi Wang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Kun Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Beijing Life Science Academy, Beijing 102209, China
| | - Xiaoyan Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Xuetao Qi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Cheng Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Changping Laboratory, Beijing 102206, China
| | - Haojie Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Shan Shao
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Yue Tian
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Tingting Gong
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Jianyuan Luo
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing 100191, China
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Shuang Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Feifei Liao
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Fengyu Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China.
| | - Dongxin Wang
- Department of Anesthesiology, Peking University First Hospital, Beijing 10034, China.
| | - Catherine C L Wong
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China.
| | - Ming Yi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Medical Innovation Center (Taizhou) of Peking University, Taizhou 225316, China.
| | - You Wan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Medical Innovation Center (Taizhou) of Peking University, Taizhou 225316, China.
| |
Collapse
|
297
|
Grune E, Nattenmüller J, Kiefer LS, Machann J, Peters A, Bamberg F, Schlett CL, Rospleszcz S. Subphenotypes of body composition and their association with cardiometabolic risk - Magnetic resonance imaging in a population-based sample. Metabolism 2025; 164:156130. [PMID: 39743039 DOI: 10.1016/j.metabol.2024.156130] [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: 10/01/2024] [Revised: 12/05/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND For characterizing health states, fat distribution is more informative than overall body size. We used population-based whole-body magnetic resonance imaging (MRI) to identify distinct body composition subphenotypes and characterize associations with cardiovascular disease (CVD) risk. METHODS Bone marrow, visceral, subcutaneous, cardiac, renal, hepatic, skeletal muscle and pancreatic adipose tissue were measured by MRI in n = 299 individuals from the population-based KORA cohort. Body composition subphenotypes were identified by data-driven k-means clustering. CVD risk was calculated by established scores. RESULTS We identified five body composition subphenotypes, which differed substantially in CVD risk factor distribution and CVD risk. Compared to reference subphenotype I with favorable risk profile, two high-risk phenotypes, III&V, had a 3.8-fold increased CVD risk. High-risk subphenotype III had increased bone marrow and skeletal muscle fat (26.3 % vs 11.4 % in subphenotype I), indicating ageing effects, whereas subphenotype V showed overall high fat contents, and particularly elevated pancreatic fat (25.0 % vs 3.7 % in subphenotype I), indicating metabolic impairment. Subphenotype II had a 2.7-fold increased CVD risk, and an unfavorable fat distribution, probably smoking-related, while BMI was only slightly elevated. Subphenotype IV had a 2.8-fold increased CVD risk with comparably young individuals, who showed high blood pressure and hepatic fat (17.7 % vs 3.0 % in subphenotype I). CONCLUSIONS Whole-body MRI can identify distinct body composition subphenotypes associated with different degrees of cardiometabolic risk. Body composition profiling may enable a more comprehensive risk assessment than individual fat compartments, with potential benefits for individualized prevention.
Collapse
Affiliation(s)
- Elena Grune
- Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute of Epidemiology, Helmholtz Munich, Neuherberg, Germany; Pettenkofer School of Public Health, LMU Munich, Munich, Germany
| | - Johanna Nattenmüller
- Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute for Radiology and Nuclear Medicine Hirslanden Clinic St. Anna, Lucerne, Switzerland
| | - Lena S Kiefer
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, Eberhard Karls University of Tuebingen, Tuebingen, Germany; Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Jürgen Machann
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Medical Faculty, Ludwig-Maximilians-Universität (LMU), Munich, Germany; German Center for Cardiovascular Disease Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Fabian Bamberg
- Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christopher L Schlett
- Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Susanne Rospleszcz
- Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute of Epidemiology, Helmholtz Munich, Neuherberg, Germany.
| |
Collapse
|
298
|
Wu W, Huynh K, Du JC, She G, Duong T, Ziemann M, Zhao WB, Deng XL, Meikle PJ, Du XJ. Hippo pathway activation causes multiple lipid derangements in a murine model of cardiomyopathy. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159590. [PMID: 39709046 DOI: 10.1016/j.bbalip.2024.159590] [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: 07/19/2024] [Revised: 12/02/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Metabolic reprogramming occurs in cardiomyopathy and heart failure contributing to progression of the disease. Activation of cardiac Hippo pathway signaling has been implicated in mediating mitochondrial dysfunction and metabolic reprogramming in cardiomyopathy, albeit influence of Hippo pathway on lipid profile is unclear. Using a dual-omics approach, we determined alterations of cardiac lipids in a mouse model of cardiomyopathy due to enhanced Hippo signaling and explored molecular mechanisms. Lipidomic profiling discovered multiple alterations in lipid classes, notably reduction of triacylglycerol, diacylglycerol, phospholipids and ether lipids, and elevation of sphingolipids and lysophosphatidylcholine. Mechanistically, we found downregulated expression of PPARα and PGC-1α at mRNA and protein levels, and downregulated expression of PPARα-target genes, indicating attenuated transcriptional activity of PPARα/PGC-1α. Lipidomics-guided transcriptomic analysis revealed dysregulated expression of gene sets that were responsible for enhanced biosynthesis of ceramides, suppression of TG biosynthesis, storage, hydrolysis and mitochondrial fatty acid oxidation, and reduction of peroxisome-localized biosynthesis of ether lipids. Collectively, Hippo pathway activation with attenuated PPARα/PGC-1α signaling is the underlying mechanism for alterations in cardiac lipids in cardiomyopathy and failing heart.
Collapse
Affiliation(s)
- Wei Wu
- Department of Cardiology, Shaanxi Provincial Hospital, Xi'an, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Jin-Chan Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Gang She
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Thy Duong
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Mark Ziemann
- School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, Australia; Bioinformatics Working Group, Burnet Institute, Melbourne, Victoria, Australia
| | - Wei-Bo Zhao
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Endocrinology, The Ninth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| | - Xiao-Jun Du
- Department of Cardiology, Shaanxi Provincial Hospital, Xi'an, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China; Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| |
Collapse
|
299
|
Houttu V, Boulund U, Troelstra M, Csader S, Stols-Gonçalves D, Mak AL, Dijk AMV, Bouts J, Winkelmeijer M, Verdoes X, van den Berg-Faay S, Lek D, Ronteltap T, de Haan F, Jorstad H, Männistö V, Savonen K, Pentikäinen H, Hanhineva K, Babu AF, Panagiotou G, van Delden O, Verheij J, Doukas M, Nederveen A, Schwab U, Grefhorst A, Nieuwdorp M, Holleboom AG. Deep phenotyping of patients with MASLD upon high-intensity interval training. JHEP Rep 2025; 7:101289. [PMID: 40051412 PMCID: PMC11883402 DOI: 10.1016/j.jhepr.2024.101289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 03/09/2025] Open
Abstract
Background & Aims Exercise is a key component of lifestyle management in patients with metabolic dysfunction-associated steatotic liver disease (MASLD), but neither its therapeutic effect on the active stage of the disease, that is metabolic dysfunction-associated steatohepatitis (MASH) nor the mediating mechanisms have been characterized. Therefore, we performed multi-omic phenotyping of patients with MASLD-MASH on an exercise program. Methods Fifteen patients with MASLD conducted high-intensity interval training (HIIT) combined with home-based training for 12 weeks. MASLD was evaluated using histology, transient elastography, and multiparametric magnetic resonance imaging (MRI) before and after the intervention. Change in maximal oxygen consumption (VO2max) and MRI-determined liver fat were compared with a control group of patients with MASLD (n = 22). RNA sequencing was performed on liver, muscle, and fat biopsies of patients in the exercise group. Stool was analyzed by shotgun metagenomics and untargeted metabolomics was performed on plasma, urine, adipose, and stool. Results HIIT increased VO2max by 10.1% and improved mitochondrial metabolism in skeletal muscle, indicating improved cardiorespiratory fitness and adherence. VO2max increased significantly in the exercise group compared with controls. Histologically, no reduction in steatosis, MASH, or liver fibrosis was observed; however, transient elastography tended to improve. MRI-determined liver fat did not change in the exercise group compared with controls. HIIT induced changes in mRNA expression of genes related to beiging of adipose tissue and fibrogenesis in liver. In addition, specific gut microbial taxa and metabolites changed. Conclusions HIIT increased cardiorespiratory fitness and induced beneficial gene expression changes in muscle, adipose tissue, and liver, but without translation into histological improvement of MASLD. Longer exercise intervention trials are warranted to validate or refute current recommendations for exercise as a cornerstone treatment for MASLD-MASH. Impact and implications Despite exercise being considered as a key component of lifestyle management for steatotic liver disease, neither the clinical effects nor the mechanisms involved are completely understood. We show that a high-intensity interval training (HIIT) program in 15 patients with metabolic dysfunction-associated steatotic liver disease (MASLD) improved cardiorespiratory fitness, compared with 22 control patients with MASLD who did not participate in an exercise program, however, it did not improve MASLD. HIIT induced a positive effect on fat tissue and muscle metabolism which was accompanied with changes in certain gut bacteria and metabolites in blood and urine. These findings improve our understanding of the effects of exercise on the whole-body metabolism in relation to steatotic liver disease. As such, this study provides a basis for future exercise interventions in patients with MASLD, required to thoroughly test current guideline advice for exercise as a cornerstone treatment for MASLD of all stages. Clinical trial registry Dutch Trial Register (registration number NL7932).
Collapse
Affiliation(s)
- Veera Houttu
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ulrika Boulund
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marian Troelstra
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne Csader
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Daniela Stols-Gonçalves
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne Linde Mak
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne-Marieke van Dijk
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Julia Bouts
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Maaike Winkelmeijer
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Xanthe Verdoes
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sandra van den Berg-Faay
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Donne Lek
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Ted Ronteltap
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Ferdinand de Haan
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Harald Jorstad
- Department of Cardiology, Amsterdam Movement Sciences, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland, and Kuopio University Hospital, Kuopio, Finland
| | - Kai Savonen
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | | | - Kati Hanhineva
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Afekta Technologies Ltd., Kuopio, Finland
- Department of Life Technologies, Food Chemistry, and Food Development Unit, University of Turku, Turku, Finland
| | - Ambrin Farizah Babu
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Afekta Technologies Ltd., Kuopio, Finland
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research, and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
- Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
- Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Otto van Delden
- Department of Interventional Radiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Joanne Verheij
- Department of Pathology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Michial Doukas
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Aart Nederveen
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ursula Schwab
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Aldo Grefhorst
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Adriaan Georgius Holleboom
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
300
|
Antoniadou C, Gavriilidis E, Ritis K, Tsilingiris D. Anemia in diabetes mellitus: Pathogenetic aspects and the value of early erythropoietin therapy. Metabol Open 2025; 25:100344. [PMID: 39886103 PMCID: PMC11780985 DOI: 10.1016/j.metop.2024.100344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 02/01/2025] Open
Abstract
Anemia is a frequent, yet increasingly recognized, comorbidity in diabetes mellitus (DM), with prevalence often driven by multifactorial mechanisms. Hematinic deficiencies, common in this population, may arise from associated comorbidities or medications, such as metformin, as well as other drugs commonly employed for DM-related conditions. Among contributing factors, diabetic kidney disease (DKD) plays a pivotal role, with anemia developing more frequently and being more pronounced in earlier stages, than in CKD of other causes. This enhanced susceptibility stems primarily from the combined impact of impaired renal oxygen sensing and deficient erythropoietin (EPO) production linked to tubulointerstitial fibrosis. Additional mechanisms comprise glomerular dysfunction, shortened erythrocyte lifespan, uremia-induced bone marrow suppression, and increased bleeding risk. DM is also recognized as a chronic low-grade inflammatory condition, with its inflammatory burden driving iron maldistribution, suppression of erythropoiesis, and resistance to EPO. The diagnostic approach of anemia in DM mirrors that in the general population. Addressing modifiable causes such as hematinic deficiencies, and other chronic conditions, such as DKD and bone marrow disorders, is paramount. In total, the underlying pathophysiology of anemia in DM primarily reflects a state of absolute or relative EPO deficiency and/or diminished bone marrow responsiveness, effectively corresponding to 'anemia of chronic disease. Early initiation of EPO therapy, even in DM patients without overt DKD, may mitigate disease progression and improve outcomes. Future research should focus on diabetes-specific strategies integrating optimal EPO use, potentially implementing targeted management of renal and inflammatory contributors to anemia.
Collapse
Affiliation(s)
- Christina Antoniadou
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
- Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Efstratios Gavriilidis
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
- Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Konstantinos Ritis
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
- Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Dimitrios Tsilingiris
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
- Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|