1
|
Xiang L, Wang G, Zhuang Y, Luo L, Yan J, Zhang H, Li X, Xie C, He Q, Peng Y, Chen H, Li Q, Li X, Guo L, Lv G, Ding Y. Safety and efficacy of GLP-1/FGF21 dual agonist HEC88473 in MASLD and T2DM: A randomized, double-blind, placebo-controlled study. J Hepatol 2025; 82:967-978. [PMID: 39709140 DOI: 10.1016/j.jhep.2024.12.006] [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: 06/13/2024] [Revised: 11/14/2024] [Accepted: 12/01/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND & AIMS Glucagon-like peptide-1 (GLP-1) and fibroblast growth factor 21 (FGF21) are key regulators of glucose and lipid metabolism. In the present study, we assessed the safety and efficacy of a novel GLP-1/FGF21 dual agonist HEC88473 for the treatment of metabolic dysfunction-associated steatotic liver disease (MASLD) combined with type 2 diabetes mellitus (T2DM). METHODS This was a randomized, double-blind, placebo-controlled, multiple-ascending-dose phase Ib/IIa trial. Sixty patients with MASLD and T2DM were randomized (10:2) to receive HEC88473 (5.1, 15.3, 30.6, 45.9, or 68.0 mg) or placebo via weekly subcutaneous injection for 5 weeks. RESULTS After 5 weeks of treatment with HEC88473, MRI-proton density fat fraction (MRI-PDFF) was reduced in a dose-proportional manner. The largest relative mean change reached -47.21% (p = 0.0143) in the 30.6 mg cohort, compared with -15.05% in the placebo group, with a higher proportion of >30% relative reductions in patients with baseline PDFF >8%. The 5-week treatment with HEC88473 significantly reduced levels of HbA1c (glycated hemoglobin), as well as fasting and postprandial glucose levels. The largest mean change in HbA1c was -1.10% in the 68.0 mg cohort, compared with -0.31% in the placebo group. Improvement was also observed in participants' lipid profiles. Most adverse events were mild to moderate in severity. The most frequently reported adverse events were gastrointestinal disorders (n = 29, 48.3%). CONCLUSIONS Herein, we report the clinical safety and proof-of-concept data for the GLP-1/FGF21 dual agonist HEC88473. A 5-week treatment with HEC88473 was generally safe and well tolerated, with multiple positive effects observed, including reduced liver fat, and improved glycemic control, insulin resistance and lipid metabolism, together indicating comprehensive improvement in metabolic syndrome. IMPACT AND IMPLICATIONS In this randomized, double-blind, placebo-controlled phase Ib/IIa study, we assessed clinical safety, pharmacodynamic and pharmacokinetic data of the GLP-1/FGF21 dual agonist HEC88473 in patients with MASLD (metabolic dysfunction-associated steatotic liver disease) and T2DM (type 2 diabetes mellitus). HEC88473 was generally safe and well tolerated. The GLP-1/FGF21 dual agonist significantly reduced the hepatic fat fraction assessed using MRI-proton density fat fraction, and improved glycemic control and lipid profiles with only 5 weeks' treatment, leading to comprehensive improvement in metabolic syndrome. The present results suggest that HEC88473 could be a promising treatment option in this patient population. CLINICAL TRIAL NUMBER Chinese Drug Trial Identifier (http://www.chinadrugtrials.org.cn/index.html): CTR20211088. CLINICALTRIALS GOV: NCT05943886.
Collapse
Affiliation(s)
- Lin Xiang
- Phase I Clinical Trial Center, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Yulei Zhuang
- Dongguan HEC Biopharmaceutical R&D Co., Ltd, China
| | - Lin Luo
- Dongguan HEC Biopharmaceutical R&D Co., Ltd, China
| | - Jiangyu Yan
- Dongguan HEC Biopharmaceutical R&D Co., Ltd, China
| | - Hong Zhang
- Phase I Clinical Trial Center, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Xiaojiao Li
- Phase I Clinical Trial Center, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Can Xie
- Dongguan HEC Biopharmaceutical R&D Co., Ltd, China
| | - Qingwei He
- Dongguan HEC Biopharmaceutical R&D Co., Ltd, China
| | - Yuyu Peng
- Dongguan HEC Biopharmaceutical R&D Co., Ltd, China
| | - Hong Chen
- Phase I Clinical Trial Center, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Qianqian Li
- Phase I Clinical Trial Center, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Xiaoping Li
- Dongguan HEC Biopharmaceutical R&D Co., Ltd, China
| | - Linfeng Guo
- Dongguan HEC Biopharmaceutical R&D Co., Ltd, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China.
| | - Yanhua Ding
- Phase I Clinical Trial Center, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
2
|
Naidu D, Althaf Umar KP, Muhsina K, Augustine S, Jeengar MK, S K K. Zingiberaceae in Cardiovascular Health: A review of adipokine modulation and endothelial protection via adipocyte-endothelial crosstalk mechanism. Curr Nutr Rep 2025; 14:66. [PMID: 40366476 DOI: 10.1007/s13668-025-00656-x] [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] [Accepted: 04/28/2025] [Indexed: 05/15/2025]
Abstract
PURPOSE OF THE REVIEW Although adipose tissue controls metabolism and protects vital organs, its importance to general health is being highlighted by the rise in type 2 diabetes and cardiovascular disease. Adipokines produced by adipose cells are essential regulators of metabolism, glucose homeostasis, and inflammatory response. It also protects vascular endothelial cells for its potential implications for cardiovascular protection. Understanding its intricate involvement in adipose tissue-endothelial communication is critical in developing targeted therapeutics to treat cardiovascular conditions linked with obesity and metabolic dysregulation. Spices from the Zingiberaceae family, such as cardamom, turmeric, and ginger, have anti-inflammatory and anti-oxidant properties that help reduce oxidative stress, vascular dysfunction, and adipocyte-endothelial crosstalk which are all linked to the etiology of CVD. Comprehensive molecular insights into how they modulate adipokine signalling, inflammatory pathways, and ROS-induced adipocyte-vascular interactions remain unexplored, demanding additional translational and clinical validation. With an emphasis on patients with obesity and metabolic dysregulation, the investigation aims to elucidate the mechanisms by which the spice as whole/bioactive constituents of the Zingiberaceae family may provide protection against CVD by integrating previous studies. RECENT FINDINGS Current research continues to support the use of spices from the Zingiberaceae family, such as ginger, turmeric, cardamom, and pepper, as potential therapeutic agents for addressing metabolic complications like obesity, type II diabetes, and CVDs. These natural remedies may modulate adipocyte-endothelial crosstalk and inflammation by modulating important signalling pathways such as AMPK, AKT, PPAR, and NF-κB.. CONCLUSION This review provides a complete summary of existing knowledge, opening the way for future research and prospective therapeutic applications of Zingiberaceae spices in cardiovascular health management.
Collapse
Affiliation(s)
- Disha Naidu
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - K P Althaf Umar
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - K Muhsina
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Sanu Augustine
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Manish Kumar Jeengar
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India.
| | - Kanthlal S K
- Department of Pharmacology, Sree Krishna College of Pharmacy and Research Centre, Parassala, Thiruvananthapuram, Kerala, 695502, India.
| |
Collapse
|
3
|
Sun G, Zhao HQ, Huang YY, Guo ZY, Zhang L, Zhu H, Wang XY, Ye HN, Chen CP. Adiponectin receptor agonist adipoRon alleviates imiquimod-induced murine psoriasis. Int Immunopharmacol 2025; 154:114568. [PMID: 40184813 DOI: 10.1016/j.intimp.2025.114568] [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: 10/19/2024] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Psoriasis is a chronic inflammatory skin disease involving inflammation, immune responses and keratinocytes proliferation. It has been suggested that adiponectin/adiponectin receptor 1 (AdipoR1) signaling plays a role in regulating psoriatic skin inflammation. AdipoRon is a small molecule agonist of AdipoR1 and AdipoR2. The effect of adipoRon on psoriasis has not been elucidated. In this study, using a GEO database, we found that the expression of adiponectin was substantially decreased in skin lesions of psoriasis patients. This reduction was also validated in an imiquimod-induced psoriasis mouse model. Interestingly, we found that topical administration of adipoRon significantly ameliorated skin lesions induced by imiquimod. The critical pro-inflammatory cytokines (IL-6, IL-17A and IL-23) and the infiltration of macrophages, especially M1 macrophages were dramatically decreased while the infiltration of M2 macrophages were slightly increased in the skin lesions upon adipoRon treatment. Mechanistically, adipoRon inhibited macrophage inflammation and keratinocytes proliferation via activation of AMPK signaling pathway. Collectively, our study demonstrates that adipoRon displayed anti-inflammatory activity and anti-proliferation of keratinocytes, and attenuated psoriatic response. Activating AdipoR1 signaling pathway by adipoRon or others may represent a novel therapeutic approach to psoriasis.
Collapse
Affiliation(s)
- Geng Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hai-Qian Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan-Yuan Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhan-Ying Guo
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Lin Zhang
- Department of Medical Laboratory,The Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Hao Zhu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Xin-Yue Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hao-Nan Ye
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Cai-Ping Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
4
|
Ashraf A, Sarwar M, Arshad K, Talat MA, Saleh K. Evaluating miR-107 and adiponectin as biomarkers in obstructive sleep apnea: Associations with neurotransmitters and metabolic regulation. PLoS One 2025; 20:e0322128. [PMID: 40323993 PMCID: PMC12052186 DOI: 10.1371/journal.pone.0322128] [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: 11/14/2024] [Accepted: 03/17/2025] [Indexed: 05/07/2025] Open
Abstract
This study aimed to examine the levels of microRNA-107 (miR-107) and its correlation with neurotransmitters (glutamate, serotonin, melatonin) and adiponectin in patients with obstructive sleep apnea (OSA) compared to healthy controls. The results showed that serum levels of miR107, melatonin, and adiponectin were significantly lower in OSA patients compared to controls, while serotonin and glutamate levels were significantly higher. Spearman correlation analysis revealed that in the control group, miR-107 levels were moderately correlated with glutamate (negative) and adiponectin (positive), but these associations were disrupted in the OSA group. Receiver operating characteristic (ROC) curve analysis demonstrated that miR-107 had excellent diagnostic performance, with 100% sensitivity and 89.3% specificity at a cut-off of 3.0 ng/mL. Adiponectin also showed strong diagnostic potential, with 78% sensitivity and 89% specificity. In contrast, serotonin, melatonin, and glutamate exhibited more moderate diagnostic accuracy. These findings suggest that miR-107 and adiponectin could serve as promising biomarkers for diagnosing OSA, and targeting miR-107 to modulate metabolic factors may offer novel therapeutic approaches for improving OSA management.
Collapse
Affiliation(s)
- Asifa Ashraf
- The University of Lahore, Institute of Molecular Biology & Biotechnology, Lahore, Pakistan
- Biochemistry, Khawaja Muhammad Safdar Medical College, Sialkot, Pakistan
| | - Muhammad Sarwar
- The University of Lahore, Institute of Molecular Biology & Biotechnology, Lahore, Pakistan
| | - Kaleem Arshad
- Biochemistry, Khawaja Muhammad Safdar Medical College, Sialkot, Pakistan
| | - Muhammad Ali Talat
- Biochemistry, Khawaja Muhammad Safdar Medical College, Sialkot, Pakistan
| | - Khudija Saleh
- Biochemistry, Khawaja Muhammad Safdar Medical College, Sialkot, Pakistan
| |
Collapse
|
5
|
Martins FF, Amarante MDSM, Oliveira DS, Vasques‐Monteiro IML, Souza‐Mello V, Daleprane JB, Camillo CDS. Obesity, White Adipose Tissue, and Adipokines Signaling in Male Reproduction. Mol Nutr Food Res 2025; 69:e70054. [PMID: 40195898 PMCID: PMC12087738 DOI: 10.1002/mnfr.70054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/19/2025] [Accepted: 03/17/2025] [Indexed: 04/09/2025]
Abstract
Currently, obesity is a global pandemic characterized by systemic metabolic complications that negatively impact several organs, including white adipose tissue (WAT) and the tissues of the male reproductive system. Since the discovery of leptin in 1994, WAT has been recognized as a dynamic endocrine organ for secreting a series of molecules with hormonal functions, collectively called adipokines. The link between obesity, WAT, adipokines, and the male reproductive system is direct and little explored. With changes in nutritional status, WAT undergoes morphofunctional changes, and the secretion of adipokines is altered, negatively impacting reproductive mechanisms, including steroidogenesis and spermatogenesis. In this review, we address in an updated way the structural and functional characteristics of WAT as well as the link between obesity and changes in the signaling pathways of the adipokines leptin, adiponectin, resistin, visfatin, apelin, chemerin, omentin-1, vaspin, and asprosin in male reproduction. Understanding the relationship between obesity, these adipokines, and reproductive dysfunction can contribute to new strategies for the treatment of subfertility and male infertility.
Collapse
Affiliation(s)
| | | | - Daiana Santana Oliveira
- Laboratory of MorphometryMetabolism and Cardiovascular DiseasesBiomedical CenterInstitute of BiologyRio de Janeiro State UniversityRio de JaneiroBrazil
| | - Isabela Macedo Lopes Vasques‐Monteiro
- Department of Basic and Experimental NutritionLaboratory for Studies of Interactions Between Nutrition and GeneticsLEINGRio de Janeiro State UniversityRio de JaneiroBrazil
| | - Vanessa Souza‐Mello
- Laboratory of MorphometryMetabolism and Cardiovascular DiseasesBiomedical CenterInstitute of BiologyRio de Janeiro State UniversityRio de JaneiroBrazil
| | - Julio Beltrame Daleprane
- Department of Basic and Experimental NutritionLaboratory for Studies of Interactions Between Nutrition and GeneticsLEINGRio de Janeiro State UniversityRio de JaneiroBrazil
| | | |
Collapse
|
6
|
Lindfors S, Schmotz C, Lewandowski D, Hau A, Saikko L, Lehtonen E, Majaniemi V, Karhe M, Naams JB, Nisen H, Tienari J, Saleem MA, Pfeil K, Bugger H, Pietiläinen KH, Mirtti T, Palczewski K, Lehtonen S. Integrin Trafficking, Fibronectin Architecture, and Glomerular Injury upon Adiponectin Receptor 1 Depletion. J Am Soc Nephrol 2025; 36:825-844. [PMID: 39874092 PMCID: PMC12059104 DOI: 10.1681/asn.0000000611] [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: 09/04/2023] [Accepted: 01/14/2025] [Indexed: 01/30/2025] Open
Abstract
Key Points Glomerular expression of adiponectin receptor 1 (AdipoR1) was lower in people with type 2 diabetes and correlates with podocyte loss. AdipoR1 knockout induced glomerular injury and fibrosis in mice, predominantly in males. AdipoR1 knockdown podocytes showed impaired trafficking of active integrin β 1, fibronectin accumulation, impaired adhesion, and increased apoptosis. Background Deficiency of adiponectin and its downstream signaling may contribute to the pathogenesis of kidney injury in type 2 diabetes. Adiponectin activates intracellular signaling using adiponectin receptor 1 (AdipoR1) and adiponectin receptor 2, but the role of adiponectin receptor–mediated signaling in glomerular injury in type 2 diabetes remains unknown. Methods The expression of AdipoR1 in the kidneys of people with type 2 diabetes and the expression of podocyte proteins or injury markers in the kidneys of AdipoR1 knockout (AdipoR1-KO) mice and immortalized AdipoR1-deficient human podocytes were investigated by immunohistochemistry and immunoblotting. The functional role of AdipoR1 was studied in AdipoR1-deficient podocytes by performing assays for apoptosis, cytokine secretion, mechanical stress, adhesion, and endocytic trafficking. Results Glomerular AdipoR1 expression was lower in type 2 diabetes and associated kidney disease, correlating with higher body mass index and podocyte loss. Male AdipoR1-KO mice showed typical signs of early diabetic kidney disease, including albuminuria, glomerular structural abnormalities, and lower expression of central podocyte proteins; females were less affected. Podocyte apoptosis increased in female and male AdipoR1-KO mice, and excessive podocyte loss, potentially due to detachment, was detected in males. AdipoR1 deficiency impaired the yes-associated protein–mediated mechanoresponse and induced accumulation of the extracellular matrix (ECM) protein fibronectin in the glomeruli in vivo and podocytes in vitro . Functionally, AdipoR1 deficiency impaired endocytosis of the ECM receptor active integrin β 1, disturbed focal adhesion turnover, and remodulated podocyte-derived ECM, thereby reducing podocyte adhesion. Conclusions AdipoR1 deficiency in mice resulted in the development of kidney injury predominantly in males. Mechanistically, AdipoR1 loss in podocytes impaired endocytosis of active integrin β 1, which plausibly compromised focal adhesion dynamics, disturbed fibronectin matrix turnover, and hindered podocyte adhesion.
Collapse
Affiliation(s)
- Sonja Lindfors
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Constanze Schmotz
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Dominik Lewandowski
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, Irvine, California
| | - Annika Hau
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Leena Saikko
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eero Lehtonen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Ville Majaniemi
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Minna Karhe
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jette-Britt Naams
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Harry Nisen
- Abdominal Center, Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jukka Tienari
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Moin A. Saleem
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Katharina Pfeil
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Heiko Bugger
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Kirsi H. Pietiläinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Obesity Research Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Healthy Weight Hub, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Tuomas Mirtti
- Department of Pathology, University of Helsinki, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, Irvine, California
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, California
- Department of Chemistry, University of California, Irvine, Irvine, California
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California
| | - Sanna Lehtonen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
Sindhwani R, Bora KS, Hazra S. The dual challenge of diabesity: pathophysiology, management, and future directions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4891-4912. [PMID: 39680103 DOI: 10.1007/s00210-024-03713-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024]
Abstract
Diabesity, the concurrent occurrence of obesity and type-2 diabetes mellitus (T2DM), represents a pressing global health challenge characterized by intricate pathophysiological mechanisms and a wide range of associated comorbidities. Central to its development are insulin resistance, metabolic syndrome, and chronic low-grade inflammation mediated by dysregulated adipokine secretion and systemic metabolic dysfunction. These mechanisms underpin the progression of diabesity and its complications, including cardiovascular disease and hypertension. Management strategies encompass lifestyle interventions focusing on tailored dietary modifications and structured physical activity, pharmacological treatments targeting both glycemic control and weight loss, and surgical interventions such as bariatric surgery, which have demonstrated efficacy in achieving durable outcomes. Clinical trials and meta-analyses underscore the comparative advantages of different treatment modalities in terms of efficacy, safety, and sustainability. Moreover, long-term follow-up studies emphasize the critical need for sustained multidisciplinary interventions to prevent relapse and enhance patient outcomes. Future advancements in management include exploring precision medicine approaches that integrate individual metabolic profiles, lifestyle factors, and emerging therapeutic innovations. A multidisciplinary approach combining advanced therapeutic strategies and patient-centered care remains pivotal for optimizing management and improving prognoses for individuals with diabesity. This review highlights the complex interplay between obesity and T2DM, offering comprehensive insights into their pathophysiology, clinical presentation, and management paradigms.
Collapse
Affiliation(s)
- Ritika Sindhwani
- University Institute of Pharma Sciences, Chandigarh University, Mohali, 140413, Punjab, India
| | - Kundan Singh Bora
- University Institute of Pharma Sciences, Chandigarh University, Mohali, 140413, Punjab, India.
| | - Subhajit Hazra
- University Institute of Pharma Sciences, Chandigarh University, Mohali, 140413, Punjab, India
| |
Collapse
|
8
|
Wei Y, Yang J, Zu W, Wang M, Zhao Y. Progression in the In Vitro Macrophage Expansion. J Immunol Res 2025; 2025:9994439. [PMID: 40331017 PMCID: PMC12052461 DOI: 10.1155/jimr/9994439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Macrophages play essential roles in homeostasis and disease, and they were considered terminally differentiated cells that cannot proliferate. However, growing evidence shows that macrophages can self-renew in homeostasis and multiple pathological states in vivo and artificial induction in vitro. With the rise of immune cell therapy based on macrophages, large-scale in vitro expansion of macrophages has become more and more urgent. However, the proliferation of macrophages in vitro is still inefficient because of the heterogeneity of macrophages, complicated crosstalk between macrophages and their microenvironments, and poor understanding of macrophage proliferation regulations. In this review, we summarized the discoveries known to stimulate macrophage proliferation in vitro, including cytokines, small molecule compounds, metabolites, the composition of pathogens and apoptotic cells, natural product extracts, gene editing, and other factors, as well as related mechanisms. It can be concluded that the promotion of macrophage proliferation in vitro covers various approaches and mechanisms. However, it is still necessary to test more strategies and learn more macrophage proliferation mechanisms to achieve large-scale engineering expansion of macrophages in vitro.
Collapse
Affiliation(s)
- Yunpeng Wei
- Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518107, China
| | - Jingzhao Yang
- Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518107, China
| | - Wenhong Zu
- Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518107, China
| | - Mengran Wang
- Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518107, China
| | - Yong Zhao
- Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518107, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
9
|
Kalkman HO, Smigielski L. Ceramides may Play a Central Role in the Pathogenesis of Alzheimer's Disease: a Review of Evidence and Horizons for Discovery. Mol Neurobiol 2025:10.1007/s12035-025-04989-0. [PMID: 40295359 DOI: 10.1007/s12035-025-04989-0] [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: 02/06/2025] [Accepted: 04/19/2025] [Indexed: 04/30/2025]
Abstract
While several hypotheses have been proposed to explain the underlying mechanisms of Alzheimer's disease, none have been entirely satisfactory. Both genetic and non-genetic risk factors, such as infections, metabolic disorders and psychological stress, contribute to this debilitating disease. Multiple lines of evidence indicate that ceramides may be central to the pathogenesis of Alzheimer's disease. Tumor necrosis factor-α, saturated fatty acids and cortisol elevate the brain levels of ceramides, while genetic risk factors, such as mutations in APP, presenilin, TREM2 and APOE ε4, also elevate ceramide synthesis. Importantly, ceramides displace sphingomyelin and cholesterol from lipid raft-like membrane patches that connect the endoplasmic reticulum and mitochondria, disturbing mitochondrial oxidative phosphorylation and energy production. As a consequence, the flattening of lipid rafts alters the function of γ-secretase, leading to increased production of Aβ42. Moreover, ceramides inhibit the insulin-signaling cascade via at least three mechanisms, resulting in the activation of glycogen synthase kinase-3 β. Activation of this kinase has multiple consequences, as it further deteriorates insulin resistance, promotes the transcription of BACE1, causes hyperphosphorylation of tau and inhibits the transcription factor Nrf2. Functional Nrf2 prevents apoptosis, mediates anti-inflammatory activity and improves blood-brain barrier function. Thus, various seemingly unrelated Alzheimer's disease risk factors converge on ceramide production, whereas the elevated levels of ceramides give rise to the well-known pathological features of Alzheimer's disease. Understanding and targeting these mechanisms may provide a promising foundation for the development of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Hans O Kalkman
- Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Lukasz Smigielski
- Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Tao Z, Luo Z, Zou Z, Ye W, Hao Y, Li X, Zheng K, Wu J, Xia J, Zhao Y, Wang Y, Zhang X. Novel insights and an updated review of metabolic syndrome in immune-mediated organ transplant rejection. Front Immunol 2025; 16:1580369. [PMID: 40330480 PMCID: PMC12052740 DOI: 10.3389/fimmu.2025.1580369] [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] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Metabolic syndrome (MetS) is a group of symptoms that are characterized by abnormal changes in metabolic substances such as glucose, lipids, proteins, and bile acids. MetS is a common complication after organ transplantation and can further affect the survival and physiological function of the graft by reprograming the patient's immune environment. Additionally, MetS can influence the occurrence of post-transplant complications, such as infections. In recent years, research into the epidemiology and mechanisms of MetS has grown significantly. In this review, we summarize the mechanisms of MetS after transplantation and the mechanisms of hyperglycemia, insulin resistance, hyperlipidemia, abnormal bile acids, and abnormal amino acids on the body's immune cells as related to the effect of metabolic disorders on immune rejection after liver, kidney, heart, skin and other organ transplantation. Finally, we provide an overview of current treatment strategies and offer insights into potential future therapies for managing MetS in transplant recipients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yang Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| | - Yongjun Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Bielach-Bazyluk A, Bossowski F, Skorupska M, Mysliwiec H, Bossowski AT, Flisiak I. Psoriasis in Obese Adolescents with Diabetes-From Common Molecular Background to Vicious Circle of Metabolic Syndrome-Case Report and Review of Literature. Cells 2025; 14:610. [PMID: 40277935 PMCID: PMC12026325 DOI: 10.3390/cells14080610] [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: 03/17/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
Psoriasis and type 1 diabetes mellitus (T1DM) are chronic autoimmune diseases sharing common immunological pathways, particularly the involvement of interleukin 17 (IL-17), driving Th17-mediated inflammation. This review explores the overlap between psoriasis, obesity, T1DM, and necrobiosis lipoidica (NL), a skin condition associated with diabetes. Obesity exacerbates inflammation through immune cell activation in adipose tissue and the release of proinflammatory adipokines, such as leptin, resistin, and IL-18, which enhance autoimmune responses and insulin resistance. Leptin promotes the differentiation of Th1 and Th17 cells, which are central to autoimmune responses in both psoriasis and T1DM. The coexistence of psoriasis, T1DM, and insulin resistance further complicates metabolic control, increasing the risk of complications like diabetic nephropathy and cardiovascular disease. Biologic treatments targeting IL-17A and IL-17F offer promising therapeutic options for managing both skin and metabolic symptoms. The early identification and management of metabolic risk factors, along with personalized interventions, are essential to improve clinical outcomes in patients with psoriasis and T1DM, particularly in obese individuals. This case report and review highlight the complex interplay of these conditions and emphasize the need for integrated treatment strategies.
Collapse
Affiliation(s)
- Angelika Bielach-Bazyluk
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland; (H.M.)
| | - Filip Bossowski
- Students’ Scientific Society at the Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland
| | - Magdalena Skorupska
- Students’ Scientific Society at the Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland
| | - Hanna Mysliwiec
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland; (H.M.)
| | - Artur Tadeusz Bossowski
- Department of Pediatrics, Endocrinology, Diabetology with Cardiology Divisions, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Iwona Flisiak
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland; (H.M.)
| |
Collapse
|
12
|
Tilg H, Ianiro G, Gasbarrini A, Adolph TE. Adipokines: masterminds of metabolic inflammation. Nat Rev Immunol 2025; 25:250-265. [PMID: 39511425 DOI: 10.1038/s41577-024-01103-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Adipose tissue is an immunologically active organ that controls host physiology, partly through the release of mediators termed adipokines. In obesity, adipocytes and infiltrating leukocytes produce adipokines, which include the hormones adiponectin and leptin and cytokines such as tumour necrosis factor and IL-1β. These adipokines orchestrate immune responses that are collectively referred to as metabolic inflammation. Consequently, metabolic inflammation characterizes metabolic disorders and promotes distinct disease aspects, such as insulin resistance, metabolic dysfunction-associated liver disease and cardiovascular complications. In this unifying concept, adipokines participate in the immunological cross-talk that occurs between metabolically active organs in metabolic diseases, highlighting the fundamental role of adipokines in obesity and their potential for therapeutic intervention. Here, we summarize how adipokines shape metabolic inflammation in mice and humans, focusing on their contribution to metabolic disorders in the setting of obesity and discussing their value as therapeutic targets.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Gianluca Ianiro
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
13
|
Yang HS, Kim SN, Ro JH, Hur M. Adiponectin and All-Cause Mortality in Patients with Chronic Kidney Disease: A Systematic Review and Meta-Analysis. Metabolites 2025; 15:230. [PMID: 40278358 PMCID: PMC12028947 DOI: 10.3390/metabo15040230] [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: 02/09/2025] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Elevated levels of adiponectin in chronic kidney disease (CKD) have been paradoxically associated with increased mortality. This meta-analysis aimed to evaluate the association between circulating adiponectin levels and all-cause mortality in patients with CKD, in total and various subgroups. Methods: We systematically searched PubMed, Embase, and Cochrane Library from their inception to December 2024 for studies examining baseline adiponectin levels and observed mortality outcomes in patients with CKD. Studies were included if they evaluated CKD stages 2-5 patients, measured baseline circulating adiponectin levels, and reported hazard ratios (HRs) for all-cause mortality. We excluded non-original research, studies of acute conditions, normal kidney function, kidney transplantation, and those using log-transformed or standardized HRs. HRs with a 95% confidence interval (CI) for all-cause mortality risk per 1 µg/mL increase in adiponectin were extracted and analyzed using the Comprehensive Meta-Analysis Version 4. Study quality was assessed using the Newcastle-Ottawa Scale. Results: Twelve studies with 2523 subjects were included. The pooled unadjusted HR was 1.003 (95% CI: 0.981-1.025) using a random-effects model (I2 = 79%). Subgroup analyses demonstrated increased mortality risk with elevated adiponectin levels in non-Asia (HR 1.021 [95% CI: 1.006-1.037], p = 0.006), studies with female proportion <47% (HR 1.021 [95% CI: 1.009-1.033], p < 0.001), and studies with body mass index ≥25 kg/m2 (HR 1.023 [95% CI: 1.008-1.038], p = 0.003). In contrast, higher adiponectin levels were associated with decreased mortality risk in the peritoneal dialysis group (HR 0.956 [95% CI: 0.934-0.979], p < 0.001) and female proportion ≥47% group (HR 0.929 [95% CI: 0.874-0.988], p = 0.019). Discussion/Conclusions: This meta-analysis revealed that elevated adiponectin levels have varying associations with the risk of all-cause mortality across CKD patient subgroups. These findings suggest that the prognostic value of adiponectin levels in CKD may be modulated by demographic and clinical factors. Limitations include poor generalizability with underrepresentation of early-stage CKD. This research received no external funding and was not registered.
Collapse
Affiliation(s)
- Hyun Suk Yang
- Department of Cardiovascular Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul 05029, Republic of Korea;
| | - Soo-Nyung Kim
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul 05029, Republic of Korea;
| | - Jung-Hoon Ro
- Department of Biomedical Engineering, Pusan National University School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea;
| | - Mina Hur
- Department of Laboratory Medicine, Konkuk University School of Medicine, Konkuk University Medical Center, Seoul 05030, Republic of Korea
| |
Collapse
|
14
|
Halpern D, Farber I, Anav Y, Tsitrina A, Lewis EC, Silberstein E. Alpha-1-Antitrypsin Enhances Fat Graft Survival in a Murine Model. Adv Wound Care (New Rochelle) 2025. [PMID: 40105893 DOI: 10.1089/wound.2024.0176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Objective: Fat grafting is widely applied for various purposes, including volume restoration, improving tissue quality, and promoting wound healing, but it has poor long-term graft survival predictability. Alpha-1-antitrypsin (AAT) administration is hypothesized to improve fat graft outcomes by expediting inflammatory resolution and graft vascularity and reducing necrosis. Approach: Mice heterozygote to human AAT was grafted fat under the scalp alongside 400 µg/graft AAT or albumin (ALB) on days 0 and 3. Graft volume was determined by micro-magnetic resonance imaging, and explants were assessed for viability, histology, immunohistochemistry, and expression of selected genes. AAT expression was examined in hypoxia-exposed adipose-derived stem cells (ADSCs). Results: After 90 days, AAT-treated grafts maintained higher volumes (70.06% vs. 34.54%, n = 8, p = 0.02) and displayed improved tissue quality. On day 10 after grafting, grafts exhibited more blood vessels (mean 1.94/mm2 vs. 0.33/mm2) and 6.25-fold more adiponectin transcript levels (n = 12, p = 0.02). Although day-3 interleukin (IL)-1β expression was 5-fold greater in AAT-treated grafts (n = 6, p = 0.4), day-10 IL-1β expression was 2-fold lower compared to ALB-treated grafts (n = 22, p = 0.01). In the Methoxynitrosulfophenyl-tetrazolium carboxanilide (XTT) assay, day-3 AAT-treated grafts were 1.56-fold more metabolically functional (n = 6, p = 0.04) and exhibited greater perilipin-positive regions (18.5% versus 3.1%). Hypoxia-exposed ADSC expressed 9-fold higher AAT transcript levels (p = 0.04). Innovation: Fat grafting outcomes improved by early AAT treatment, probably by accelerating inflammatory resolution. Due to its marked safety profile, the study's findings are for adjunct clinical-grade AAT therapy. Conclusion: AAT has a promising potential to be utilized as a fat graft outcome enhancer in terms of volume retention predictability and tissue quality.
Collapse
Affiliation(s)
- Dor Halpern
- Plastic and Reconstructive Surgery Department, Soroka University Medical Centre, Beer Sheva, Israel
| | - Idan Farber
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Yuval Anav
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
- Joyce and Irving Goldman Medical School, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Alexandra Tsitrina
- Ilse Katz Institute for Nanoscale Science and Technology, Ben Gurion University of the Negev,Beer Sheva, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Eldad Silberstein
- Plastic and Reconstructive Surgery Department, Soroka University Medical Centre, Beer Sheva, Israel
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
15
|
Mahabamunuge J, Sekula NM, Lepore C, Kudrimoti M, Upadhyay A, Alshowaikh K, Li HJ, Seifer DB, AlAshqar A. The Molecular Basis of Polycystic Ovary Syndrome and Its Cardiometabolic Correlates: Exploring the Intersection and Its Clinical Implications-A Narrative Review. Biomedicines 2025; 13:709. [PMID: 40149685 PMCID: PMC11940587 DOI: 10.3390/biomedicines13030709] [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: 01/26/2025] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Recent studies have highlighted the association between polycystic ovary syndrome (PCOS) and cardiometabolic diseases, leading to an improved understanding of the underlying mechanistic factors. PCOS significantly increases cardiovascular risk by predisposing individuals to various subclinical and clinical conditions, including atherosclerosis and type 2 diabetes mellitus. Additionally, it interacts synergistically with other traditional cardiovascular risk factors, such as obesity, hyperlipidemia, and insulin resistance. Several molecular mechanisms involving genetics, epigenetics, adipokine secretion, hyperandrogenemia, and hyperinsulinemia play a role in the relationship between PCOS and these comorbidities. For instance, androgen excess has been implicated in the development of hypertension, type 2 diabetes mellitus, endothelial dysfunction, and ultimately, broader cardiovascular disease. A deeper understanding of these underlying mechanisms facilitates the development of diagnostic, preventative, and therapeutic strategies directed at reducing cardiometabolic morbidity. This narrative review summarizes the current evidence, explores the potential clinical implications of these findings, and discusses emerging therapies to reduce cardiometabolic morbidity in women with PCOS.
Collapse
Affiliation(s)
- Jasmin Mahabamunuge
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Nicole M. Sekula
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Christina Lepore
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Meghana Kudrimoti
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Animesh Upadhyay
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Khadija Alshowaikh
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Howard J. Li
- Division of Reproductive Endocrinology and Infertility, National Institutes of Health, Bethesda, MD 20892, USA;
| | - David B. Seifer
- Division of Reproductive Endocrinology and Infertility, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Abdelrahman AlAshqar
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| |
Collapse
|
16
|
Li J, Liu T, Xian M, Zhou K, Wei J. The Power of Exercise: Unlocking the Biological Mysteries of Peripheral-Central Crosstalk in Parkinson's Disease. J Adv Res 2025:S2090-1232(25)00143-2. [PMID: 40049515 DOI: 10.1016/j.jare.2025.03.001] [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: 07/23/2024] [Revised: 01/06/2025] [Accepted: 03/01/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Exercise is a widely recognized non-pharmacological treatment for Parkinson's Disease (PD). The bidirectional regulation between the brain and peripheral organs has emerged as a promising area of research, with the mechanisms by which exercise impacts PD closely linked to the interplay between peripheral signals and the central nervous system. AIM OF REVIEW This review aims to summarize the mechanisms by which exercise influences peripheral-central crosstalk to improve PD, discuss the molecular processes mediating these interactions, elucidate the pathways through which exercise may modulate PD pathophysiology, and identify directions for future research. KEY SCIENTIFIC CONCEPTS OF REVIEW This review examines how exercise-induced cytokine release promotes neuroprotection in PD. It discusses how exercise can stimulate cytokine secretion through various pathways, including the gut-brain, muscle-brain, liver-brain, adipose-brain, and bone-brain axes, thereby alleviating PD symptoms. Additionally, the potential contributions of the heart-brain, lung-brain, and spleen-brain axes, as well as multi-axis crosstalk-such as the brain-gut-muscle and brain-gut-bone axes-are explored in the context of exercise therapy. The study highlights the need for further research into peripheral-central crosstalk and outlines future directions to address challenges in clinical PD therapy.
Collapse
Affiliation(s)
- Jingwen Li
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Ke Zhou
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China.
| | - Jianshe Wei
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China; Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
17
|
Li H, Gong Y, Wang Y, Sang W, Wang C, Zhang Y, Zhang H, Liu P, Liu M, Sun H. β-Sitosterol modulates osteogenic and adipogenic balance in BMSCs to suppress osteoporosis via regulating mTOR-IMP1-Adipoq axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156400. [PMID: 39848018 DOI: 10.1016/j.phymed.2025.156400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/05/2025] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Osteoporosis (OP) is a prevalent global health concern, impacting millions of individuals, especially the elderly. The etiology of senile OP is associated with the imbalance of osteogenic and adipogenic differentiation in the bone marrow mesenchymal stem cells (BMSCs). The imbalance of BMSCs differentiation fate will leading to bone loss and lipids accumulation. β-sitosterol, a naturally occurring phytosterol which is abundant in plants and has a similar structure to cholesterol, demonstrates diverse bioactivities, including lipid-lowering effect and osteogenesis-inducing effects. These effects indicate that β-sitosterol might have anti-OP effects. Nevertheless, the precise mechanism underlying β-sitosterol's anti-osteoporotic efficacy via modulating BMSCs differentiation fate remains obscure. PURPOSE This study endeavors to elucidate whether β-sitosterol has the potential to augment the osteogenic differentiation of BMSCs while mitigating their adipogenic differentiation, thereby exerting an anti-OP effect; and to reveal its molecular mechanisms of action. METHODS In this study, a dosage form HP-β-cyclodextrin-coated β-sitosterol was developed for intragastric administration in mice to enhancing its bioavailability. Subsequently by using an integrative approach encompassing bioinformatics, computer molecular simulations, high-throughput sequencing, and in vitro/vivo as well as in-tube experiments, we investigated the anti-osteoporotic and bone healing effects of β-sitosterol and delineated its underlying mechanisms. RESULTS Our findings demonstrate that β-sitosterol exhibits anti-osteoporotic and bone healing effects both in vitro and in vivo by modulating the osteogenic and adipogenic differentiation of BMSCs. Mechanistically, these effects are mediated through the direct inhibition of mTOR's kinase activity independent of mediating autophagy, leading to the suppression of the mTOR-IMP1-Adipoq axis in BMSCs. CONCLUSION These results unveil β-sitosterol as a promising therapeutic agent for OP, shedding light on its underlying mechanisms. This research contributes potential candidates for diagnostic and therapeutic interventions in the realm of OP.
Collapse
Affiliation(s)
- Hao Li
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China; Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Ying Gong
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yanna Wang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Wanyu Sang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China; Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Changyuan Wang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Yukun Zhang
- Three Gorges Medical College, Wanzhou, Chongqing, China
| | - Hanrui Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Peixuan Liu
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Mozhen Liu
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Huijun Sun
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China; Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.
| |
Collapse
|
18
|
Landim-Vieira M, Nieto Morales PF, ElSafty S, Kahmini AR, Ranek MJ, Solís C. The role of mechanosignaling in the control of myocardial mass. Am J Physiol Heart Circ Physiol 2025; 328:H622-H638. [PMID: 39739566 DOI: 10.1152/ajpheart.00277.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 12/03/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Regulation of myocardial mass is key for maintaining cardiovascular health. This review highlights the complex and regulatory relationship between mechanosignaling and myocardial mass, influenced by many internal and external factors including hemodynamic and microgravity, respectively. The heart is a dynamic organ constantly adapting to changes in workload (preload and afterload) and mechanical stress exerted on the myocardium, influencing both physiological adaptations and pathological remodeling. Mechanosignaling pathways, such as the mitogen-activated protein kinases (MAPKs) and the phosphoinositide 3-kinases and serine/threonine kinase (PI3K/Akt) pathways, mediate downstream effects on gene expression and play key roles in transducing mechanical cues into biochemical signals, thereby modulating cellular processes, including control of myocardial mass. Dysregulation of these processes can lead to pathological cardiac remodeling, such as hypertrophic cardiomyopathy. Furthermore, recent studies have highlighted the importance of protein quality control mechanisms, such as the ubiquitin-proteasome system, in settings of extreme physiological conditions that alter the heart workload such as pregnancy and microgravity. Overall, this review provides a thorough insight into how mechanical signals are converted into chemical signals to regulate myocardial mass in both healthy and diseased conditions.
Collapse
Affiliation(s)
- Maicon Landim-Vieira
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Paula F Nieto Morales
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Summer ElSafty
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Aida Rahimi Kahmini
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Christopher Solís
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
19
|
Suh JH, Lee Y, Jin SP, Kim EJ, Seo EY, Li N, Oh JH, Kim SJ, Lee SH, Lee DH, Cho S, Chung JH. Adiponectin Prevents Skin Inflammation in Rosacea by Suppressing S6 Phosphorylation in Keratinocytes. J Invest Dermatol 2025; 145:548-558.e5. [PMID: 39122145 DOI: 10.1016/j.jid.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/20/2024] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
Numerous recent evidence highlights epidemiological connections between rosacea and metabolic disorders. However, the precise path through which metabolic factors impact rosacea risk is still unclear. Therefore, this study aims to investigate the role of adiponectin, a crucial adipokine that regulates metabolic homeostasis, in the pathogenesis of rosacea. We elucidated a detrimental feedback loop between rosacea-like skin inflammation and decreased levels of skin adiponectin. To elaborate, rosacea lesional skin exhibits diminished adiponectin expression compared with nonlesional areas in the same patients. Induction of rosacea-like inflammation reduced adiponectin levels in the skin by generating inflammatory cytokines that suppress adiponectin production from subcutaneous adipocytes. Conversely, complete depletion of adiponectin exacerbated rosacea-like features in the mouse model. Mechanistically, adiponectin deficiency led to heightened S6 phosphorylation, a marker of the mTORC1 signaling pathway, in the epidermis. Adiponectin significantly inhibited S6 phosphorylation in cultured keratinocytes. Notably, replenishing adiponectin whole protein or topically applying an agonist for adiponectin receptor 1 successfully improved rosacea-like features in mice. This study contributes to understanding the role of adiponectin in skin inflammation associated with rosacea pathophysiology, suggesting that restoring adiponectin function in the skin could be a potential therapeutic strategy.
Collapse
Affiliation(s)
- Joong Heon Suh
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Youngae Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Seon-Pil Jin
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Eun Ju Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Eun Young Seo
- Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Na Li
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Jang-Hee Oh
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Sung Joon Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea; Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Si-Hyung Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Soyun Cho
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University Medical Research Center, Seoul, Republic of Korea; Institute of Aging, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
20
|
Zhang Y, Qu Z, Mao Z, Liu H, Wang W, Jia L. Adiponectin targets the AMPK/mTOR signaling pathway to alleviate cognitive impairment in epilepsy. Mol Med Rep 2025; 31:64. [PMID: 39749696 PMCID: PMC11726285 DOI: 10.3892/mmr.2025.13429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/10/2024] [Indexed: 01/04/2025] Open
Abstract
Among patients with chronic epilepsy, 70‑80% have cognitive impairment. To investigate the relationship between adiponectin (ADPN) and the cognitive level in epilepsy and its mechanism, 20 epileptic patients and 20 healthy controls were included for the assessment of the cognitive level. An ELISA was used to evaluate the serum ADPN level. An epileptic rat model was established and treated with AdipoRon, an ADPN receptor (AdipoR) agonist, which binds to AdipoR1 and AdipoR2. The Morris water maze test was used to assess the cognitive function of rats, and the expression levels of the synapsis‑associated proteins postsynaptic density protein 95 (PSD95), synaptosomal associated protein 25 (SNAP25) and synaptophysin (SYP), as well as AMP‑activated protein kinase (AMPK), mTOR, phosphorylated (p‑)AMPK and p‑mTOR were determined by immunoblotting. Serum ADPN levels were positively correlated with the Montreal cognitive assessment score. AdipoRon improved the cognitive function of epileptic rats, maintained the structural integrity of hippocampal neurons and reduced neuronal damage. It also promoted the mRNA expression of AdipoR1 and AdipoR2 in the hippocampus. Furthermore, AdipoRon increased the expression of the synapsis‑associated proteins PSD95, SNAP25 and SYP by activating the AMPK/mTOR signaling pathway. ADPN improved cognitive impairment in epilepsy by targeting the AMPK/mTOR signaling pathway, providing novel insights for the treatment of epilepsy.
Collapse
Affiliation(s)
- Yaoyuan Zhang
- Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
- Department of Neurology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075061, P.R. China
| | - Zhenzhen Qu
- Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Zhuofeng Mao
- Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Hu Liu
- Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Weiping Wang
- Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Lijing Jia
- Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
21
|
Qian Y, Qi Y, Lin J, Zhang T, Mo L, Xue Q, Zheng N, Niu Y, Dong X, Shi Y, Jiang Y. AdipoRon ameliorates chronic ethanol induced cardiac necroptosis by reducing ceramide mediated mtROS. Free Radic Biol Med 2025; 229:237-250. [PMID: 39805512 DOI: 10.1016/j.freeradbiomed.2025.01.018] [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: 10/14/2024] [Revised: 12/30/2024] [Accepted: 01/11/2025] [Indexed: 01/16/2025]
Abstract
Chronic ethanol (EtOH) consumption has been widely recognized as a significant contributor to cardiotoxicity. However, no specific treatment is currently available to ameliorate chronic ethanol induced cardiotoxicity. Adiponectin receptor agonist AdipoRon exerts protective effects in multiple organs through alleviating lipotoxicity. Our previous study showed that chronic ethanol consumption increased de novo ceramide synthesis and necroptosis in myocardium. In this study, we investigated the role of AdipoRon on ceramide metabolism and necroptosis in chronic ethanol-treated myocardium. Eight-week-old C57/BL6J mice were fed with a Lieber-Decarli diet containing vehicle or AdipoRon for 12 weeks. Cardiac function, histology and oxidative stress were assessed. We found that chronic ethanol treatment decreased expression of AdipoR2 in myocardium and H9c2 cells, whereas AdipoRon improved cardiac function, reduced myocardium ceramide levels and suppressed necroptosis. By pharmacological interventions, RNA interference and point mutations in AdipoR2, we demonstrated that AdipoRon reduced ceramide levels through PPARα mediated lipid metabolism rather than AdipoR2's ceramidase activity. Using transmission electron microscope and reactive oxygen species (ROS) staining, we showed that chronic ethanol induced myocardium mitochondria damage and mitochondrial reactive oxygen species (mtROS) accumulation. Meanwhile, we found that AdipoRon ameliorated chronic ethanol induced cardiac necroptosis via the SIRT3-SOD2-mtROS pathway. Moreover, C6 ceramide treatment recapitulated chronic ethanol in inducing mtROS and necroptosis, whereas the ceramide synthesis inhibitors myriocin (MYR) and fumonisin B1 (FB1) attenuated chronic ethanol induced mtROS and necroptosis. Collectively, AdipoRon ameliorates chronic ethanol induced cardiac necroptosis by reducing ceramide de novo synthesis and mtROS, which highlights the therapeutic potential of targeting ceramide metabolism and oxidative stress pathways in treating ethanol induced cardiotoxicity.
Collapse
Affiliation(s)
- Yile Qian
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yanyu Qi
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Junyi Lin
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Tianyi Zhang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Lingjie Mo
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qiupeng Xue
- Forensic Science and Information Technology Research Centre of Supreme People's Procuratorate, Beijing, 100726, China
| | - Nianchang Zheng
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yaqin Niu
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiaoru Dong
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yan Shi
- Academy of Forensic Science Shanghai Key Laboratory of Forensic Medicine, Shanghai, 200063, China.
| | - Yan Jiang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
22
|
Tan A, Dunseath G, Thomas RL, Prior SL, Bracken RM, Churm R. Effect of home-based exercise with or without a Mediterranean-style diet on adiposity markers in postmenopausal women: A randomized-control trial. Physiol Rep 2025; 13:e70239. [PMID: 39921234 PMCID: PMC11805804 DOI: 10.14814/phy2.70239] [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: 09/17/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
Advancing age and estrogen deficiency increases susceptibility of post-menopausal women (PMW) to abdominal obesity and manifestation of cardiometabolic disease. There is limited evidence on the effect of lifestyle interventions on adiposity markers within at-risk PMW. Therefore, this study aims to evaluates an 8-weeks of home-based, equipment-free, interval training (HEFIT) with or without Mediterranean-style diet (MD) on adiposity markers in physically inactive, postmenopausal women with overweight/obesity. Thirty PMW (56.7 ± 3.9 years, BMI: 30.5 ± 5.2 kg/m2) were randomly assigned to three groups: (i) Ex; HEFIT thrice weekly/week, (ii) EX + MD, or (iii) CTL; control. Visceral Adiposity Index (VAI), body weight, BMI, waist and hip circumference (WC; HC), visceral adipose tissue (VAT), total body fat percentage, leptin, and adiponectin were determined pre- and post-8-week intervention. There was no significant between group effect on VAI. Compared to CTL, a significant between group reduction was seen in weight, BMI, and WC in both EX and EX+D (p < 0.05). Leptin and adiponectin remained unchanged in all groups (p > 0.05). Adherence rates were 85% and 96% for EX and EX+MD, respectively, and 80% of EX+D of participants had optimal adherence to diet. Concluding HEFIT with or without dietary changes could improve adiposity in overweight/obese postmenopausal women.
Collapse
Affiliation(s)
- Abbigail Tan
- Applied Sports Technology, Exercise and Medicine (A‐STEM) Research Centre, Faculty of Science and EngineeringSwansea UniversitySwanseaUK
| | - Gareth Dunseath
- Diabetes Research Group, Grove BuildingSwansea UniversitySwanseaUK
| | | | - Sarah L. Prior
- Diabetes Research Group, Grove BuildingSwansea UniversitySwanseaUK
| | - Richard M. Bracken
- Applied Sports Technology, Exercise and Medicine (A‐STEM) Research Centre, Faculty of Science and EngineeringSwansea UniversitySwanseaUK
| | - Rachel Churm
- Applied Sports Technology, Exercise and Medicine (A‐STEM) Research Centre, Faculty of Science and EngineeringSwansea UniversitySwanseaUK
| |
Collapse
|
23
|
Biagioli V, Mela F, Ferraro P, Villano G, Orsini A, Diana MC, Striano P, Santangelo A. The Interplay Between Gut Microbiota, Adipose Tissue, and Migraine: A Narrative Review. Nutrients 2025; 17:337. [PMID: 39861467 PMCID: PMC11768392 DOI: 10.3390/nu17020337] [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: 12/24/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Migraine, a prevalent neurovascular disorder, affects millions globally and is associated with significant morbidity. Emerging evidence suggests a crucial role of the gut microbiota and adipose tissue in the modulation of migraine pathophysiology, particularly through mechanisms involving neuroinflammation and metabolic regulation. MATERIAL AND METHODS A narrative review of the literature from 2000 to 2024 was conducted using the PubMed database. Studies addressing the relationships between microbiota, adipose tissue, and migraine-including dietary interventions and their impact-were analyzed. RESULTS The findings highlight a bidirectional gut-brain axis, with gut microbiota influencing neuroinflammation via metabolites such as short-chain fatty acids (SCFAs). Obesity exacerbates migraine severity through chronic inflammation and the dysregulation of adipocytokines like leptin and adiponectin. Dietary patterns, such as low glycemic index diets and Mediterranean diets, and the use of prebiotics, probiotics, and postbiotics show potential in migraine management. CONCLUSIONS This review underscores the need for integrative approaches targeting the microbiota-gut-brain axis and adipose tissue in migraine therapy. Future studies should explore longitudinal effects and personalized interventions to optimize outcomes.
Collapse
Affiliation(s)
- Valentina Biagioli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
| | - Federica Mela
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Paola Ferraro
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Gianmichele Villano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
| | - Alessandro Orsini
- Pediatric Neurology, Pediatric Department, AOUP Santa Chiara Hospital, 56100 Pisa, Italy
| | - Maria Cristina Diana
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Andrea Santangelo
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| |
Collapse
|
24
|
Russo L, Babboni S, Andreassi MG, Daher J, Canale P, Del Turco S, Basta G. Treating Metabolic Dysregulation and Senescence by Caloric Restriction: Killing Two Birds with One Stone? Antioxidants (Basel) 2025; 14:99. [PMID: 39857433 PMCID: PMC11763027 DOI: 10.3390/antiox14010099] [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: 12/20/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Cellular senescence is a state of permanent cell cycle arrest accompanied by metabolic activity and characteristic phenotypic changes. This process is crucial for developing age-related diseases, where excessive calorie intake accelerates metabolic dysfunction and aging. Overnutrition disturbs key metabolic pathways, including insulin/insulin-like growth factor signaling (IIS), the mammalian target of rapamycin (mTOR), and AMP-activated protein kinase. The dysregulation of these pathways contributes to insulin resistance, impaired autophagy, exacerbated oxidative stress, and mitochondrial dysfunction, further enhancing cellular senescence and systemic metabolic derangements. On the other hand, dysfunctional endothelial cells and adipocytes contribute to systemic inflammation, reduced nitric oxide production, and altered lipid metabolism. Numerous factors, including extracellular vesicles, mediate pathological communication between the vascular system and adipose tissue, amplifying metabolic imbalances. Meanwhile, caloric restriction (CR) emerges as a potent intervention to counteract overnutrition effects, improve mitochondrial function, reduce oxidative stress, and restore metabolic balance. CR modulates pathways such as IIS, mTOR, and sirtuins, enhancing glucose and lipid metabolism, reducing inflammation, and promoting autophagy. CR can extend the health span and mitigate age-related diseases by delaying cellular senescence and improving healthy endothelial-adipocyte interactions. This review highlights the crosstalk between endothelial cells and adipocytes, emphasizing CR potential in counteracting overnutrition-induced senescence and restoring vascular homeostasis.
Collapse
Affiliation(s)
- Lara Russo
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Serena Babboni
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Maria Grazia Andreassi
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Jalil Daher
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, El-Koura 100, Lebanon;
| | - Paola Canale
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Serena Del Turco
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Giuseppina Basta
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| |
Collapse
|
25
|
Chen T, Ren Q, Ma F. New insights into constitutive neutrophil death. Cell Death Discov 2025; 11:6. [PMID: 39800780 PMCID: PMC11725587 DOI: 10.1038/s41420-025-02287-1] [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: 06/15/2024] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
Neutrophils undergo rapid aging and death known as constitutive or spontaneous death. Constitutive neutrophil death (CND) contributes to neutrophil homeostasis and inflammation resolution. CND has long been considered to be apoptotic until our findings reveal that it was a heterogeneous combination of diverse death. Furthermore, dead neutrophils retain functional roles via multiple manners. This review provides an overview of current research on the mechanism and modulation of CND. More noteworthy, we also summarize the after-death events of neutrophils. The fate of neutrophils can be changed under pathological conditions, so the involvement of CND in diseases and CND-related therapeutic strategies are also addressed.
Collapse
Affiliation(s)
- Tong Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key laboratory for prevention and control of hematological disease treatment related infection, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key laboratory for prevention and control of hematological disease treatment related infection, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Fengxia Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key laboratory for prevention and control of hematological disease treatment related infection, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
26
|
Yang X, Wang X, Yang Z, Lu H. Iron-Mediated Regulation in Adipose Tissue: A Comprehensive Review of Metabolism and Physiological Effects. Curr Obes Rep 2025; 14:4. [PMID: 39753935 DOI: 10.1007/s13679-024-00600-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/11/2024] [Indexed: 01/14/2025]
Abstract
PURPOSE OF REVIEW Review the latest data regarding the intersection of adipose tissue (AT) and iron to meet the needs of AT metabolism and the progression of related diseases. RECENT FINDINGS Iron is involved in fundamental biological metabolic processes and is precisely fine-tuned within the body to maintain cellular, tissue and even systemic iron homeostasis. AT not only serves as an energy storage depot but also represents the largest endocrine organ in the human body, maintaining systemic metabolic homeostasis. It is involved in physiological processes such as energy storage, insulin sensitivity regulation and lipid metabolism. As a unique iron-sensing tissue, AT expresses related regulatory factors, including the classic hepcidin, ferroportin (FPN), iron regulatory protein/iron responsive element (IRP/IRE) and ferritin. Consequently, the interaction between AT and iron is intricately intertwined. Imbalance of iron homeostasis produces the potential risks of steatosis, impaired glucose tolerance and insulin resistance, leading to AT dysfunction diseases, including obesity, type 2 diabetes and metabolic dysfunction-associated steatotic liver disease (MASLD). Despite the role of AT iron has garnered increasing attention in recent years, a comprehensive review that systematically organizes the connection between iron and AT remains lacking. Given the necessity of iron homeostasis, emphasizing its potential impact on AT function and metabolism regulation provides valuable insights into physiological effects such as adipocyte differentiation and thermogenesis. Futhermore, regulators including adipokines, mitochondria and macrophages have been mentioned, along with analyzing the novel perspective of iron as a key mediator influencing the fat-gut crosstalk.
Collapse
Affiliation(s)
- Xinyu Yang
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Xianghong Wang
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Zhe Yang
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Hongyun Lu
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China.
| |
Collapse
|
27
|
Matsui M, Fukuda A, Onishi S, Ushiro K, Nishikawa T, Asai A, Kim SK, Nishikawa H. Metabolic Syndrome and Somatic Composition: A Large Cross-sectional Analysis. In Vivo 2025; 39:381-389. [PMID: 39740891 PMCID: PMC11705143 DOI: 10.21873/invivo.13839] [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: 09/07/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND/AIM To elucidate the relationship between metabolic syndrome (Mets) and somatic composition [fat mass, fat-free (FF) mass, and fat to fat-free (F-FF) ratio] among health checkup recipients (7,776 males and 10,121 females). PATIENTS AND METHODS We classified study subjects into four types considering Japanese criteria for Mets; Type A is for males with waist circumference (WC) <85 cm and females with WC <90 cm, Type B is for males with WC ≥85 cm and females with WC ≥90 cm, but without any metabolic abnormalities, Type C is for males with WC ≥85 cm and females with WC ≥90 cm and one metabolic disorder (pre-Mets), and Type D is Mets. We compared baseline characteristics among types of A, B, C, and D. RESULTS F index, FF index, and F-FF ratio showed an increasing trend with increasing risk factors for Mets in both sexes. CONCLUSION This study demonstrates a clear correlation between somatic composition and the severity of metabolic syndrome (Mets). As Mets risk factors increase, fat mass, fat-free mass, and the fat-to-fat-free ratio also rise, indicating that body composition shifts with disease progression. These findings emphasize the need for early intervention, such as exercise and diet, to manage somatic composition imbalances and reduce complications like insulin resistance.
Collapse
Affiliation(s)
- Masahiro Matsui
- Second Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Akira Fukuda
- Osaka Medical and Pharmaceutical University Health Science Clinic, Takatsuki, Japan
| | - Saori Onishi
- Second Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Kosuke Ushiro
- Second Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Tomohiro Nishikawa
- Second Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Akira Asai
- Second Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Soo Ki Kim
- Department of Gastroenterology, Kobe Asahi Hospital, Kobe, Japan
| | - Hiroki Nishikawa
- Second Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan;
| |
Collapse
|
28
|
Liu C, Xin Y, Huang Y, Xu L, Zhou R, Wang Y, Wang W. Reduction of Hepatic Fat Content by Dulaglutide for the Treatment of Diabetes Mellitus: A Two-Centre Open, Single-Arm Trial. Endocrinol Diabetes Metab 2025; 8:e70021. [PMID: 39718468 DOI: 10.1002/edm2.70021] [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: 07/24/2024] [Revised: 10/07/2024] [Accepted: 11/06/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND With the elevated level of NAFLD prevalence, the incidence of diabetes, hypertension, metabolic syndrome and other diseases is also significantly elevated. GLP-1RA can exert weight loss, glucose-lowering effects and various nonglycaemic effects. However, the relationship between quantitative reduction in hepatic fat content and improvement of pancreatic islet function by GLP-1RA is unclear. METHODS This trial was a single-arm open cohort study. A total of 38 patients with T2DM and NAFLD were enrolled in the GLP-1RA treatment group. The included patients were tested for biochemical and blood glucose levels, adiponectin and FGF21 levels, and liver fat content was measured using MRI. Measure the above indicators again after at least 3 months of GLP-1RA treatment. Divided into Q1 (average decrease of 0.37%) and Q2 (average decrease of 8.6%) groups based on the degree of reduction in liver fat content. RESULTS Q2 group showed an average reduction in liver fat content of 8.6%, a decrease in glycated haemoglobin of 18.17%, a weight loss of 7.29% and an increase in fasting c-peptide release by 1.03%, 1-h and 2-h postprandial c-peptide release by 28.86% and 18.28% respectively. In contrast, Q1 group had an average reduction in liver fat content of 0.37%, a decrease in glycated haemoglobin of only 6.53%, a weight loss of 3.41%, a decrease in fasting c-peptide release by 1.91% and an increase in 1-h and 2-h postprandial c-peptide release by 19.18% and 11.66% respectively. CONCLUSION Reduction in liver fat content effectively improves pancreatic islet function secretion, particularly postprandial c-peptide secretion, especially in the first hour after a meal. This improvement leads to a decrease in glycated haemoglobin levels and promotes better compliance with blood glucose control.
Collapse
Affiliation(s)
- Chuanfeng Liu
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yu Xin
- Department of Endocrinology and Metabolic Diseases, Jiaozuo People's Hospital, Jiaozuo, China
- Department of Endocrinology and Metabolic Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yajing Huang
- Department of Endocrinology and Metabolic Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lili Xu
- Department of Endocrinology and Metabolic Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruizhi Zhou
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yangang Wang
- Department of Endocrinology and Metabolic Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Wang
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
29
|
Wang Z, Liu X, Sheng L, Xie Y, Feng W, Yu L. Effects of duration of high-fat diet on adipocyte hyperplasia in rat epididymis. Obes Res Clin Pract 2025; 19:54-62. [PMID: 39922761 DOI: 10.1016/j.orcp.2025.02.001] [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: 08/21/2023] [Revised: 06/25/2024] [Accepted: 02/01/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND High-fat diet (HFD) contributes to obesity and enhances the expression of mature adipocyte markers. However, the effect of HFD on adipocyte hyperplasia remains controversial. This may be due to variations in the duration of HFD. This study aimed to investigate the effects of different durations of HFD on adipocyte hyperplasia and the expression of mature adipocyte-related markers in obese rats. METHODS We divided 32 Sprague-Dawley rats into four groups: B (standard diet control), H1 (HFD for four weeks), H2 (HFD for eight weeks), and H3 (HFD for 12 weeks). We evaluated the morphological changes in epididymal fat cells, measured serum inflammatory markers using enzyme-linked immunosorbent assay (ELISA) kits, and quantified adipocyte hyperplasia and maturation markers using western blotting. RESULTS We observed progressive increases in body weight, epididymal fat weight, serum leptin, TNF-α, IL-6, irisin, PPARγ, adiponectin, and FNDC5 protein expression over 8 weeks of HFD. 12 weeks of HFD intervention resulted in significant decreases in irisin, PPARγ, adiponectin, and FNDC5. Concurrently, the expression of perilipin A and ATGL declined with prolonged HFD. CONCLUSIONS Our results suggest that the duration of HFD significantly affects adipocyte ability to undergo hyperplasia in the epididymis of obese rats. Specifically, 4 weeks of HFD did not change the capacity for adipocyte hyperplasia, while 8 weeks of the diet enhanced this capacity. Interestingly, a longer diet duration (12 weeks) led to a decrease in adipocyte hyperplasia.
Collapse
Affiliation(s)
- Zhaoxin Wang
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Xiujuan Liu
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China.
| | - Lei Sheng
- Department of Scientific Research, Nanjing Sport Institute, Nanjing, China.
| | - Yuting Xie
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Wanyu Feng
- Department of Science Experiment Center, Nanjing Sport Institute, Nanjing, China
| | - Li Yu
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| |
Collapse
|
30
|
Jain U, Srivastava P, Sharma A, Sinha S, Johari S. Impaired Fibroblast Growth Factor 21 (FGF21) Associated with Visceral Adiposity Leads to Insulin Resistance: The Core Defect in Diabetes Mellitus. Curr Diabetes Rev 2025; 21:e260424229342. [PMID: 38676505 DOI: 10.2174/0115733998265915231116043813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/06/2023] [Accepted: 09/20/2023] [Indexed: 04/29/2024]
Abstract
The Central nervous system (CNS) is the prime regulator of signaling pathways whose function includes regulation of food intake (consumption), energy expenditure, and other metabolic responses like glycolysis, gluconeogenesis, fatty acid oxidation, and thermogenesis that have been implicated in chronic inflammatory disorders. Type 2 diabetes mellitus (T2DM) and obesity are two metabolic disorders that are linked together and have become an epidemic worldwide, thus raising significant public health concerns. Fibroblast growth factor 21 (FGF21) is an endocrine hormone with pleiotropic metabolic effects that increase insulin sensitivity and energy expenditure by elevating thermogenesis in brown or beige adipocytes, thus reducing body weight and sugar intake. In contrast, during starvation conditions, FGF21 induces its expression in the liver to initiate glucose homeostasis. Insulin resistance is one of the main anomalies caused by impaired FGF21 signaling, which also causes abnormal regulation of other signaling pathways. Tumor necrosis factor alpha (TNF-α), the cytokine released by adipocytes and inflammatory cells in response to chronic inflammation, is regarded major factor that reduces the expression of FGF21 and modulates underlying insulin resistance that causes imbalanced glucose homeostasis. This review aims to shed light on the mechanisms underlying the development of insulin resistance in obese individuals as well as the fundamental flaw in type 2 diabetes, which is malfunctioning obese adipose tissue.
Collapse
Affiliation(s)
- Unnati Jain
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India
| | - Priyanka Srivastava
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India
| | - Ashwani Sharma
- Insight BioSolutions, Rue Joseph Colin, 35000 Rennes, France
| | - Subrata Sinha
- Centre of Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh, Assam 786004, India
| | - Surabhi Johari
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
31
|
Yang Y, Ma X, Li Y, Jin L, Zhou X. The evolving tumor-associated adipose tissue microenvironment in breast cancer: from cancer initiation to metastatic outgrowth. Clin Transl Oncol 2024:10.1007/s12094-024-03831-8. [PMID: 39720985 DOI: 10.1007/s12094-024-03831-8] [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: 11/12/2024] [Accepted: 12/09/2024] [Indexed: 12/26/2024]
Abstract
Adipocytes represent a significant proportion of breast tissue, comprising between 3.7 and 37% of stromal tissue. They play a pivotal role in metabolic regulation, energy supply, metabolic regulation, support effects, and cytokine release within the breast. In breast cancer (BC) tissue, adipocytes engage in intricate crosstalk with BC cells, playing a key role in tumor proliferation, invasion, metastasis formation, and metabolic remodeling. This is due to the provision of hormones, adipokines, and fatty acids to tumor cells by the adipocytes. With the initiation of metastatic outgrowth of BC, the peritumoral adipose tissue exhibits abundant and intricate changes based on its original construction and function, which convert it into a tumor-associated adipose tissue microenvironment (TAAME). It includes some specific adipocytes: adipose-derived stem cells (ASCs), cancer-associated adipocytes (CAAs), adipocyte-derived fibroblasts (ADFs), etc. From a mechanistic standpoint, specific adipocytes can facilitate the proliferation, invasion, metastasis, and angiogenesis of BC cells by secreting a multitude of cytokines (IL-6) and adipokines (leptin), which collectively create an environment conducive to BC progression. It is of paramount importance to recognize the TAAME as a crucial target for the diagnosis, treatment, and drug resistance of BC. Consequently, the review presents an overview of the characteristics and interactions of specific adipocytes within TAAME cell populations. This will facilitate the development of more effective personalized therapies against BC progression, relapse, and metastasis.
Collapse
Affiliation(s)
- Yang Yang
- College of Life Science, Northeast Forestry University, Harbin, 150000, China
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, 133000, China
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
- Zhejiang Orient Gene Biotech Co., Ltd, Huzhou, 313300, China
| | - Xiao Ma
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, 133000, China
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Yue Li
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, 133000, China
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Lihua Jin
- College of Life Science, Northeast Forestry University, Harbin, 150000, China
| | - Xianchun Zhou
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, 133000, China.
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, 133000, China.
- Central Laboratory, Yanbian University Hospital, Ju Zi Road No.1327, Yanji, 133002, China.
| |
Collapse
|
32
|
Xiao H, Xing T, Qiu M, Zhang G, Yang G, Chen W, Hu D, Xue D, Peng J, Du B. Adiponectin deficiency prevents chronic colitis-associated colonic fibrosis via inhibiting CXCL13 production. J Adv Res 2024:S2090-1232(24)00610-6. [PMID: 39725008 DOI: 10.1016/j.jare.2024.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
INTRODUCTION Colonic fibrosis is a long-term complication of inflammatory bowel disease (IBD), often leading to functional impairment, intestinal obstruction, and surgery. Adiponectin (APN) is an adipokine derived from adipocytes that plays a pleiotropic role in fibrosis regulation, depending on tissue and cell type specific or disease context, but its role in colonic fibrosis remains unclear. OBJECTIVE To explore the role and involved mechanism of APN in chronic colitis-associated colonic fibrosis. METHODS Studies were performed in GEO database, colonic tissues of UC patients, dextran sulfate sodium (DSS)-induced colonic fibrosis in male wild-type (WT) and APN-/- mice, mouse L929 and human CCD-18Co fibroblasts treated with recombinant CXCL13 protein, and colonic fibrosis in WT mice infected with shRNA of CXCL13. RESULTS APN was highly expressed in the colonic tissues of UC patients and positively correlated with the colonoscopy score and colonic fibrosis markers COL1A1 and COL3A1. APN deficiency significantly improved chronic colitis-induced colonic fibrosis in mice with down-regulating collagenase accumulation and expressions of TGF-β, α-SMA, COL1A1, COL3A1, and MMP-9 in colonic tissues. Transcriptomics showed that APN deficiency mainly affected cytokine-cytokine receptor interactions, especially CXCL13 signaling. Follow-up studies showed that APN deficiency significantly decreased the number of colonic F4/80+CD206+CXCL13+ macrophages by weakening Akt phosphorylation. Additional experiments confirmed that CXCL13 notably increased the expressions of α-SMA and COL1A1 in mouse and human fibroblasts by activating p-Akt, p-p38, p-ERK, and p-JNK. Moreover, inhibiting CXCL13 with shRNA significantly ameliorated colonic fibrosis in mice with DSS-induced chronic colitis. Immunohistochemistry analysis revealed high expression of CXCL13 in the colon tissues of patients with UC, showing a positive correlation with APN, COL1A1, and COL3A1. CONCLUSION APN contributes to the progression of colonic fibrosis and can exacerbate this condition by regulating the secretion of CXCL13 in the colon, offering potential new perspectives on the pathophysiology of colonic fibrosis.
Collapse
Affiliation(s)
- Haitao Xiao
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Tianhang Xing
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China; Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China; Hebei Key Laboratory of Natural Products Activity Components and Function, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei 066004, China
| | - Miao Qiu
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Guangtao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Gongli Yang
- Department of Gastroenterology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Wenke Chen
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Die Hu
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Deao Xue
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Jiao Peng
- Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China.
| | - Bin Du
- Hebei Key Laboratory of Natural Products Activity Components and Function, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei 066004, China.
| |
Collapse
|
33
|
Papakonstantinou I, Tsioufis K, Katsi V. Spotlight on the Mechanism of Action of Semaglutide. Curr Issues Mol Biol 2024; 46:14514-14541. [PMID: 39728000 DOI: 10.3390/cimb46120872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024] Open
Abstract
Initially intended to control blood glucose levels in patients with type 2 diabetes, semaglutide, a potent glucagon-like peptide 1 analogue, has been established as an effective weight loss treatment by controlling appetite. Integrating the latest clinical trials, semaglutide in patients with or without diabetes presents significant therapeutic efficacy in ameliorating cardiometabolic risk factors and physical functioning, independent of body weight reduction. Semaglutide may modulate adipose tissue browning, which enhances human metabolism and exhibits possible benefits in skeletal muscle degeneration, accelerated by obesity and ageing. This may be attributed to anti-inflammatory, mitochondrial biogenesis, antioxidant and autophagy-regulating effects. However, most of the supporting evidence on the mechanistic actions of semaglutide is preclinical, demonstrated in rodents and not actually confirmed in humans, therefore warranting caution in the interpretation. This article aims to explore potential innovative molecular mechanisms of semaglutide action in restoring the balance of several interlinking aspects of metabolism, pointing to distinct functions in inflammation and oxidative stress in insulin-sensitive musculoskeletal and adipose tissues. Moreover, possible applications in protection from infections and anti-aging properties are discussed. Semaglutide enhancement of the core molecular mechanisms involved in the progress of obesity and diabetes, although mostly preclinical, may provide a framework for future research applications in human diseases overall.
Collapse
Affiliation(s)
- Ilias Papakonstantinou
- 4th Department of Internal Medicine, Evangelismos General Hospital, 10676 Athens, Greece
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 11527 Athens, Greece
| | - Vasiliki Katsi
- 1st Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 11527 Athens, Greece
| |
Collapse
|
34
|
Ghosh A, Chénier I, Leung YH, Oppong AK, Peyot ML, Madiraju SRM, Al-Khairi I, Abubaker J, Al-Mulla F, Prentki M, Abu-Farha M. Adipocyte Angptl8 deletion improves glucose and energy metabolism and obesity associated inflammation in mice. iScience 2024; 27:111292. [PMID: 39640567 PMCID: PMC11617963 DOI: 10.1016/j.isci.2024.111292] [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: 04/08/2024] [Revised: 08/28/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Angiopoietin-like protein 8 (Angptl8), expressed in the liver and adipocytes, forms a complex with Angptl3 or Angptl4, which regulates lipoprotein lipase and triglyceride metabolism. However, the precise functions of adipocyte Angptl8 remain elusive. Here we report that adipocyte-specific inducible Angptl8-knockout (AT-A8-KO) male mice on normal diet showed minor phenotypic changes, but after a high-fat high fructose (HFHF) diet, exhibited decreased body weight gain and glycemia, elevated rectal temperature and early dark phase energy expenditure compared to the Cre controls. AT-A8-KO mice also displayed improved glucose tolerance, a trend for better insulin sensitivity, improved insulin-stimulated glucose uptake in adipose tissues, and reduced visceral adipose tissue crown-like structures, plasma MCP-1 and leptin levels. The results indicate the importance of adipose Angptl8 in the context of nutri-stress and obesity, as its deletion in mice promotes a metabolically healthy obese phenotype by slightly ameliorating obesity, improving glucose and energy homeostasis, and mitigating inflammation.
Collapse
Affiliation(s)
- Anindya Ghosh
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche Du Centre Hospitalier de L’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Isabelle Chénier
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche Du Centre Hospitalier de L’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Yat Hei Leung
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche Du Centre Hospitalier de L’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Abel K. Oppong
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche Du Centre Hospitalier de L’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Marie-Line Peyot
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche Du Centre Hospitalier de L’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - S. R. Murthy Madiraju
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche Du Centre Hospitalier de L’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Irina Al-Khairi
- Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Jehad Abubaker
- Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Translational Research Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Marc Prentki
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche Du Centre Hospitalier de L’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Mohamed Abu-Farha
- Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
- Translational Research Department, Dasman Diabetes Institute, Dasman 15462, Kuwait
| |
Collapse
|
35
|
Boisclair YR, Giesy SL. Endocrine adaptations to demanding physiological states in ruminants. J Dairy Sci 2024:S0022-0302(24)01377-8. [PMID: 39647618 DOI: 10.3168/jds.2024-25799] [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: 09/30/2024] [Accepted: 11/11/2024] [Indexed: 12/10/2024]
Abstract
Highly productive ruminants rely on hormonally driven adaptations to prioritize the use of limiting nutrients during the demanding phases of the pregnancy-lactation cycle. Glucose, the predominant oxidative fuel of fetal life and the absolute precursor of mammary lactose synthesis, illustrates the need and benefit of such adaptations. Endocrine mechanisms such as insulin resistance and/or hypoinsulinemia favor the diversion of maternal glucose to the placenta or mammary gland where uptake is independent of insulin. Research in dairy cows in the 1980s and 1990s identified growth hormone as a peripherally acting signal opposing the effects of insulin. The following decades have seen the discovery of a new generation of signals secreted almost exclusively by adipose tissue, skeletal muscle or liver, dynamically regulated by metabolic challenges, and engaged in cross-organ communication. The understanding of these signals in the coordination of metabolism in ruminants has been limited by the availability of assays to measure their circulating concentrations and materials to perform functional studies. Nevertheless, emerging data point to their importance during demanding physiological states in ruminants, including early lactation in dairy cows and late pregnancy in sheep. Examples include modulation of insulin action by liver-derived fibroblast growth factor 21 (FGF21) and regulation of energy allocation among tissues by the action of the adipose-derived hormone leptin via its ability to control the hypothalamic-pituitary-thyroid axis. Recent studies investigating the regulation and action of FGF21 and leptin in dairy cows and sheep will be used to illustrate the potential of recently discovered signals to coordinate metabolism during physiologically demanding states such as early lactation.
Collapse
Affiliation(s)
- Y R Boisclair
- Department of Animal Science, Cornell University, Ithaca, New York 14853.
| | - S L Giesy
- Department of Animal Science, Cornell University, Ithaca, New York 14853
| |
Collapse
|
36
|
Nhoek P, Hwang S, Huh J, Pel P, Park CW, Khiev P, Kim HW, Noh M, Chin YW. Butanolides and clerodane diterpenes from the twigs of Casearia grewiifolia and their effects on adiponectin secretion. Bioorg Chem 2024; 153:107890. [PMID: 39427630 DOI: 10.1016/j.bioorg.2024.107890] [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: 08/17/2024] [Revised: 10/07/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024]
Abstract
Three butanolides derivatives, grewiifolides A-C, and nine clerodane diterpenes, grewiifolins M-U, as well as a known sterol were isolated from the twigs of Casearia grewiifolia. The chemical structures and configurations of all isolates were established by various spectroscopic means and chemical derivatization. In a cell-based phenotypic assay using the adipogenesis model of human bone marrow mesenchymal stem cells (hBM-MSCs), grewiifolide B significantly promoted adiponectin-secretion with EC50 value of 24.8 µM. In target identification studies, butanolide derivatives were selectively bound to PPARγ with Ki values of 4.65, 0.55, and 17.8 µM, respectively. Further functional analysis and molecular modeling revealed that grewiifolide B promotes adiponectin-secretion through PPARγ full agonism.
Collapse
Affiliation(s)
- Piseth Nhoek
- Natural Product Research Institute and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seokyoung Hwang
- Natural Product Research Institute and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jungmoo Huh
- Natural Product Research Institute and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Pisey Pel
- Natural Product Research Institute and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Chan-Woong Park
- Natural Product Research Institute and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Piseth Khiev
- Royal University of Phnom Penh, Department of Biology, Russian Federation Boulevard, Khan Toul Kork, Phnom Penh 12156, Cambodia
| | - Hyun Woo Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Gyeonggi-do 04620, Republic of Korea
| | - Minsoo Noh
- Natural Product Research Institute and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Young-Won Chin
- Natural Product Research Institute and Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
37
|
Oster H. Adipose endothelin signaling-An unusual suspect linking obesity to insulin resistance. Acta Physiol (Oxf) 2024; 240:e14241. [PMID: 39377169 DOI: 10.1111/apha.14241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024]
Affiliation(s)
- Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
- Center of Brain, Behavior & Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| |
Collapse
|
38
|
Galigniana NM, Ruiz MC, Piwien-Pilipuk G. FK506 binding protein 51: Its role in the adipose organ and beyond. J Cell Biochem 2024; 125:e30351. [PMID: 36502528 DOI: 10.1002/jcb.30351] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/04/2022] [Accepted: 11/15/2022] [Indexed: 02/17/2024]
Abstract
There is a great body of evidence that the adipose organ plays a central role in the control not only of energy balance, but importantly, in the maintenance of metabolic homeostasis. Interest in the study of different aspects of its physiology grew in the last decades due to the pandemic of obesity and the consequences of metabolic syndrome. It was not until recently that the first evidence for the role of the high molecular weight immunophilin FK506 binding protein (FKBP) 51 in the process of adipocyte differentiation have been described. Since then, many new facets have been discovered of this stress-responsive FKBP51 as a central node for precise coordination of many cell functions, as shown for nuclear steroid receptors, autophagy, signaling pathways as Akt, p38 MAPK, and GSK3, as well as for insulin signaling and the control of glucose homeostasis. Thus, the aim of this review is to integrate and discuss the recent advances in the understanding of the many roles of FKBP51 in the adipose organ.
Collapse
Affiliation(s)
- Natalia M Galigniana
- Laboratory of Nuclear Architecture, Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marina C Ruiz
- Laboratory of Nuclear Architecture, Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina
| | - Graciela Piwien-Pilipuk
- Laboratory of Nuclear Architecture, Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina
| |
Collapse
|
39
|
Zhang T, Liu J, Liu X, Wang Q, Zhang H. The causal impact of gut microbiota on circulating adipokine concentrations: a two-sample Mendelian randomization study. Hormones (Athens) 2024; 23:789-799. [PMID: 38564143 DOI: 10.1007/s42000-024-00553-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE Evidence from previous experimental and observational research demonstrates that the gut microbiota is related to circulating adipokine concentrations. Nevertheless, the debate as to whether gut microbiome composition causally influences circulating adipokine concentrations remains unresolved. This study aimed to take an essential step in elucidating this issue. METHODS We used two-sample Mendelian randomization (MR) to causally analyze genetic variation statistics for gut microbiota and four adipokines (including adiponectin, leptin, soluble leptin receptor [sOB-R], and plasminogen activator inhibitor-1 [PAI-1]) from large-scale genome-wide association studies (GWAS) datasets. A range of sensitivity analyses was also conducted to assess the stability and reliability of the results. RESULTS The composite results of the MR and sensitivity analyses revealed 22 significant causal associations. In particular, there is a suggestive causality between the family Clostridiaceae1 (IVW: β = 0.063, P = 0.034), the genus Butyrivibrio (IVW: β = 0.029, P = 0.031), and the family Alcaligenaceae (IVW: β=-0.070, P = 0.014) and adiponectin. Stronger causal effects with leptin were found for the genus Enterorhabdus (IVW: β=-0.073, P = 0.038) and the genus Lachnospiraceae (NK4A136 group) (IVW: β=-0.076, P = 0.01). Eight candidate bacterial groups were found to be associated with sOB-R, with the phylum Firmicutes (IVW: β = 0.235, P = 0.03) and the order Clostridiales (IVW: β = 0.267, P = 0.028) being of more interest. In addition, the genus Roseburia (IVW: β = 0.953, P = 0.022) and the order Lactobacillales (IVW: β=-0.806, P = 0.042) were suggestive of an association with PAI-1. CONCLUSION This study reveals a causal relationship between the gut microbiota and circulating adipokines and may help to offer novel insights into the prevention of abnormal concentrations of circulating adipokines and obesity-related diseases.
Collapse
Affiliation(s)
- Tongxin Zhang
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Jingyu Liu
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Xiao Liu
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Qian Wang
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China.
| | - Huawei Zhang
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
40
|
Li L, Zhao S, Leng Z, Chen S, Shi Y, Shi L, Li J, Mao K, Tang H, Meng B, Wang Y, Shang G, Liu H. Pathological mechanisms and related markers of steroid-induced osteonecrosis of the femoral head. Ann Med 2024; 56:2416070. [PMID: 39529511 PMCID: PMC11559024 DOI: 10.1080/07853890.2024.2416070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Osteonecrosis of the femoral head (ONFH) is a refractory orthopedic disease with a high disability rate. Long-term administration of steroids is the most common pathogenic factor for non-traumatic ONFH. Early diagnosis of steroid-induced osteonecrosis of the femoral head (SONFH) is difficult and mainly depends on imaging. OBJECTIVES The objectives of this review were to examine the pathological mechanisms of SONFH, summarize related markers of SONFH, and identify areas for future studies. METHODS We reviewed studies on pathological mechanisms and related markers of SONFH and discussed the relationship between them, as well as clinical applications and the outlook of potential markers. RESULTS The pathological mechanisms of SONFH included decreased osteogenesis, lipid accumulation, increased intraosseous pressure, and microcirculation disruption. Differential proteomics and genomics play crucial roles in the occurrence, progression, and outcome of SONFH, providing novel insights into SONFH. Additionally, the biological functions of mesenchymal stem cells (MSCs) and exosomes (Exos) in SONFH have attracted increasing attention. CONCLUSIONS The pathological mechanisms of SONFH are complex. The related markers mentioned in the current review can predict the occurrence and progression of SONFH, which will help provide effective early clinical prevention and treatment strategies for SONFH.
Collapse
Affiliation(s)
- Longyu Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shangkun Zhao
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zikuan Leng
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yifang Shi
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijun Shi
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinfeng Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Keya Mao
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, China
| | - Hai Tang
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bin Meng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yisheng Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guowei Shang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
41
|
Su S, Tian R, Jiao Y, Zheng S, Liang S, Liu T, Tian Z, Cao X, Xing Y, Ma C, Ni P, Yu F, Jiang T, Wang J. Ubiquitination and deubiquitination: Implications for the pathogenesis and treatment of osteoarthritis. J Orthop Translat 2024; 49:156-166. [PMID: 40226783 PMCID: PMC11993839 DOI: 10.1016/j.jot.2024.09.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 01/12/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative disease that affects multiple cells and associated extracellular matrix (ECM). Chondrocytes and chondroextracellular matrix together constitute articular cartilage tissue. Any factors that affect the activity of chondrocytes and destroy the metabolic balance of the chondrocyte ECM will lead to the inability of articular cartilage to perform normal functions. The articular subchondral bone and articular cartilage must be coordinated to resist enough friction and mechanical stress, so the articular subchondral bone lesion will aggravate the articular cartilage defect and vice versa. Synoviocytes, including fibroblast-like synoviocytes (FLSs) and synovial macrophages at the joint, are also important factors that cause low-grade chronic progressive inflammation of OA. Regulation of phenotype transformation of synovial macrophages has become another possible target for the clinical treatment of OA. Ubiquitination and deubiquitination are the main post-translational protein modification pathways in the human body, which are widely involved in multiple signaling pathways and physiological processes. Naturally, they also play a very important regulatory role in the occurrence and development of OA. These effects are summarized in this review, including (A) regulating the aging and apoptosis of chondrocytes, FLSs and osteoblasts; (B) regulation of ECM degradation; (C) regulation of macrophage phenotypic transformation; (D) modulation of skeletal muscle and adipose tissues. Ubiquitination targeting drugs for OA treatment are also listed. Depending on the high efficiency of ubiquitination and deubiquitination, understanding OA-related ubiquitination pathways can help design more efficient drugs to treat OA and provide more potential targets for clinical treatment. The Translational Potential of This Article. In this paper, the ubiquitination-related pathways in osteoarthritis (OA), including aging, apoptosis and autophagy in chondrocytes, osteoblasts, FLSs and macrophages were investigated. In particular, several ubiquitination-related targets are expected to be effective approaches for OA clinical treatment. In addition, in the process of OA occurrence and development, the complex relationship between the local joint area and other tissues including skeletal muscle and adipose tissue is also discussed. These myokines and adipokines from musculoskeletal tissues are all expected to become efficient targets for OA treatment apart from the joint itself. In addition, those myokines secreted by cardiovascular tissues would show potential therapeutic effects as well. What if altering the contents for these ubiquitination-related targets in the serum through exercise will provide a new idea for OA therapy or prevent OA from deteriorating continuously?
Collapse
Affiliation(s)
- Shibo Su
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Hainan Provincial Stem Cell Research Institute, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, 571199, China
| | - Ruijiao Tian
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Hainan Provincial Stem Cell Research Institute, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, 571199, China
| | - Yang Jiao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Shudan Zheng
- Plastic Surgery, Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, China
| | - Siqiang Liang
- Zhongke Comprehensive Medical Transformation Center Research Institute (Hainan) Co., Ltd, Haikou, 571199, China
| | - Tianyi Liu
- Department of Pharmacology, Zibo Hospital of Traditional Chinese Medicine, Zibo, 255300, China
| | - Ziheng Tian
- School of Clinical Medicine, Jining Medical University, Jining, 272002, China
| | - Xiuhong Cao
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, 571199, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, 571199, China
| | - Yanlong Xing
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, 571199, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, 571199, China
| | - Chuqing Ma
- The Second Clinical College, Hainan Medical University, Haikou, 571199, China
| | - Panli Ni
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Hainan Provincial Stem Cell Research Institute, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, 571199, China
| | - Fabiao Yu
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, 571199, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, 571199, China
| | - Tongmeng Jiang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Hainan Provincial Stem Cell Research Institute, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, 571199, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, 571199, China
| | - Juan Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Hainan Provincial Stem Cell Research Institute, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, 571199, China
| |
Collapse
|
42
|
Peng Z, Zeng Y, Zeng X, Tan Q, He Q, Wang S, Wang J. 6-Gingerol improves lipid metabolism disorders in skeletal muscle by regulating AdipoR1/AMPK signaling pathway. Biomed Pharmacother 2024; 180:117462. [PMID: 39316973 DOI: 10.1016/j.biopha.2024.117462] [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: 07/21/2024] [Revised: 09/08/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND To delve into the precise mechanisms by which 6-gingerol ameliorates lipid metabolism disorders in skeletal muscle. METHODS The level of triglycerides (TG) was used to evaluate lipid deposition. In skeletal muscle, transmission electron microscopy (TEM) was employed to observe mitochondrial morphology. Additionally, PCR was applied to detect mitochondrial biogenesis, and levels of malondialdehyde (MDA), catalase (CAT), glutathione, r-glutamyl cysteingl+glycine (GSH) and nicotinamide adenine dinucleotide (NADH) were measured to assess mitochondrial oxidative stress levels. In vivo, flow cytometry and immunofluorescence assays were conducted to quantify reactive oxygen species (ROS) and mitochondrial membrane potential (MMP). Furthermore, the Seahorse XF assays was utilized to assess mitochondrial respiratory capacity. Fluorescence confocal microscopy and molecular docking were applied to analyze the binding of 6-gingerol and adiponectin receptor 1 (AdipoR1). The expression of AdipoR1, AMPK, PGC-1α and SIRT1 were detected by Western Blot. RESULTS In vivo, 6-gingerol could reduce body weight in mice induced by a high-fat diet, enhance metabolic profiles in plasma, decrease lipid accumulation in skeletal muscle and liver, and elevate adiponectin levels. In skeletal muscle, it could restore mitochondrial morphology, boost mitochondrial copy number and biogenesis, and mitigate oxidative stress. In vitro, 6-gingerol may directly interact with AdipoR1 to upregulate the expression of downstream proteins p-AMPK, SIRT1, and PGC-1α, leading to a reduction in lipid deposition, a decrease in ROS production, an increase in mitochondrial membrane potential, and an enhancement of mitochondrial respiratory capacity in C2C12 myotubes. CONCLUSION 6-Gingerol ameliorated lipid metabolism in skeletal muscle by regulating the AdipoR1/AMPK signaling pathway.
Collapse
Affiliation(s)
- Ze Peng
- Chongqing University of Chinese Medicine, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China; Chongqing Medical University, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China
| | - Yan Zeng
- Chongqing Medical University, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China
| | - Xin Zeng
- Chongqing University of Chinese Medicine, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China; Chongqing Medical University, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China
| | - Qi Tan
- Chongqing Medical University, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China
| | - Qifeng He
- Chongqing Medical University, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China
| | - Shang Wang
- Chongqing University of Chinese Medicine, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China; Chongqing Medical University, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China.
| | - Jianwei Wang
- Chongqing University of Chinese Medicine, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China; Chongqing Medical University, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China.
| |
Collapse
|
43
|
Xiao Q, Tang L, Chen S, Mei Y, Wang C, Yang J, Shang J, Li S, Wang W. Two-Pronged Attack: Dual Activation of Fat Reduction Using Near-Infrared-Responsive Nanosandwich for Targeted Anti-Obesity Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406985. [PMID: 39324577 DOI: 10.1002/advs.202406985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/06/2024] [Indexed: 09/27/2024]
Abstract
Excessive fat accumulation and chronic inflammation are two typical characteristics of obesity. AMP-activated protein kinase (AMPK), a master regulator of energy metabolism, is involved in adipogenesis, lipogenesis, and inflammation modulation in adipose tissue (AT). Thus, effective lipid reduction and anti-inflammation through AMPK regulation play vital roles in treating obesity. Herein, an anti-obesity nanosandwich is fabricated through attaching polymetformin (PolyMet) onto photothermal agent black phosphorus nanosheets (BP). PolyMet activates AMPK to inhibit adipogenesis, promote browning, and mitigate AT inflammation by decreasing macrophage infiltration, repolarizing macrophage phenotype, and downregulating pro-inflammatory cytokines. Additionally, BP induces lipolysis and apoptosis of adipocytes and macrophages through a photothermal effect. By further functionalization using hyaluronic acid (HA) and MMP2 substrate-linking P3 peptide-modified HA (P3-HA), an enhanced anti-obesity effect is obtained by dual-targeting of P3 and HA, and HA-mediated CD44 poly-clustering after MMP2 cleavage. Upon laser irradiation, the designed nanosandwich (P3-HA/PM@BP) effectively inhibits obesity development in obese mice, increases M2/M1 ratio in AT, reduces the serum levels of cholesterol/triglyceride and improves insulin sensitivity, exhibiting promising research potential to facilitate the clinical development of modern anti-obesity therapies.
Collapse
Affiliation(s)
- Qiaqia Xiao
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Lu Tang
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Siying Chen
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Yijun Mei
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Chuying Wang
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Jing Yang
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Jing Shang
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Shengliang Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing, 211198, P. R. China
| |
Collapse
|
44
|
Prieur X, Cao L. Precision medicine: toward restoring fat with gene therapy in inherited lipodystrophy. Gene Ther 2024; 31:560-562. [PMID: 39317737 DOI: 10.1038/s41434-024-00489-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Affiliation(s)
- Xavier Prieur
- l'Institut du Thorax, INSERM, CNRS, Nantes Université, CHU Nantes, Nantes, France.
| | - Lei Cao
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA.
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
45
|
Tang J, Lei Y, Pignalosa A, Hsu HH, Abdul-Sater AA, Sweeney G. Development of a non-invasive bioassay for adiponectin target engagement in mice. iScience 2024; 27:110994. [PMID: 39435143 PMCID: PMC11492082 DOI: 10.1016/j.isci.2024.110994] [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: 06/11/2024] [Revised: 07/23/2024] [Accepted: 09/16/2024] [Indexed: 10/23/2024] Open
Abstract
Adiponectin-based therapeutic strategies are promising for managing metabolic diseases and reducing inflammation, prompting the development of adiponectin receptor agonists. However, monitoring their pharmacodynamic actions in clinical applications is challenging. This study aimed to identify peripheral biomarkers to monitor adiponectin actions using ALY688, an adiponectin receptor agonist peptide. RNA sequencing analysis of whole blood identified a cluster of genes that were significantly increased in the ALY688-treated group compared to the control. This gene cluster was validated by qPCR and further confirmed in human peripheral blood mononuclear cells treated with ALY688 ex vivo. We also confirmed a functional outcome of ALY688 action in mice as our study also demonstrated the anti-inflammatory effect of ALY688 in a sublethal LPS mouse model. In summary, a newly identified gene cluster signature is suitable for assessing the pharmacodynamic action of adiponectin or its mimetics in blood samples.
Collapse
Affiliation(s)
- Jialing Tang
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Yubin Lei
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Angelica Pignalosa
- Allysta Pharmaceuticals Inc., 500 108th Avenue NE, Suite 1100, Bellevue, WA 98004, USA
| | - Henry H. Hsu
- Allysta Pharmaceuticals Inc., 500 108th Avenue NE, Suite 1100, Bellevue, WA 98004, USA
| | - Ali A. Abdul-Sater
- School of Kinesiology and Health Science, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Gary Sweeney
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
46
|
Wu X, Tian Y, Zhang N, Ren Y, Zhang Z, Zhao Y, Guo Y, Gong Y, Zhang Y, Li D, Li H, Jiang R, Li G, Liu X, Kang X, Tian Y. The role of AdipoQ on proliferation, apoptosis, and hormone Secretion in chicken primary adenohypophysis cells. Poult Sci 2024; 103:104137. [PMID: 39142032 PMCID: PMC11379664 DOI: 10.1016/j.psj.2024.104137] [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: 04/01/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 08/16/2024] Open
Abstract
Adiponectin (AdipoQ), an adipokine secreted by adipocytes, has been reported to exist widely in various cell types and tissues, including the adenohypophysis of chickens. However, the molecular mechanism by which AdipoQ regulates the function of chicken adenohypophysis remains elusive. In this study, we investigated the effects of AdipoQ on proliferation, apoptosis, secretion of related hormones (FSH, LH, TSH, GH, PRL and ACTH) and expression of related genes (FSHβ, LHβ, GnRHR, TSHβ, GH, PRL and ACTH) in primary adenohypophysis cells of chickens by using real-time fluorescent quantitative PCR (RT-qPCR), cell counting kit-8 (CCK-8), flow cytometry, enzyme-linked immunosorbent assay (ELISA) and Western blot (WB) assays. Our results showed that AdipoQ promoted the proliferation of chicken primary adenohypophysis cells, up-regulated the mRNA expression of proliferation-related genes CDK1, PCNA, CCND1 and P21 (P < 0.05), as well as the increased protein expression of CDK1 and PCNA (P < 0.05). Furthermore, AdipoQ inhibited apoptosis of chicken primary adenohypophysis cells, resulting in down-regulation of pro-apoptotic genes Caspase3, Fas, and FasL mRNA expression, and decreased Caspase3 protein expression (P < 0.05). Moreover, there was an up-regulation of anti-apoptotic gene Bcl2 mRNA and protein expression (P < 0.05). Additionally, AdipoQ suppressed the secretion of FSH, LH, TSH, GH, PRL, and ACTH (P < 0.05), as well as the mRNA expression levels of related genes (P < 0.05). Treatment with AdipoRon (a synthetic substitute for AdipoQ) and co-treatment with RNA interference targeting AdipoQ receptors 1/2 (AdipoR1/2) had no effect on the secretion of FSH, LH, TSH, GH, PRL, and ACTH, as well as the mRNA expression levels of the related genes. This suggests that AdipoQ's regulation of hormone secretion and related gene expression is mediated by the AdipoR1/2 signaling axis. Importantly, we further demonstrated that the mechanism of AdipoQ on FSH, LH, TSH and GH secretion is realized through AMPK signaling pathway. In conclusion, we have revealed, for the first time the molecular mechanism by which AdipoQ regulates hormone secretion in chicken primary adenohypophysis cells.
Collapse
Affiliation(s)
- Xing Wu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yixiang Tian
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Na Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yangguang Ren
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Zihao Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yudian Zhao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yulong Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yujie Gong
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yanhua Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Donghua Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Hong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Ruirui Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Guoxi Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xiaojun Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China.
| |
Collapse
|
47
|
Wu W, Jian Y, Yuan S, Li X, Tang Y, Zeng F, Liu W, Zhao Z, Wang Y, Wang Y, Liu W. Exercise-promoted adiponectin secretion activates autolysosomes to protect the liver of ApoE -/- mice from a high-fat diet. Food Funct 2024; 15:9796-9812. [PMID: 39229645 DOI: 10.1039/d4fo02984d] [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: 09/05/2024]
Abstract
Fat is a "double-edged sword": while it is a necessary substance for the body, the long-term intake of excessive fat will cause obesity, with the liver subjected to lipotoxicity as it accumulates. It will then continue to deteriorate, eventually leading to liver failure, which is a negative impact of high-fat food intake. Research has shown that exercise can reverse the side effects of a chronic high-fat diet and help the body to mitigate the harmful effects of lipotoxicity. In our study, it was found that moderate-intensity cardio-training (MICT) and high-intensity interval exercise (HIIT) effectively protected the livers of high-fat diet (HFD) ApoE-/- mice against lipotoxicity. Previous results demonstrated that 12 weeks of HFD resulted in a significant elevation of CD36 in the livers of C57BL/6J mice, while knockdown of CD36 did not reduce the accumulation of fat in the liver. Therefore, we used ApoE-/- mice as experimental subjects. Although HFD caused the development of hyperlipidemia and atherosclerosis, it is interesting to note that, due to the knockdown of ApoE, the livers of ApoE-/- mice in the non-exercise group did not show significant lipid deposition; however, after 12 weeks of MICT and HIIT, the livers of ApoE-/- mice showed significant lipid deposition. After we analyzed the lipid metabolism in their livers, we found that this was caused by the promotion of transport of peripheral fat into the liver due to exercise. Moreover, 12 weeks of exercise effectively reduced atherosclerosis, and the livers of ApoE-/- mice in the exercise group were not damaged by lipotoxicity. The results showed that a 12-week exercise treatment activated AMPK in the livers of HFD ApoE-/- mice through the APN-AdipoR1 signaling pathway, improved hepatic lipid metabolism disorders, and promoted the nuclear translocation of TFEB to enhance autophagic-lysosomal lipid scavenging. After the peripheral lipid is input into the liver due to exercise, the energy generated through gluconeogenesis can be used to replenish the energy consumed by exercise and maintain the normal operation of various functions in the liver, based on which the high autophagic flux in the liver can be maintained and the lipid clearance rate can be enhanced to protect the liver from lipotoxicity.
Collapse
Affiliation(s)
- Weijia Wu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Ye Jian
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Shunling Yuan
- Yangtze University, College of Arts and Sciences, Jingzhou 434020, China
| | - Xuan Li
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Yingzhe Tang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Fanqi Zeng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Wenjing Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Zhe Zhao
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Yirong Wang
- Hunan Sports Vocational College, Changsha 410019, China
| | - Yiyang Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| |
Collapse
|
48
|
Odimegwu CL, Uwaezuoke SN, Chikani UN, Mbanefo NR, Adiele KD, Nwolisa CE, Eneh CI, Ndiokwelu CO, Okpala SC, Ogbuka FN, Odo KE, Ohuche IO, Obiora-Izuka CE. Targeting the Epigenetic Marks in Type 2 Diabetes Mellitus: Will Epigenetic Therapy Be a Valuable Adjunct to Pharmacotherapy? Diabetes Metab Syndr Obes 2024; 17:3557-3576. [PMID: 39323929 PMCID: PMC11423826 DOI: 10.2147/dmso.s479077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/03/2024] [Indexed: 09/27/2024] Open
Abstract
Although genetic, environmental, and lifestyle factors largely contribute to type 2 diabetes mellitus (T2DM) risk, the role of epigenetics in its pathogenesis is now well established. The epigenetic mechanisms in T2DM mainly consist of DNA methylation, histone modifications and regulation by noncoding RNAs (ncRNAs). For instance, DNA methylation at CpG islands in the promoter regions of specific genes encoding insulin signaling and glucose metabolism suppresses these genes. Modulating the enzyme mediators of these epigenetic marks aims to restore standard gene expression patterns and improve glycemic control. In targeting these epigenetic marks, using epigenetic drugs such as DNA methyltransferase (DNAMT), histone deacetylase (HDAC) and histone acetyltransferase (HAT) inhibitors has led to variable success in humans and experimental murine models. Specifically, the United States' Food and Drug Administration (US FDA) has approved DNAMT inhibitors like 5-azacytidine and 5-aza-2'-deoxycytidine for use in diabetic retinopathy: a T2DM microvascular complication. These DNAMT inhibitors block the genes for methylation of mitochondrial superoxide dismutase 2 (SOD2) and matrix metallopeptidase 9 (MMP-9): the epigenetic marks in diabetic retinopathy. Traditional pharmacotherapy with metformin also have epigenetic effects in T2DM and positively alter disease outcomes when combined with epigenetic drugs like DNAMT and HDAC inhibitors, raising the prospect of using epigenetic therapy as a valuable adjunct to pharmacotherapy. However, introducing small interfering RNAs (siRNAs) in cells to silence specific target genes remains in the exploratory phase. Future research should focus on regulating gene expression in T2DM using long noncoding RNA (lncRNA) molecules, another type of ncRNA. This review discusses the epigenetics of T2DM and that of its macro- and microvascular complications, and the potential benefits of combining epigenetic therapy with pharmacotherapy for optimal results.
Collapse
Affiliation(s)
- Chioma Laura Odimegwu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Samuel Nkachukwu Uwaezuoke
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ugo N Chikani
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ngozi Rita Mbanefo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ken Daberechi Adiele
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | - Chizoma Ihuarula Eneh
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Chibuzo Obiora Ndiokwelu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Somkenechi C Okpala
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Francis N Ogbuka
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Kenneth E Odo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | | |
Collapse
|
49
|
Raz M, Milo T, Glass DS, Mayo A, Alon U. Endocrine gland size is proportional to its target tissue size. iScience 2024; 27:110625. [PMID: 39224518 PMCID: PMC11367476 DOI: 10.1016/j.isci.2024.110625] [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: 10/25/2023] [Revised: 06/26/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Endocrine glands secrete hormones into the circulation to target distant tissues and regulate their functions. The qualitative relationship between hormone-secreting organs and their target tissues is well established, but a quantitative approach is currently limited. Quantification is important, as it could allow us to study the endocrine system using engineering concepts of optimality and tradeoffs. In this study, we collected literature data on 24 human hormones secreted from dedicated endocrine cells. We find that the number of endocrine cells secreting a hormone is proportional to the number of its target cells. A single endocrine cell serves approximately 2,000 target cells, a relationship that spans 6 orders of magnitude of cell numbers. This suggests an economic principle of cells working near their maximal capacity, and glands that are no bigger than they need to be.
Collapse
Affiliation(s)
- Moriya Raz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tomer Milo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - David S. Glass
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
50
|
Chen S, Yang X, Gu H, Wang Y, Xu Z, Jiang Y, Wang Y. Predictive etiological classification of acute ischemic stroke through interpretable machine learning algorithms: a multicenter, prospective cohort study. BMC Med Res Methodol 2024; 24:199. [PMID: 39256656 PMCID: PMC11384709 DOI: 10.1186/s12874-024-02331-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 09/05/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND The prognosis, recurrence rates, and secondary prevention strategies varied significantly among different subtypes of acute ischemic stroke (AIS). Machine learning (ML) techniques can uncover intricate, non-linear relationships within medical data, enabling the identification of factors associated with etiological classification. However, there is currently a lack of research utilizing ML algorithms for predicting AIS etiology. OBJECTIVE We aimed to use interpretable ML algorithms to develop AIS etiology prediction models, identify critical factors in etiology classification, and enhance existing clinical categorization. METHODS This study involved patients with the Third China National Stroke Registry (CNSR-III). Nine models, which included Natural Gradient Boosting (NGBoost), Categorical Boosting (CatBoost), Extreme Gradient Boosting (XGBoost), Random Forest (RF), Light Gradient Boosting Machine (LGBM), Gradient Boosting Decision Tree (GBDT), Adaptive Boosting (AdaBoost), Support Vector Machine (SVM), and logistic regression (LR), were employed to predict large artery atherosclerosis (LAA), small vessel occlusion (SVO), and cardioembolism (CE) using an 80:20 randomly split training and test set. We designed an SFS-XGB with 10-fold cross-validation for feature selection. The primary evaluation metrics for the models included the area under the receiver operating characteristic curve (AUC) for discrimination and the Brier score (or calibration plots) for calibration. RESULTS A total of 5,213 patients were included, comprising 2,471 (47.4%) with LAA, 2,153 (41.3%) with SVO, and 589 (11.3%) with CE. In both LAA and SVO models, the AUC values of the ML models were significantly higher than that of the LR model (P < 0.001). The optimal model for predicting SVO (AUC [RF model] = 0.932) outperformed the optimal LAA model (AUC [NGB model] = 0.917) and the optimal CE model (AUC [LGBM model] = 0.846). Each model displayed relatively satisfactory calibration. Further analysis showed that the optimal CE model could identify potential CE patients in the undetermined etiology (SUE) group, accounting for 1,900 out of 4,156 (45.7%). CONCLUSIONS The ML algorithm effectively classified patients with LAA, SVO, and CE, demonstrating superior classification performance compared to the LR model. The optimal ML model can identify potential CE patients among SUE patients. These newly identified predictive factors may complement the existing etiological classification system, enabling clinicians to promptly categorize stroke patients' etiology and initiate optimal strategies for secondary prevention.
Collapse
Affiliation(s)
- Siding Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China
- Changping Laboratory, Beijing, China
| | - Xiaomeng Yang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China
| | - Hongqiu Gu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China
| | - Yanzhao Wang
- School of Statistics, Renmin University of China, No. 59 Zhongguancun Street, Haidian District, Beijing, 100872, China
| | - Zhe Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China
| | - Yong Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.
- Changping Laboratory, Beijing, China.
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing, 100091, China.
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.
- Changping Laboratory, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
- Clinical Center for Precision Medicine in Stroke, Capital Medical University, Beijing, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|