1
|
Mkhize BC, Mosili P, Ngubane PS, Sibiya NH, Khathi A. Increased Plasma Levels of ACE and Ang II in Prediabetes May Contribute to Adipose Tissue Dysfunction. Int J Mol Sci 2025; 26:5517. [PMID: 40564980 PMCID: PMC12192874 DOI: 10.3390/ijms26125517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/31/2025] [Accepted: 06/04/2025] [Indexed: 06/28/2025] Open
Abstract
Adipose tissue is essential for the regulation of insulin sensitivity and cytokine production, which are key processes in maintaining metabolic homeostasis. Previous studies have shown a link between the renin-angiotensin system (RAS) and adipose tissue dysfunction in type 2 diabetes (T2D); however, the role of RAS in prediabetes remains underexplored. This study aimed to analyze the association between RAS components and adipose tissue dysfunction in the prediabetic state. This observational, cross-sectional study was conducted between 21/05/21 and 20/05/24 and analyzed RAS markers in plasma samples. This study was conducted at King Edward Hospital, focusing on individuals from outpatient clinics. The study included non-prediabetic (NPD), prediabetic (PD), and T2D individuals (n = 40 per group) aged 25-45 years. The participants were selected based on fasting blood glucose levels and HbA1c criteria. Plasma RAS markers and adipose function markers were measured in each participant. Primary outcomes included HOMA-IR, HbA1c, and plasma levels of ACE1, Ang II, ACE2, Ang 1-7, adiponectin, adipsin, MCP-1, and HDL. PD participants had significantly altered glycaemic control (HOMA-IR: 2.1 ± 0.4 vs. 3.9 ± 0.8; HbA1c: 4.9 ± 0.4 vs. 5.9 ± 0.6) compared to NPD. Plasma ACE1 (162.0 ± 10.55 vs. 180.3 ± 7.546) and Ang II (20.26 ± 2.404 vs. 25 ± 1.752) were elevated, while adiponectin (29.08 ± 5.72 vs. 23.22 ± 4.93) and HDL (1.01 ± 0.11 vs. 0.67 ± 0.11) were reduced in PD. Alterations in RAS manifest early in prediabetes and are associated with adipose tissue dysfunction. These findings suggest that RAS dysregulation contributes to early metabolic disturbances in prediabetes.
Collapse
Affiliation(s)
- Bongeka Cassandra Mkhize
- School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Durban 3629, KwaZulu-Natal, South Africa;
| | - Palesa Mosili
- Department of Human Physiology, University of KwaZulu-Natal, Westville 4000, KwaZulu-Natal, South Africa; (P.M.); (P.S.N.)
| | - Phikelelani Sethu Ngubane
- Department of Human Physiology, University of KwaZulu-Natal, Westville 4000, KwaZulu-Natal, South Africa; (P.M.); (P.S.N.)
| | | | - Andile Khathi
- Department of Human Physiology, University of KwaZulu-Natal, Westville 4000, KwaZulu-Natal, South Africa; (P.M.); (P.S.N.)
| |
Collapse
|
2
|
Pereira de Lima R, Li A, Gilani A, Rubio-Navarro A, Warren CD, Kong IY, Geri JB, Lo JC. C3aR1 on β cells enhances β cell function and survival to maintain glucose homeostasis. Mol Metab 2025; 96:102134. [PMID: 40189102 PMCID: PMC12018202 DOI: 10.1016/j.molmet.2025.102134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/23/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
OBJECTIVE Pancreatic β cell dysfunction is critical to the development of type 2 diabetes (T2D). Our previous studies suggested that C3aR1 on β cells promotes insulin secretion and cell survival. However, as C3aR1 is expressed on many other cell types including within the islets, whole-body C3aR1 knockout models confound the analyses of direct impacts on β cells. METHODS To clarify the role of C3aR1 in β cells under T2D conditions, we generated β cell-specific C3aR1 knockout mice. We assessed glucose homeostasis, focusing on β cell function and mass under metabolic stress conditions, to interrogate the effects of C3aR1 on β cells in a mouse model of T2D. We performed proteomic analyses on islets from control and β cell-specific C3aR1 knockout mice. To determine potential translational relevance, C3AR1 was assessed alongside glucose-stimulated insulin secretion in human islets. RESULTS We show that the complement receptor C3aR1 on β cells plays an essential role in maintaining β cell homeostasis, especially under the metabolic duress of obesity and T2D. Male mice with β cell specific deletion of C3ar1 (β-C3aR1 KO) exhibit worse glucose tolerance and lower insulin levels when fed regular or high fat diet. Under high fat diet, β-C3aR1 KO also have diminished β cell mass. Islets from β-C3aR1 KO mice demonstrate impaired insulin secretion. β cells lacking C3aR1 display increased susceptibility to lipotoxicity-mediated cell death. Markers of β cell identity are decreased in β-C3aR1 KO mice while stress markers are elevated. Disruption of C3ar1 on β cells ablates the insulin secretory response to C3a, establishing a signaling axis between C3a and β cell-derived C3aR1. Islet proteomic analyses highlight the MAPK pathway and mitochondrial dysfunction with C3aR1 loss in β cells. Finally, we show that C3AR1 is positively correlated with insulin secretion in human islets. CONCLUSIONS These findings indicate that C3aR1 expression on β cells is necessary to maintain optimal β cell function and preserve β cell mass in T2D.
Collapse
Affiliation(s)
- Renan Pereira de Lima
- Division of Cardiology, Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ang Li
- Division of Cardiology, Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ankit Gilani
- Division of Cardiology, Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Charles D Warren
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Isabella Y Kong
- Division of Pediatric Hematology/Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Jacob B Geri
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - James C Lo
- Division of Cardiology, Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
3
|
Sriboonaied P, Phuangbubpha P, Saetan P, Charoensuksai P, Charoenpanich A. Dual Modulation of Adipogenesis and Apoptosis by PPARG Agonist Rosiglitazone and Antagonist Betulinic Acid in 3T3-L1 Cells. Biomedicines 2025; 13:1340. [PMID: 40564064 PMCID: PMC12190099 DOI: 10.3390/biomedicines13061340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Revised: 05/27/2025] [Accepted: 05/28/2025] [Indexed: 06/28/2025] Open
Abstract
Background/Objectives: Disruptions in adipose tissue dynamics contribute to obesity-related metabolic disorders, emphasizing the need for targeted therapies focusing on adipose tissue cells, including progenitor cells and adipocytes. Peroxisome proliferator-activated receptor gamma (PPARG) ligands are potent insulin sensitizers used in type 2 diabetes treatment. This study investigated the effects of rosiglitazone, a PPARG agonist, and betulinic acid, a PPARG antagonist, on adipogenesis and apoptosis in 3T3-L1 pre-adipocytes. Method: 3T3-L1 pre-adipocytes were treated with rosiglitazone or betulinic acid during adipogenic differentiation. Lipid droplet formation was used to evaluate adipogenesis. Cell growth and cell death were assessed using the resazurin-based cell viability assay, trypan blue exclusion assay, LDH assay, and Annexin V/PI staining. Quantitative PCR was conducted to examine the expression of genes associated with adipogenesis and apoptosis. Results: Betulinic acid reduced adipogenesis only when administered daily for eight days. Rosiglitazone did not alter the overall lipid quantity; however, it promoted a shift toward fewer but larger lipid droplets. Both compounds increased Adipoq and Cfd expression, and betulinic acid also elevated Fabp4. Rosiglitazone induced stronger cell aggregation. Despite increased cell death, overall viability was maintained. Apoptotic cell death was enhanced by both compounds and confirmed via Annexin V/PI staining and flow cytometry, accompanied by downregulation of Ccnd1 and Bcl2. Additionally, rosiglitazone markedly increased the expression of Cebpa, a key regulator that can modulate lipid droplet formation and the balance between cell growth and death. Conclusions: Rosiglitazone and betulinic acid differentially modulate adipogenesis and apoptosis in 3T3-L1 cells, revealing a complex interplay between lipid accumulation and programmed cell death. Together, the findings underscore the potential of dual PPARG-targeting approaches for metabolic disease interventions.
Collapse
Affiliation(s)
- Patsawee Sriboonaied
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand; (P.S.); (P.P.); (P.S.)
| | - Pornwipa Phuangbubpha
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand; (P.S.); (P.P.); (P.S.)
| | - Puretat Saetan
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand; (P.S.); (P.P.); (P.S.)
| | - Purin Charoensuksai
- Department of Biomedicine and Health Informatics and Bioactives from Natural Resources Research Collaboration for Excellence in Pharmaceutical Sciences (BNEP), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand;
| | - Adisri Charoenpanich
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand; (P.S.); (P.P.); (P.S.)
| |
Collapse
|
4
|
Laker RC, Egolf S, Will S, Lantier L, McGuinness OP, Brown C, Bhagroo N, Oldham S, Kuszpit K, Alfaro A, Li X, Kang T, Pellegrini G, Andréasson AC, Kajani S, Sitaula S, Larsen MR, Rhodes CJ. GLP-1R/GCGR dual agonism dissipates hepatic steatosis to restore insulin sensitivity and rescue pancreatic β-cell function in obese male mice. Nat Commun 2025; 16:4714. [PMID: 40399267 PMCID: PMC12095689 DOI: 10.1038/s41467-025-59773-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/29/2025] [Indexed: 05/23/2025] Open
Abstract
An early driver of Type 2 diabetes mellitus (T2D) is ectopic fat accumulation, especially in the liver, that impairs insulin sensitivity. In T2D, GLP-1R/GCGR dual-agonists reduce glycaemia, body weight and hepatic steatosis. Here, we utilize cotadutide, a well characterized GLP-1R/GCGR dual-agonist, and demonstrate improvement of insulin sensitivity during hyperinsulinemic euglycemic clamp following sub-chronic dosing in male, diet-induced obese (DIO) mice. Phosphoproteomic analyses of insulin stimulated liver from cotadutide-treated mice identifies previously unknown and known phosphorylation sites on key insulin signaling proteins associated with improved insulin sensitivity. Cotadutide or GCGR mono-agonist treatment also increases brown adipose tissue (BAT) insulin-stimulated glucose uptake, while GLP-1R mono-agonist shows a weak effect. BAT from cotadutide-treated mice have induction of UCP-1 protein, increased mitochondrial area and a transcriptomic profile of increased fat oxidation and mitochondrial activity. Finally, the cotadutide-induced improvement in insulin sensitivity is associated with reduction of insulin secretion from isolated pancreatic islets indicating reduced insulin secretory demand. Here we show, GLP-1R/GCGR dual agonism provides multimodal efficacy to decrease hepatic steatosis and consequently improve insulin sensitivity, in concert with recovery of endogenous β-cell function and reduced insulin demand. This substantiates GLP-1R/GCGR dual-agonism as a potentially effective T2D treatment.
Collapse
Affiliation(s)
- Rhianna C Laker
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
| | - Shaun Egolf
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Sarah Will
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Louise Lantier
- Vanderbilt University Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - Owen P McGuinness
- Vanderbilt University Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - Charles Brown
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, AstraZeneca, Gaithersburg, MD, USA
| | - Nicholas Bhagroo
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Stephanie Oldham
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Kyle Kuszpit
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, AstraZeneca, Gaithersburg, MD, USA
| | - Alex Alfaro
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, AstraZeneca, Gaithersburg, MD, USA
| | - Xidan Li
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Taewook Kang
- Department of Biochemistry and Molecular Biology, PR group, University of Southern Denmark, Odense, Denmark
| | - Giovanni Pellegrini
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anne-Christine Andréasson
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sarina Kajani
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Sadichha Sitaula
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, PR group, University of Southern Denmark, Odense, Denmark
| | - Christopher J Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
| |
Collapse
|
5
|
Cuesta‐Gomez N, Castro C, Rosko M, Seeberger K, Korbutt GS. Sex Differences in Maturation and Function of Neonatal Porcine Islets Upon Transplantation in Mice. Xenotransplantation 2025; 32:e70039. [PMID: 40243327 PMCID: PMC12005065 DOI: 10.1111/xen.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/08/2025] [Accepted: 03/14/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Neonatal porcine islets (NPIs) can mature into a mixed population of endocrine cells that can restore glucose control in mice, pigs, and non-human primates, representing a potential alternative islet source for clinical beta cell replacement therapy. However, it remains unclear how conditions in the recipient influence the maturation and function of these cells. Here, we investigated the impact of host sex on NPIs implanted under the kidney capsule of male and female B6.129S7-Rag1tm1Mom (B6/Rag-/-) mice. METHODS Diabetic mice were transplanted with 3000 NPIs under the kidney capsule. All mice were monitored for reversal of hyperglycemia and glucose clearance at 8- and 20-weeks post-transplant. Grafts were assessed for cell composition and insulin content. RESULTS Female mice demonstrated improved glucose clearance at 8- and 20-weeks post-transplant compared to their male counterparts. Improved glucose clearance correlated with accelerated diabetes reversal in females (8 weeks vs. 12 weeks in males) and increased rates of euglycemic achievement (17/18 in females vs. 14/19 in males). However, grafts collected from male mice exhibited an increased percentage of insulin-positive cells as well as increased insulin content. CONCLUSION The sex of the host influences the outcomes of NPI transplantation, showcasing the relevance of understanding the role of sex as a biological variable in islet transplantation.
Collapse
Affiliation(s)
- Nerea Cuesta‐Gomez
- Department of SurgeryUniversity of AlbertaEdmontonAlbertaCanada
- Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Chelsea Castro
- Department of SurgeryUniversity of AlbertaEdmontonAlbertaCanada
- Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Mandy Rosko
- Department of SurgeryUniversity of AlbertaEdmontonAlbertaCanada
- Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Karen Seeberger
- Department of SurgeryUniversity of AlbertaEdmontonAlbertaCanada
- Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| | - Gregory S. Korbutt
- Department of SurgeryUniversity of AlbertaEdmontonAlbertaCanada
- Alberta Diabetes InstituteUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
6
|
Jiao J, Gao F, Zhao H, Jiang M, Zhou Y, Liu D, Fang S, Gao D, Wang Z, Yang Z, Yuan H. Exploring the Plasma Proteome: Identifying Hub Proteins linking Aging, Homeostasis, and Organ Function. Int J Med Sci 2025; 22:1109-1123. [PMID: 40027189 PMCID: PMC11866526 DOI: 10.7150/ijms.107750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/17/2025] [Indexed: 03/05/2025] Open
Abstract
As effectors of interactions between genes and the environment, plasma proteins can monitor homeostasis and reflect the aging state of an organism. However, biomarkers of aging that are associated with homeostasis are still unclear. This study investigates the phenotype-related plasma proteome profiles of healthy individuals and to identify proteins that are specifically related to aging and physiological indices and their expression patterns across the lifespan. From September 2020 to March 2021, 71 participants aged over 20 to 100 years were enrolled in this cross-sectional study. Data were analyzed from April 2021 to December 2023. The plasma proteome was analyzed to identify proteins that are specifically related to aging and their expression patterns across the lifespan. Then, hub proteins were screened through correlation of aging proteins with physiological and biochemical phenotypes. Based on levels of plasma proteins, physiological indices are associated with age. Additionally, these differences in protein expression correlate with age and physiological indices. Finally, we identified 20 hub proteins that correlate with both physiological indices and age, and these proteins are involved in oxidative stress, inflammation and metabolism. Bibliometric analysis confirmed that 8 hub proteins (CD44, CD14, IGF2, CFD, LBP, IGFBP3, EFEMP1, and AHSG) associated with age affect organ function by mediating homeostasis. Plasma proteins associated with both age and physiological indices are involved in oxidative stress, inflammation, and metabolism. This is the first investigation to link aging and homeostasis based on plasma proteins.
Collapse
Affiliation(s)
- Juan Jiao
- Department of Clinical Laboratory, the Seventh Medical Center, Chinese PLA General Hospital, Beijing 100700, P.R. China
| | - Fei Gao
- Department of Research & Development, Beijing IPE Center for Clinical Laboratory CO, Beijing 100176, P.R. China
| | - Hongye Zhao
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Mingjun Jiang
- Respiratory Department, Beijing Children's Hospital, Capital Medical University, China National Clinical Research Center of Respiratory Diseases, National Center for Children's Health, Beijing 100045, P.R. China
| | - Yan Zhou
- General Practice Department, Beijing Hospital, Beijing 100730, P.R. China
| | - Dizhi Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, P.R. China
| | - Sihang Fang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, P.R. China
| | - Danni Gao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, P.R. China
| | - Zhaoping Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, P.R. China
| | - Ze Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, P.R. China
| | - Huiping Yuan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, P.R. China
| |
Collapse
|
7
|
Zhao K, Zhang H, Ding W, Yu X, Hou Y, Liu X, Li X, Wang X. Adipokines regulate the development and progression of MASLD through organellar oxidative stress. Hepatol Commun 2025; 9:e0639. [PMID: 39878681 PMCID: PMC11781772 DOI: 10.1097/hc9.0000000000000639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), which is increasingly being recognized as a leading cause of chronic liver pathology globally, is increasing. The pathophysiological underpinnings of its progression, which is currently under active investigation, involve oxidative stress. Human adipose tissue, an integral endocrine organ, secretes an array of adipokines that are modulated by dietary patterns and lifestyle choices. These adipokines intricately orchestrate regulatory pathways that impact glucose and lipid metabolism, oxidative stress, and mitochondrial function, thereby influencing the evolution of hepatic steatosis and progression to metabolic dysfunction-associated steatohepatitis (MASH). This review examines recent data, underscoring the critical interplay of oxidative stress, reactive oxygen species, and redox signaling in adipokine-mediated mechanisms. The role of various adipokines in regulating the onset and progression of MASLD/MASH through mitochondrial dysfunction and endoplasmic reticulum stress and the underlying mechanisms are discussed. Due to the emerging correlation between adipokines and the development of MASLD positions, these adipokines are potential targets for the development of innovative therapeutic interventions for MASLD management. A comprehensive understanding of the pathogenesis of MASLD/MASH is instrumental for identifying therapies for MASH.
Collapse
Affiliation(s)
- Ke Zhao
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
| | - Heng Zhang
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
- Central laboratory, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wenyu Ding
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
| | - Xiaoshuai Yu
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
- Central laboratory, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yanli Hou
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
| | - Xihong Liu
- Department of Pathology, The Fourth People’s Hospital of Jinan, Jinan, Shandong, China
| | - Xinhua Li
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
| | - Xiaolei Wang
- Central laboratory, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Central laboratory, Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Jinan, Shandong, China
- First school of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
8
|
Homan EA, Gilani A, Rubio-Navarro A, Johnson MA, Schaepkens OM, Cortada E, Pereira de Lima R, Stoll L, Lo JC. Complement 3a receptor 1 on macrophages and Kupffer cells is not required for the pathogenesis of metabolic dysfunction-associated steatotic liver disease. eLife 2025; 13:RP100708. [PMID: 39773465 PMCID: PMC11709426 DOI: 10.7554/elife.100708] [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] [Indexed: 01/30/2025] Open
Abstract
Together with obesity and type 2 diabetes, metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing global epidemic. Activation of the complement system and infiltration of macrophages has been linked to progression of metabolic liver disease. The role of complement receptors in macrophage activation and recruitment in MASLD remains poorly understood. In human and mouse, C3AR1 in the liver is expressed primarily in Kupffer cells, but is downregulated in humans with MASLD compared to obese controls. To test the role of complement 3a receptor (C3aR1) on macrophages and liver resident macrophages in MASLD, we generated mice deficient in C3aR1 on all macrophages (C3aR1-MφKO) or specifically in liver Kupffer cells (C3aR1-KpKO) and subjected them to a model of metabolic steatotic liver disease. We show that macrophages account for the vast majority of C3ar1 expression in the liver. Overall, C3aR1-MφKO and C3aR1-KpKO mice have similar body weight gain without significant alterations in glucose homeostasis, hepatic steatosis and fibrosis, compared to controls on a MASLD-inducing diet. This study demonstrates that C3aR1 deletion in macrophages or Kupffer cells, the predominant liver cell type expressing C3ar1, has no significant effect on liver steatosis, inflammation or fibrosis in a dietary MASLD model.
Collapse
Affiliation(s)
- Edwin A Homan
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Maya A Johnson
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Odin M Schaepkens
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Eric Cortada
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Renan Pereira de Lima
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Lisa Stoll
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - James C Lo
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| |
Collapse
|
9
|
Cortada E, Yao J, Xia Y, Dündar F, Zumbo P, Yang B, Rubio-Navarro A, Perder B, Qiu M, Pettinato AM, Homan EA, Stoll L, Betel D, Cao J, Lo JC. Cross-species single-cell RNA-seq analysis reveals disparate and conserved cardiac and extracardiac inflammatory responses upon heart injury. Commun Biol 2024; 7:1611. [PMID: 39627536 PMCID: PMC11615278 DOI: 10.1038/s42003-024-07315-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
The immune system coordinates the response to cardiac injury and controls regenerative and fibrotic scar outcomes in the heart and subsequent chronic low-grade inflammation associated with heart failure. Adult mice and humans lack the ability to fully recover while adult zebrafish spontaneously regenerate after heart injury. Here we profile the inflammatory response to heart cryoinjury in zebrafish and coronary artery ligation in mouse using single cell transcriptomics. We interrogate the extracardiac reaction to cardiomyocyte necrosis to assess the specific peripheral tissue and immune cell reaction to chronic stress. Cardiac macrophages play a critical role in determining tissue homeostasis by healing versus scarring. We identify distinct transcriptional clusters of monocytes/macrophages (mono/Mϕ) in each species and find analogous pairs in zebrafish and mice. However, the reaction to myocardial injury is largely disparate between mice and zebrafish. The dichotomous response to heart damage between the murine and zebrafish mono/Mϕ and/or the presence of distinct zebrafish mono/Mϕ subtypes may underlie the impaired regenerative process in adult mammals and humans. Our study furnishes a direct cross-species comparison of immune responses between regenerative and profibrotic myocardial injury models, providing a useful resource to the fields of regenerative biology and cardiovascular research.
Collapse
Affiliation(s)
- Eric Cortada
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Jun Yao
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Yu Xia
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Friederike Dündar
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Paul Zumbo
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Boris Yang
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Björn Perder
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Miaoyan Qiu
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Anthony M Pettinato
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
| | - Edwin A Homan
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Lisa Stoll
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA.
- Institute for Computational Biomedicine, Division of Hematology and Medical, Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Jingli Cao
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA.
| | - James C Lo
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA.
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
10
|
Zhang X, Jiang M, Zhang X, Zuo Y, Zhang H, Zhang T, Yang L, Lin J, Zhang Y, Dai X, Ge W, Sun C, Yang F, Zhang J, Liu Y, Wang Y, Qiang H, Yang X, Sun D. Adipsin improves diabetic hindlimb ischemia through SERPINE1 dependent angiogenesis. Cardiovasc Diabetol 2024; 23:429. [PMID: 39623437 PMCID: PMC11613494 DOI: 10.1186/s12933-024-02526-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Adipsin (complement factor D, CFD), as the first described adipokine, is well-known for its regulatory effects in diabetic cardiovascular complications. However, its role in diabetic hind-limb ischemia was not clarified. This study aimed to evaluate the possible therapeutic effect of Adipsin in hind-limb ischemia in type 2 diabetic mice and to elucidate the molecular mechanisms involved. METHODS A high-fat diet and streptozotocin (HFD/STZ)-induced diabetic mouse model, and a transgenic mouse model with adipose tissue-specific overexpression of Adipsin (Adipsin-Tg) were employed. Hindlimb ischemia was established by femoral artery ligation, and blood flow recovery was monitored using Laser Doppler perfusion imaging. Molecular mechanisms underlying Adipsin-potentiated angiogenesis were examined using RNA sequencing and co-immunoprecipitation/mass spectrometry (Co-IP/MS) analyses. RESULTS Adipsin expression was upregulated in non-diabetic mice following HLI, while suppressed in diabetic mice, indicating its potential role in ischemic recovery which is impaired in diabetes. Adipsin-Tg mice exhibited significantly improved blood flow recovery, increased capillary density, and enhanced muscle regeneration in comparison with non-transgenic (NTg) diabetic mice. Adipsin facilitated proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) under hyperglycemic and hypoxic conditions. Additionally, it enhanced phosphorylation of AKT, ERK, and eNOS pathways both in vivo and in vitro. RNA sequencing and co-immunoprecipitation/mass spectrometry (Co-IP/MS) analyses identified that Adipsin promoted angiogenesis by interacting with SERBP1, which disrupted the binding of SERBP1 to SERPINE1 mRNA, resulting in reduced SERPINE1 expression and the subsequent activation of the VEGFR2 signaling cascade. CONCLUSIONS Adipsin promotes angiogenesis and facilitates blood perfusion recovery in diabetic mice with HLI by downregulating SERPINE1 through interaction with SERBP1. These findings elucidate a novel therapeutic potential for Adipsin in the management of PAD in diabetic patients, highlighting its role in enhancing angiogenesis and tissue repair.
Collapse
MESH Headings
- Animals
- Ischemia/physiopathology
- Ischemia/metabolism
- Ischemia/genetics
- Hindlimb
- Neovascularization, Physiologic
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/complications
- Humans
- Male
- Mice, Inbred C57BL
- Regional Blood Flow
- Human Umbilical Vein Endothelial Cells/metabolism
- Complement Factor D/metabolism
- Complement Factor D/genetics
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/metabolism
- Signal Transduction
- Mice, Transgenic
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/physiopathology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/etiology
- Cell Proliferation
- Recovery of Function
- Mice
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Cell Movement
- Angiogenesis Inducing Agents/pharmacology
- Cells, Cultured
- Microvascular Density
- Angiogenesis
Collapse
Affiliation(s)
- Xiaohua Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yixuan Zuo
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Huanle Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Tingting Zhang
- Xijing 986 Hospital Department, Air Force Medical University, Xi'an, China
| | - Liyu Yang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xinchun Dai
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Wen Ge
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Chuang Sun
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Fang Yang
- Basic Medical Teaching Experiment Center, Basic Medical College, Air Force Medical University, Xi'an, China
| | - Jiye Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yue Liu
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yangyang Wang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Huanhuan Qiang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xiaojie Yang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China.
| |
Collapse
|
11
|
Park JH, Nguyen TN, Shim HM, Yu GI, Ha EY, Cho H. Identification of Adipsin as a Biomarker of Beta Cell Function in Patients with Type 2 Diabetes. J Clin Med 2024; 13:7351. [PMID: 39685809 DOI: 10.3390/jcm13237351] [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: 10/28/2024] [Revised: 11/20/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Adipsin, an adipokine, is known to play an important role in maintaining the function of pancreatic beta cells in mice. This study aimed to investigate whether adipsin could be a circulating biomarker for evaluating the function of beta cells in patients with type 2 diabetes (T2D). Methods: Plasma adipsin concentrations were measured using immunoassay in three distinct subject groups: normoglycemia, T2D without insulin treatment (T2D-w/o-insulin), and T2D treated with insulin (T2D-with-insulin). Adipsin expressions were evaluated in three distinct mouse groups: normal diet (ND), high-fat diet (HFD), and HFD with streptozotocin (STZ) and nicotinamide (NA). Results: The T2D-with-insulin group exhibited a significant decrease in plasma adipsin concentration (3.91 ± 1.51 μg/mL) compared to the T2D-w/o-insulin group (5.11 ± 1.53 μg/mL; p < 0.001), whereas the T2D-w/o-insulin group showed a significantly increased plasma adipsin concentration compared to the normoglycemia group (4.53 ± 1.15 μg/mL). Plasma adipsin concentration was positively correlated with fasting C-peptide level (p < 0.001), 2-h C-peptide level (p < 0.001), and 2-h C-peptidogenic index (p < 0.001) in the diabetic groups. HFD mice showed a significant increase in pancreatic islet size, plasma insulin and adipsin levels, as well as adipsin expression in white adipose tissue (WAT) compared to ND mice. In contrast, the insulin-deficient T2D model (HFD-STZ-NA) demonstrated a marked reduction in pancreatic islet size, plasma insulin and adipsin concentrations, and adipsin expression in WAT compared to the HFD mice. Conclusions: plasma adipsin may be useful for evaluating pancreatic beta cell function in patients with T2D.
Collapse
Affiliation(s)
- Jae-Hyung Park
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Thi Nhi Nguyen
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Hye Min Shim
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Gyeong Im Yu
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Eun Yeong Ha
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Hochan Cho
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| |
Collapse
|
12
|
Alic L, Dendinovic K, Papac-Milicevic N. The complement system in lipid-mediated pathologies. Front Immunol 2024; 15:1511886. [PMID: 39635529 PMCID: PMC11614835 DOI: 10.3389/fimmu.2024.1511886] [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: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
The complement system, a coordinator and facilitator of the innate immune response, plays an essential role in maintaining host homeostasis. It promotes clearance of pathogen- and danger-associated molecular patterns, regulates adaptive immunity, and can modify various metabolic processes such as energy expenditure, lipid metabolism, and glucose homeostasis. In this review, we will focus on the intricate interplay between complement components and lipid metabolism. More precisely, we will display how alterations in the activation and regulation of the complement system affect pathological outcome in lipid-associated diseases, such as atherosclerosis, obesity, metabolic syndrome, age-related macular degeneration, and metabolic dysfunction-associated steatotic liver disease. In addition to that, we will present and evaluate underlying complement-mediated physiological mechanisms, observed both in vitro and in vivo. Our manuscript will demonstrate the clinical significance of the complement system as a bridging figure between innate immunity and lipid homeostasis.
Collapse
Affiliation(s)
- Lejla Alic
- Department of Medical Biochemistry, Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Kristina Dendinovic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nikolina Papac-Milicevic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Han R, Huang H, Zhu J, Jin X, Wang Y, Xu Y, Xia Z. Adipokines and their potential impacts on susceptibility to myocardial ischemia/reperfusion injury in diabetes. Lipids Health Dis 2024; 23:372. [PMID: 39538244 PMCID: PMC11558907 DOI: 10.1186/s12944-024-02357-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Coronary artery disease has a high mortality rate and is a striking public health concern, affecting a substantial portion of the global population. On the early onset of myocardial ischemia, thrombolytic therapy and coronary revascularization could promptly restore the bloodstream and nutrient supply to the ischemic tissue, efficiently preserving less severely injured myocardium. However, the abrupt re-establishment of blood flow triggers the significant discharge of previously accumulated oxidative substances and inflammatory cytokines, leading to further harm referred to as ischemia/reperfusion (I/R) injury. Diabetes significantly raises the vulnerability of the heart to I/R injury due to disrupted glucose and lipid processing, impaired insulin sensitivity and metabolic signaling, and increased inflammatory responses. Numerous studies have indicated that adipokines are crucial in the etiology and pathogenesis of obesity, diabetes, hyperlipidemia, hypertension, and coronary artery disease. Adipokines such as adiponectin, adipsin, visfatin, chemerin, omentin, and apelin, which possess protective properties against inflammatory activity and insulin resistance, have been shown to confer myocardial protection in conditions such as atherosclerosis, myocardial hypertrophy, myocardial I/R injury, and diabetic complications. On the other hand, adipokines such as leptin and resistin, known for their pro-inflammatory characteristics, have been linked to elevated cardiac lipid deposition, insulin resistance, and fibrosis. Meteorin-like (metrnl) exhibits opposite effects in various pathological conditions. However, the data on adipokines in myocardial I/R, especially in diabetes, is still incomplete and controversial. This review focuses on recent research regarding the categorization and function of adipokines in the heart muscle, and the identification of different signaling pathways involved in myocardial I/R injury under diabetic conditions, aiming to facilitate the exploration of therapeutic strategies against myocardial I/R injury in diabetes.
Collapse
Affiliation(s)
- Ronghui Han
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Hemeng Huang
- Department of Emergency, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Jianyu Zhu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Xiaogao Jin
- Department of Anesthesiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Yongyan Wang
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China.
- Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, Hengqin, Zhuhai, People's Republic of China.
- Faculty of Pharmacy, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, People's Republic of China.
| | - Zhengyuan Xia
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China.
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
14
|
de Lima RP, Li A, Gilani A, Lo JC. C3aR1 on β cells enhances β cell function and survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.622969. [PMID: 39605339 PMCID: PMC11601266 DOI: 10.1101/2024.11.11.622969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Pancreatic β cell dysfunction is critical to the development of type 2 diabetes (T2D). We show that the complement receptor C3aR1 on β cells plays an essential role in maintaining β cell homeostasis, especially under the metabolic duress of obesity and T2D. Mice with β cell specific deletion of C3ar1 have worse glucose tolerance, lower insulin levels, and decreased β cell mass. Islets from β cell specific C3ar1 knockout (β-C3aR1 KO) mice demonstrate impaired insulin secretion. Disruption of C3ar1 on β cells ablates the insulin secretory response to C3a, establishing a signaling axis between C3a and β cell-derived C3aR1. Markers of β cell identity were decreased while stress markers were increased in β-C3aR1 KO mice. Islets from β-C3aR1 KO also exhibit increased β cell death to lipotoxicity. Finally, we show that C3AR1 is positively correlated with insulin secretion in human islets. These findings indicate that C3aR1 expression on β cells is necessary to maintain optimal β cell function and preserve β cell mass in T2D.
Collapse
|
15
|
Okita T, Kita S, Fukuda S, Kondo Y, Sakaue TA, Iioka M, Fukuoka K, Kawada K, Nagao H, Obata Y, Fujishima Y, Ebihara T, Matsumoto H, Nakagawa S, Kimura T, Nishizawa H, Shimomura I. Soluble T-cadherin secretion from endothelial cells is regulated via insulin/PI3K/Akt signalling. Biochem Biophys Res Commun 2024; 732:150403. [PMID: 39047402 DOI: 10.1016/j.bbrc.2024.150403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/27/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
AIM AND OBJECTIVE Our recent report showed that soluble T-cadherin promotes pancreatic beta-cell proliferation. However, how and where the secretion of soluble T-cadherin is regulated remain unclear. METHODS AND RESULTS Soluble T-cadherin levels significantly increased in leptin receptor-deficient db/db mice with hypoinsulinaemia or in wild-type mice treated with insulin receptor blockade by S961. Similar results were observed in human subjects; Diabetic ketoacidosis patients at the time of hospitalization had increased plasma soluble T-cadherin levels, which decreased after insulin infusion therapy. Patients with recurrent ovarian cancer who were administered a phosphatidylinositol-3 kinase (PI3K)-alpha inhibitor (a new anticancer drug) had increased plasma soluble T-cadherin and plasma C-peptide levels. Endothelial cell-specific T-cadherin knockout mice, but not skeletal muscle- or cardiac muscle-specific T-cadherin knockout mice, showed a 26 % reduction in plasma soluble T-cadherin levels and a significant increase in blood glucose levels in streptozocin-induced diabetes. The secretion of soluble T-cadherin from human endothelial cells was approximately 20 % decreased by insulin and this decrease was canceled by blockade of insulin receptor/Akt signalling, not Erk signalling. CONCLUSION We conclude that insulin regulates soluble T-cadherin levels and soluble T-cadherin secretion from endothelial cells is positively regulated by insulin/insulin receptor/Akt signalling.
Collapse
Affiliation(s)
- Tomonori Okita
- Departments of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shunbun Kita
- Departments of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Shiro Fukuda
- Departments of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Yuta Kondo
- Departments of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Taka-Aki Sakaue
- Departments of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masahito Iioka
- Departments of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Keita Fukuoka
- Departments of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Keitaro Kawada
- Departments of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hirofumi Nagao
- Departments of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshinari Obata
- Departments of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuya Fujishima
- Departments of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takeshi Ebihara
- Departments of Traumatology and Acute Critical Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hisatake Matsumoto
- Departments of Traumatology and Acute Critical Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Satoshi Nakagawa
- Departments of Obstetrics and Gynecology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tadashi Kimura
- Departments of Obstetrics and Gynecology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hitoshi Nishizawa
- Departments of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Iichiro Shimomura
- Departments of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
16
|
Matsui M, Lynch LE, Distefano I, Galante A, Gade AR, Wang HG, Gómez-Banoy N, Towers P, Sinden DS, Wei EQ, Barnett AS, Johnson K, Lima R, Rubio-Navarro A, Li AK, Marx SO, McGraw TE, Thornton PS, Timothy KW, Lo JC, Pitt GS. Multiple beta cell-independent mechanisms drive hypoglycemia in Timothy syndrome. Nat Commun 2024; 15:8980. [PMID: 39420001 PMCID: PMC11487186 DOI: 10.1038/s41467-024-52885-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
The canonical G406R mutation that increases Ca2+ influx through the CACNA1C-encoded CaV1.2 Ca2+ channel underlies the multisystem disorder Timothy syndrome (TS), characterized by life-threatening arrhythmias. Severe episodic hypoglycemia is among the poorly characterized non-cardiac TS pathologies. While hypothesized from increased Ca2+ influx in pancreatic beta cells and consequent hyperinsulinism, this hypoglycemia mechanism is undemonstrated because of limited clinical data and lack of animal models. We generated a CaV1.2 G406R knockin mouse model that recapitulates key TS features, including hypoglycemia. Unexpectedly, these mice do not show hyperactive beta cells or hyperinsulinism in the setting of normal intrinsic beta cell function, suggesting dysregulated glucose homeostasis. Patient data confirm the absence of hyperinsulinism. We discover multiple alternative contributors, including perturbed counterregulatory hormone responses with defects in glucagon secretion and abnormal hypothalamic control of glucose homeostasis. These data provide new insights into contributions of CaV1.2 channels and reveal integrated consequences of the mutant channels driving life-threatening events in TS.
Collapse
Affiliation(s)
- Maiko Matsui
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Lauren E Lynch
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Isabella Distefano
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Allison Galante
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Aravind R Gade
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Hong-Gang Wang
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Nicolas Gómez-Banoy
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Patrick Towers
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Daniel S Sinden
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Eric Q Wei
- Department of Medicine, MSRB II, 2 Genome Ct, Duke University Medical Center, Durham, NC, 27710, USA
| | - Adam S Barnett
- Department of Medicine, MSRB II, 2 Genome Ct, Duke University Medical Center, Durham, NC, 27710, USA
| | - Kenneth Johnson
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Renan Lima
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Alfonso Rubio-Navarro
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Ang K Li
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Steven O Marx
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 622 W 168th St, PH-3 Center, New York, NY, USA
- Department of Pharmacology, Vagelos College of Physicians and Surgeons, Columbia University, 622 W 168th St, PH-3 Center, New York, NY, USA
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medical College, 1300 York Ave, New York, NY, 10065, USA
| | - Paul S Thornton
- Division of Endocrinology and Diabetes, Cook Children's Medical Center, 801 7th Ave, Fort Worth, TX, 76104, USA
| | - Katherine W Timothy
- Children's Hospital Boston, Harvard Medical School, 300 Longwood Ave., Boston, MA, 02115, USA
| | - James C Lo
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Division of Cardiology, Department of Medicine, Weill Cornell Medicine, 413 E. 69th St, New York, NY, 10021, USA
| | - Geoffrey S Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA.
| |
Collapse
|
17
|
Homan EA, Gilani A, Rubio-Navarro A, Johnson MA, Schaepkens OM, Cortada E, de Lima RP, Stoll L, Lo JC. Complement 3a Receptor 1 on Macrophages and Kupffer cells is not required for the Pathogenesis of Metabolic Dysfunction-Associated Steatotic Liver Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.26.24309550. [PMID: 38978661 PMCID: PMC11230319 DOI: 10.1101/2024.06.26.24309550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Together with obesity and type 2 diabetes, metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing global epidemic. Activation of the complement system and infiltration of macrophages has been linked to progression of metabolic liver disease. The role of complement receptors in macrophage activation and recruitment in MASLD remains poorly understood. In human and mouse, C3AR1 in the liver is expressed primarily in Kupffer cells, but is downregulated in humans with MASLD compared to obese controls. To test the role of complement 3a receptor (C3aR1) on macrophages and liver resident macrophages in MASLD, we generated mice deficient in C3aR1 on all macrophages (C3aR1-MφKO) or specifically in liver Kupffer cells (C3aR1-KpKO) and subjected them to a model of metabolic steatotic liver disease. We show that macrophages account for the vast majority of C3ar1 expression in the liver. Overall, C3aR1-MφKO and C3aR1-KpKO mice have similar body weight gain without significant alterations in glucose homeostasis, hepatic steatosis and fibrosis, compared to controls on a MASLD-inducing diet. This study demonstrates that C3aR1 deletion in macrophages or Kupffer cells, the predominant liver cell type expressing C3aR1, has no significant effect on liver steatosis, inflammation or fibrosis in a dietary MASLD model.
Collapse
Affiliation(s)
- Edwin A. Homan
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Maya A. Johnson
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Odin M. Schaepkens
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Eric Cortada
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Renan Pereira de Lima
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - Lisa Stoll
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| | - James C. Lo
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, New York, 10021
| |
Collapse
|
18
|
Jiang Y, Guo JQ, Wu Y, Zheng P, Wang SF, Yang MC, Ma GS, Yao YY. Excessive or sustained endoplasmic reticulum stress: one of the culprits of adipocyte dysfunction in obesity. Ther Adv Endocrinol Metab 2024; 15:20420188241282707. [PMID: 39381518 PMCID: PMC11459521 DOI: 10.1177/20420188241282707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/22/2024] [Indexed: 10/10/2024] Open
Abstract
As the prevalence of obesity continues to rise globally, the research on adipocytes has attracted more and more attention. In the presence of nutrient overload, adipocytes are exposed to pressures such as hypoxia, inflammation, mechanical stress, metabolite, and oxidative stress that can lead to organelle dysfunction. Endoplasmic reticulum (ER) is a vital organelle for sensing cellular pressure, and its homeostasis is essential for maintaining adipocyte function. Under conditions of excess nutrition, ER stress (ERS) will be triggered by the gathering of abnormally folded proteins in the ER lumen, resulting in the activation of a signaling response known as the unfolded protein responses (UPRs), which is a response system to relieve ERS and restore ER homeostasis. However, if the UPRs fail to rescue ER homeostasis, ERS will activate pathways to damage cells. Studies have shown a role for disturbed activation of adipocyte ERS in the pathophysiology of obesity and its complications. Prolonged or excessive ERS in adipocytes can aggravate lipolysis, insulin resistance, and apoptosis and affect the bioactive molecule production. In addition, ERS also impacts the expression of some important genes. In view of the fact that ERS influences adipocyte function through various mechanisms, targeting ERS may be a viable strategy to treat obesity. This article summarizes the effects of ERS on adipocytes during obesity.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Jia-Qi Guo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Ya Wu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Peng Zheng
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Shao-Fan Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Meng-Chen Yang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Gen-Shan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Yu-Yu Yao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| |
Collapse
|
19
|
Kong Y, Wang N, Tong Z, Wang D, Wang P, Yang Q, Yan X, Song W, Jin Z, Zhang M. Role of complement factor D in cardiovascular and metabolic diseases. Front Immunol 2024; 15:1453030. [PMID: 39416783 PMCID: PMC11479899 DOI: 10.3389/fimmu.2024.1453030] [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: 06/22/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
In the genesis and progression of cardiovascular and metabolic diseases (CVMDs), adipose tissue plays a pivotal and dual role. Complement factor D (CFD, also known as adipsin), which is mainly produced by adipocytes, is the rate-limiting enzyme of the alternative pathway. Abnormalities in CFD generation or function lead to aberrant immune responses and energy metabolism. A large number of studies have revealed that CFD is associated with CVMDs. Herein, we will review the current studies on the function and mechanism of CFD in CVMDs such as hypertension, coronary heart disease, ischemia/reperfusion injury, heart failure, arrhythmia, aortic aneurysm, obesity, insulin resistance, and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yingjin Kong
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Naixin Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Zhonghua Tong
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Dongni Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Penghe Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Qiannan Yang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Xiangyu Yan
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Weijun Song
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Zexi Jin
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Maomao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| |
Collapse
|
20
|
Rivera-Gonzalez O, Mills MF, Konadu BD, Wilson NA, Murphy HA, Newberry MK, Hyndman KA, Garrett MR, Webb DJ, Speed JS. Adipocyte endothelin B receptor activation inhibits adiponectin production and causes insulin resistance in obese mice. Acta Physiol (Oxf) 2024; 240:e14214. [PMID: 39096077 PMCID: PMC11421981 DOI: 10.1111/apha.14214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024]
Abstract
AIMS Endothelin-1 (ET-1) is elevated in patients with obesity and adipose tissue of obese mice fed high-fat diet (HFD); however, its contribution to the pathophysiology of obesity is not fully understood. Genetic loss of endothelin type B receptors (ETB) improves insulin sensitivity in rats and leads to increased circulating adiponectin, suggesting that ETB activation on adipocytes may contribute to obesity pathophysiology. We hypothesized that elevated ET-1 in obesity promotes insulin resistance by reducing the secretion of insulin sensitizing adipokines, via ETB receptor. METHODS Male adipocyte-specific ETB receptor knockout (adETBKO), overexpression (adETBOX), or control littermates were fed either normal diet (NMD) or high-fat diet (HFD) for 8 weeks. RESULTS RNA-sequencing of epididymal adipose (eWAT) indicated differential expression of over 5500 genes (p < 0.05) in HFD compared to NMD controls, and changes in 1077 of these genes were attenuated in HFD adETBKO mice. KEGG analysis indicated significant increase in metabolic signaling pathway. HFD adETBKO mice had significantly improved glucose and insulin tolerance compared to HFD control. In addition, adETBKO attenuated changes in plasma adiponectin, insulin, and leptin that is observed in HFD versus NMD control mice. Treatment of primary adipocytes with ET-1 caused a reduction in adiponectin production that was attenuated in cells pretreated with an ETB antagonist. CONCLUSION These data indicate elevated ET-1 in adipose tissue of mice fed HFD inhibits adiponectin production and causes insulin resistance through activation of the ETB receptor on adipocytes.
Collapse
Affiliation(s)
- Osvaldo Rivera-Gonzalez
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216
| | - Megumi F. Mills
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216
| | - Bridget D. Konadu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216
| | - Natalie A. Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216
| | - Hayley A. Murphy
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216
| | - Madison K. Newberry
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216
| | - Kelly A. Hyndman
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham
| | - Michael R. Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216
| | - David J. Webb
- University/British Heart Foundation Centre for Cardiovascular Science|Queen’s Medical Research Institute, University of Edinburgh, UK
| | - Joshua S. Speed
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216
| |
Collapse
|
21
|
Huang P, Zhu Y, Qin J. Research advances in understanding crosstalk between organs and pancreatic β-cell dysfunction. Diabetes Obes Metab 2024; 26:4147-4164. [PMID: 39044309 DOI: 10.1111/dom.15787] [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: 06/18/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024]
Abstract
Obesity has increased dramatically worldwide. Being overweight or obese can lead to various conditions, including dyslipidaemia, hypertension, glucose intolerance and metabolic syndrome (MetS), which may further lead to type 2 diabetes mellitus (T2DM). Previous studies have identified a link between β-cell dysfunction and the severity of MetS, with multiple organs and tissues affected. Identifying the associations between pancreatic β-cell dysfunction and organs is critical. Research has focused on the interaction between the liver, gut and pancreatic β-cells. However, the mechanisms and related core targets are still not perfectly elucidated. The aims of this review were to summarize the mechanisms of β-cell dysfunction and to explore the potential pathogenic pathways and targets that connect the liver, gut, adipose tissue, muscle, and brain to pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yunling Zhu
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jian Qin
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
22
|
Mohammadyari E, Miwa T, Golla M, Song WC. Therapeutic targeting of factor D and MASP3 in complement-mediated diseases: Lessons learned from mouse studies. Eur J Immunol 2024; 54:e2350845. [PMID: 39540581 DOI: 10.1002/eji.202350845] [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: 03/27/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 11/16/2024]
Abstract
The alternative pathway (AP) plays a major role in many complement-mediated human diseases. Factor D (FD), a rate-limiting enzyme in AP complement activation, is an attractive therapeutic target. Unlike other complement proteins, FD is synthesized primarily in adipose tissue, and its levels in human blood are relatively low. However, because of FD's high turnover rate, therapeutic targeting with monoclonal antibodies and chemical inhibitors has been challenging. The recent discovery that FD activity is regulated by mannose-binding lectin-associated serine protease 3 (MASP3), through conversion of a zymogen to mature FD, has sparked interest in MASP3 inhibition as a new way to block FD function and AP complement activity. Here, we review studies of mouse models of FD and MASP3 inhibition. We additionally discuss the lessons learned from these studies and their implications for therapeutic targeting of human FD and MASP3.
Collapse
Affiliation(s)
- Eshagh Mohammadyari
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Takashi Miwa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Madhu Golla
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
23
|
Maity SK, Das Sharma A, Sarkar J, Chaudhuri T, Tantia O, Chakrabarti P. Adipose tissue-derived adipsin marks human aging in non-type 2 diabetes population. BMJ Open Diabetes Res Care 2024; 12:e004179. [PMID: 39209774 PMCID: PMC11367331 DOI: 10.1136/bmjdrc-2024-004179] [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: 03/08/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Adipsin or complement factor D is an adipokine that augments insulin secretion, is altered in various degrees of obesity, and is involved in alternative complement pathway. However, whether adipsin has any independent association with risk factors and biomarkers in patients with type 2 diabetes (T2D) remains elusive. RESEARCH DESIGN AND METHODS We performed an oral glucose tolerance test on a subset of 43 patients with T2D from the community health cohort to access the role of adipsin in insulin secretion. We further cross-sectionally examined the role of adipsin in plasma, adipose tissue (AT), and secretion in a community cohort of 353 subjects and a hospital cohort of 52 subjects. RESULTS We found that plasma adipsin has no significant correlation with insulin secretion in people with diabetes. Among the risk factors of T2D, adipsin levels were independently associated only with age, and a positive correlation between plasma adipsin and age among subjects without T2D was lost in patients with T2D. Plasma adipsin levels, AT adipsin expression, and secretion were upregulated both in T2D and aging, with a corresponding drop in Homeostatic Model Assessment for assessing β-cell function. Adipsin expression was positively associated with other aging biomarkers, such as β-galactosidase, p21, and p16. These results also corroborated with existing plasma proteomic signatures of aging, including growth, and differentiation factor-15, which strongly correlated with adipsin. CONCLUSIONS Our results demonstrate an increase in circulating adipsin in T2D and aging, and it scores as a candidate plasma marker for aging specifically in non-T2D population.
Collapse
Affiliation(s)
- Sujay Krishna Maity
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | | | - Jit Sarkar
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Tamonash Chaudhuri
- Department of Minimal Access & Bariatric Surgery, ILS Hospitals, Kolkata, India
| | - Om Tantia
- Department of Minimal Access & Bariatric Surgery, ILS Hospitals, Kolkata, India
| | - Partha Chakrabarti
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
24
|
Ghasemi Gojani E, Rai S, Norouzkhani F, Shujat S, Wang B, Li D, Kovalchuk O, Kovalchuk I. Targeting β-Cell Plasticity: A Promising Approach for Diabetes Treatment. Curr Issues Mol Biol 2024; 46:7621-7667. [PMID: 39057094 PMCID: PMC11275945 DOI: 10.3390/cimb46070453] [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: 06/24/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The β-cells within the pancreas play a pivotal role in insulin production and secretion, responding to fluctuations in blood glucose levels. However, factors like obesity, dietary habits, and prolonged insulin resistance can compromise β-cell function, contributing to the development of Type 2 Diabetes (T2D). A critical aspect of this dysfunction involves β-cell dedifferentiation and transdifferentiation, wherein these cells lose their specialized characteristics and adopt different identities, notably transitioning towards progenitor or other pancreatic cell types like α-cells. This process significantly contributes to β-cell malfunction and the progression of T2D, often surpassing the impact of outright β-cell loss. Alterations in the expressions of specific genes and transcription factors unique to β-cells, along with epigenetic modifications and environmental factors such as inflammation, oxidative stress, and mitochondrial dysfunction, underpin the occurrence of β-cell dedifferentiation and the onset of T2D. Recent research underscores the potential therapeutic value for targeting β-cell dedifferentiation to manage T2D effectively. In this review, we aim to dissect the intricate mechanisms governing β-cell dedifferentiation and explore the therapeutic avenues stemming from these insights.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.)
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.)
| |
Collapse
|
25
|
Zhang X, Tian L, Majumdar A, Scheller EL. Function and Regulation of Bone Marrow Adipose Tissue in Health and Disease: State of the Field and Clinical Considerations. Compr Physiol 2024; 14:5521-5579. [PMID: 39109972 PMCID: PMC11725182 DOI: 10.1002/cphy.c230016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Bone marrow adipose tissue (BMAT) is a metabolically and clinically relevant fat depot that exists within bone. Two subtypes of BMAT, regulated and constitutive, reside in hematopoietic-rich red marrow and fatty yellow marrow, respectively, and exhibit distinct characteristics compared to peripheral fat such as white and brown adipose tissues. Bone marrow adipocytes (BMAds) are evolutionally preserved in most vertebrates, start development after birth and expand throughout life, and originate from unique progenitor populations that control bone formation and hematopoiesis. Mature BMAds also interact closely with other cellular components of the bone marrow niche, serving as a nearby energy reservoir to support the skeletal system, a signaling hub that contributes to both local and systemic homeostasis, and a final fuel reserve for survival during starvation. Though BMAT and bone are often inversely correlated, more BMAT does not always mean less bone, and the prevention of BMAT expansion as a strategy to prevent bone loss remains questionable. BMAT adipogenesis and lipid metabolism are regulated by the nervous systems and a variety of circulating hormones. This contributes to the plasticity of BMAT, including BMAT expansion in common physiological or pathological conditions, and BMAT catabolism under certain extreme circumstances, which are often associated with malnutrition and/or systemic inflammation. Altogether, this article provides a comprehensive overview of the local and systemic functions of BMAT and discusses the regulation and plasticity of this unique adipose tissue depot in health and disease. © 2024 American Physiological Society. Compr Physiol 14:5521-5579, 2024.
Collapse
Affiliation(s)
- Xiao Zhang
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Linda Tian
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Anurag Majumdar
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
26
|
Sosibo AM, Mzimela NC, Ngubane PS, Khathi A. Hormone imbalances detected in study participants with pre-diabetes in a Durban-based clinical setting, South Africa. Int J Diabetes Dev Ctries 2024. [DOI: 10.1007/s13410-024-01363-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 05/29/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Type II diabetes mellitus onset is linked with hormonal imbalances. However, the knowledge about hormonal alterations in pre-diabetes is limited.
Objective
The study aimed to examine type II diabetes mellitus-associated hormone levels during the pre-diabetes phase in participants aged 25–45 in a Durban-based clinical setting in South Africa.
Methods
Stored plasma samples from a retrospective study collected 364 samples that were divided into pre-diabetes and non-pre-diabetes groups. From the 364, 38 samples from the group of persons without pre-diabetes and 38 from persons with glycated haemoglobin determined pre-diabetes were blindly selected. The hormone concentrations (C-peptide, cortisol, adipokines, thyroids, incretins, and sex steroids) of the study participants were measured using the BIO-RAD Bio-Plex MAGPIX instrument.
Results
Hormone imbalances in several hormones were detected in study participants with pre-diabetes. Most of the hormone dysregulation associated with T2DM begins in pre-diabetes but at a moderate level.
Conclusion
The findings reveal new possible hormone therapy targets for pre-diabetes and contribute to the growing support for targeting pre-diabetes as a preventative measure for T2DM prevention.
Collapse
|
27
|
Liu ZT, Yang GW, Zhao X, Dong SH, Jiao Y, Ge Z, Yu A, Zhang XQ, Xu XZ, Cheng ZQ, Zhang X, Wang KX. Growth hormone improves insulin resistance in visceral adipose tissue after duodenal-jejunal bypass by regulating adiponectin secretion. World J Diabetes 2024; 15:1340-1352. [PMID: 38983805 PMCID: PMC11229968 DOI: 10.4239/wjd.v15.i6.1340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/12/2024] [Accepted: 04/15/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND The mechanism of improvement of type 2 diabetes after duodenal-jejunal bypass (DJB) surgery is not clear. AIM To study the morphological and functional changes in adipose tissue after DJB and explore the potential mechanisms contributing to postoperative insulin sensitivity improvement of adipose tissue in a diabetic male rat model. METHODS DJB and sham surgery was performed in a-high-fat-diet/streptozotocin-induced diabetic rat model. All adipose tissue was weighed and observed under microscope. Use inguinal fat to represent subcutaneous adipose tissue (SAT) and mesangial fat to represent visceral adipose tissue. RNA-sequencing was utilized to evaluate gene expression alterations adipocytes. The hematoxylin and eosin staining, reverse transcription-quantitative polymerase chain reaction, western blot, and enzyme-linked immunosorbent assay were used to study the changes. Insulin resistance was evaluated by immunofluorescence. RESULTS After DJB, whole body blood glucose metabolism and insulin sensitivity in adipose tissue improved. Fat cell volume in both visceral adipose tissue (VAT) and SAT increased. Compared to SAT, VAT showed more significantly functional alterations after DJB and KEGG analysis indicated growth hormone (GH) pathway and downstream adiponectin secretion were involved in metabolic regulation. The circulating GH and adiponectin levels and GH receptor and adiponectin levels in VAT increased. Cytological experiment showed that GH stimulated adiponectin secretion and improve insulin sensitivity. CONCLUSION GH improves insulin resistance in VAT in male diabetic rats after receiving DJB, possibly by increasing adiponectin secretion.
Collapse
Affiliation(s)
- Zi-Tian Liu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Guang-Wei Yang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Xiang Zhao
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Shuo-Hui Dong
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Yang Jiao
- Department of General Surgery, Shandong University of Qilu Hospital (Qingdao), Qingdao 266000, Shandong Province, China
| | - Zheng Ge
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Ao Yu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Xi-Qiang Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Xin-Zhen Xu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Zhi-Qiang Cheng
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Xiang Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Ke-Xin Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
28
|
Yuan L, Wang T, Duan J, Zhou J, Li N, Li G, Zhou H. Expression Profiles and Bioinformatic Analysis of Circular RNAs in Db/Db Mice with Cardiac Fibrosis. Diabetes Metab Syndr Obes 2024; 17:2107-2120. [PMID: 38799279 PMCID: PMC11128257 DOI: 10.2147/dmso.s465588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Cardiac fibrosis is one of the important causes of heart failure and death in diabetic cardiomyopathy (DCM) patients. Circular RNAs (circRNAs) are covalently closed RNA molecules in eukaryotes and have high stability. Their role in myocardial fibrosis with diabetic cardiomyopathy (DCM) remain to be fully elucidated. This study aimed to understand the expression profiles of circRNAs in myocardial fibrosis with DCM, exploring the possible biomarkers and therapeutic targets for DCM. Methods At 21 weeks of age, db/db mice established the type 2 DCM model measured by echocardiography, and the cardiac tissue was extracted for Hematoxylin-eosin, Masson's trichrome staining, and transmission electron microscopy. Subsequently, the expression profile of circRNAs in myocardial fibrosis of db/db mice was constructed using microarray hybridization and verified by real-time quantitative polymerase chain reaction. A circRNA-microRNA-messenger RNA coexpression network was constructed, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis were done. Results Compared with normal control mice, db/db mice had 77 upregulated circRNAs and 135 downregulated circRNAs in their chromosomes (fold change ≥1.5, P ≤ 0.05). Moreover, the enrichment analysis of circRNA host genes showed that these differentially expressed circRNAs were mainly involved in mitogen-activated protein kinase signaling pathways. CircPHF20L1, circCLASP1, and circSLC8A1 were the key circRNAs. Moreover, circCLASP1/miR-182-5p/Wnt7a, circSLC8A1/miR-29b-1-5p/Col12a1, and most especially circPHF20L1/miR-29a-3p/Col6a2 might be three novel axes in the development of myocardial fibrosis in DCM. Conclusion The findings will provide some novel circRNAs and molecular pathways for the prevention or clinical treatment of DCM through intervention with specific circRNAs.
Collapse
Affiliation(s)
- Lingling Yuan
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Ting Wang
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Jinsheng Duan
- Department of Cardiology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Jing Zhou
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Na Li
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Guizhi Li
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| | - Hong Zhou
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050004, People’s Republic of China
| |
Collapse
|
29
|
Ma L, Gilani A, Rubio-Navarro A, Cortada E, Li A, Reilly SM, Tang L, Lo JC. Adipsin and adipocyte-derived C3aR1 regulate thermogenic fat in a sex-dependent fashion. JCI Insight 2024; 9:e178925. [PMID: 38713526 PMCID: PMC11382875 DOI: 10.1172/jci.insight.178925] [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: 12/27/2023] [Accepted: 04/26/2024] [Indexed: 05/09/2024] Open
Abstract
Thermogenesis in beige/brown adipose tissues can be leveraged to combat metabolic disorders such as type 2 diabetes and obesity. The complement system plays pleiotropic roles in metabolic homeostasis and organismal energy balance with canonical effects on immune cells and noncanonical effects on nonimmune cells. The adipsin/C3a/C3a receptor 1 (C3aR1) pathway stimulates insulin secretion and sustains pancreatic β cell mass. However, its role in adipose thermogenesis has not been defined. Here, we show that male Adipsin/Cfd-knockout mice exhibited increased energy expenditure and white adipose tissue (WAT) browning. In addition, male adipocyte-specific C3aR1-knockout mice exhibited enhanced WAT thermogenesis and increased respiration. In stark contrast, female adipocyte-specific C3aR1-knockout mice displayed decreased brown fat thermogenesis and were cold intolerant. Female mice expressed lower levels of Adipsin in thermogenic adipocytes and adipose tissues than males. C3aR1 was also lower in female subcutaneous adipose tissue than in males. Collectively, these results reveal sexual dimorphism in the adipsin/C3a/C3aR1 axis in regulating adipose thermogenesis and defense against cold stress. Our findings establish a potentially new role of the alternative complement pathway in adaptive thermogenesis and highlight sex-specific considerations in potential therapeutic targets for metabolic diseases.
Collapse
Affiliation(s)
- Lunkun Ma
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Eric Cortada
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Ang Li
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Shannon M Reilly
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - James C Lo
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
30
|
Li J, Fan Z, Chen H, Maria Da Costa E, Zhou X, Yu N. Development of a rapid and ultrasensitive magnetic chemiluminescence immunoassay for the detection of adiponectin and its clinical application. J Pharm Biomed Anal 2024; 241:115961. [PMID: 38237546 DOI: 10.1016/j.jpba.2024.115961] [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: 09/04/2023] [Revised: 12/26/2023] [Accepted: 01/01/2024] [Indexed: 02/21/2024]
Abstract
Adiponectin (ADPN), which serum/plasma adiponectin levels are closely associated with insulin resistance and type 2 diabetes, and lower adiponectin levels predict an increased risk of diabetes, is a strong indicator of diabetes risk in people at high risk of diabetes in different races. Using the unique principle and performance advantages of chemiluminescence immunoassay (CLIA), an ADPN-CLIA method with high sensitivity, high specificity and wide detection range was established based on the principle of two-steps method of sandwich-type, with the magnetic particles (MPs) as the solid phase carrier and acridinium ester (AE) as the chemiluminescence reaction system. The selection of the main raw materials required, the preparation conditions of MPs-coated antibodies, the methods of AE-labeled antibodies, sample requirements and reaction modes were optimized and evaluated. AE labeling experiment was successfully performed with the labeling efficiency of 8.366 and the antibody utilization rate of 96.8%. The chemiluminescent immunoassay for ADPN had a good linear relationship from 0 ng/mL to 250 ng/mL (R2 =0.9993), with the detection limit of 0.05 ng/mL. The coefficient of variation (CV) of intra-assay and inter-assay precision were both less than 5% respectively. The recovery rates for accuracy were from 91.26% to 107.46%. The comparison experiment of 80 clinical serum samples between the developed ADPN-CLIA with the immunoturbidimetry showed that the correlation coefficient was 0.956, and the Bland-Altman analysis showed that the limits of agreement were - 0.364 and 0.433.
Collapse
Affiliation(s)
- Jiexia Li
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, PR China; Department of Laboratory Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, PR China
| | - Zhuqiao Fan
- Guangzhou Biotron Technology Co., Ltd., Guangzhou 510530, PR China
| | - Hanqi Chen
- Department of Laboratory Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, PR China
| | - Ernestina Maria Da Costa
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Xiaomian Zhou
- Guangzhou Biotron Technology Co., Ltd., Guangzhou 510530, PR China.
| | - Nan Yu
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, PR China; Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, PR China.
| |
Collapse
|
31
|
Taneera J, Saber-Ayad MM. Preservation of β-Cells as a Therapeutic Strategy for Diabetes. Horm Metab Res 2024; 56:261-271. [PMID: 38387480 DOI: 10.1055/a-2239-2668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
The preservation of pancreatic islet β-cells is crucial in diabetes mellitus, encompassing both type 1 and type 2 diabetes. β-cell dysfunction, reduced mass, and apoptosis are central to insufficient insulin secretion in both types. Research is focused on understanding β-cell characteristics and the factors regulating their function to develop novel therapeutic approaches. In type 1 diabetes (T1D), β-cell destruction by the immune system calls for exploring immunosuppressive therapies, non-steroidal anti-inflammatory drugs, and leukotriene antagonists. Islet transplantation, stem cell therapy, and xenogeneic transplantation offer promising strategies for type 1 diabetes treatment. For type 2 diabetes (T2D), lifestyle changes like weight loss and exercise enhance insulin sensitivity and maintain β-cell function. Additionally, various pharmacological approaches, such as cytokine inhibitors and protein kinase inhibitors, are being investigated to protect β-cells from inflammation and glucotoxicity. Bariatric surgery emerges as an effective treatment for obesity and T2D by promoting β-cell survival and function. It improves insulin sensitivity, modulates gut hormones, and expands β-cell mass, leading to diabetes remission and better glycemic control. In conclusion, preserving β-cells offers a promising approach to managing both types of diabetes. By combining lifestyle modifications, targeted pharmacological interventions, and advanced therapies like stem cell transplantation and bariatric surgery, we have a significant chance to preserve β-cell function and enhance glucose regulation in diabetic patients.
Collapse
Affiliation(s)
- Jalal Taneera
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Maha M Saber-Ayad
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
32
|
Hatton-Jones KM, West NP, Thang MW, Chen PY, Davoren P, Cripps AW, Cox AJ. Gut Microbiome and Metabolic and Immune Indices in Males with or without Evidence of Metabolic Dysregulation. J Obes Metab Syndr 2024; 33:64-75. [PMID: 38508778 PMCID: PMC11000514 DOI: 10.7570/jomes23022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/23/2023] [Accepted: 11/30/2023] [Indexed: 03/22/2024] Open
Abstract
Background The contributions of the gut microbiota to obesity and metabolic disease represent a potentially modifiable factor that may explain variation in risk between individuals. This study aimed to explore relationships among microbial composition and imputed functional attributes, a range of soluble metabolic and immune indices, and gene expression markers in males with or without evidence of metabolic dysregulation (MetDys). Methods This case-control study included healthy males (n=15; 41.9±11.7 years; body mass index [BMI], 22.9±1.2 kg/m2) and males with evidence of MetDys (n=14; 46.6±10.0 years; BMI, 35.1±3.3 kg/m2) who provided blood and faecal samples for assessment of a range of metabolic and immune markers and microbial composition using 16S rRNA gene sequencing. Metagenomic functions were imputed from microbial sequence data for analysis. Results In addition to elevated values in a range of traditional metabolic, adipokine and inflammatory indices in the MetDys group, 23 immunomodulatory genes were significantly altered in the MetDys group. Overall microbial diversity did not differ between groups; however, a trend for a higher relative abundance of the Bacteroidetes (P=0.06) and a lower relative abundance of the Verrucomicrobia (P=0.09) phyla was noted in the MetDys group. Using both family- and genera-level classifications, a partial least square discriminant analysis revealed unique microbial signatures between the groups. Conclusion These findings confirm the need for ongoing investigations in human clinical cohorts to further resolve the relationships between the gut microbiota and metabolic and immune markers and risk for metabolic disease.
Collapse
Affiliation(s)
- Kyle M. Hatton-Jones
- School of Pharmacy and Medical Science, Griffith University, Southport, Australia
| | - Nicholas P. West
- School of Pharmacy and Medical Science, Griffith University, Southport, Australia
- Menzies Health Institute Queensland, Griffith University, Southport, Australia
| | - Mike W.C. Thang
- QCIF Facility for Advanced Bioinformatics, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | - Pin-Yen Chen
- School of Pharmacy and Medical Science, Griffith University, Southport, Australia
| | - Peter Davoren
- Diabetes and Endocrinology, Gold Coast University Hospital, Southport, Australia
| | - Allan W. Cripps
- Menzies Health Institute Queensland, Griffith University, Southport, Australia
- School of Medicine, Griffith University, Southport, Australia
| | - Amanda J. Cox
- School of Pharmacy and Medical Science, Griffith University, Southport, Australia
- Menzies Health Institute Queensland, Griffith University, Southport, Australia
| |
Collapse
|
33
|
Kulak K, Kuska K, Colineau L, Mckay M, Maziarz K, Slaby J, Blom AM, King BC. Intracellular C3 protects β-cells from IL-1β-driven cytotoxicity via interaction with Fyn-related kinase. Proc Natl Acad Sci U S A 2024; 121:e2312621121. [PMID: 38346191 PMCID: PMC10895342 DOI: 10.1073/pnas.2312621121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024] Open
Abstract
One of the hallmarks of type 1 but also type 2 diabetes is pancreatic islet inflammation, associated with altered pancreatic islet function and structure, if unresolved. IL-1β is a proinflammatory cytokine which detrimentally affects β-cell function. In the course of diabetes, complement components, including the central complement protein C3, are deregulated. Previously, we reported high C3 expression in human pancreatic islets, with upregulation after IL-1β treatment. In the current investigation, using primary human and rodent material and CRISPR/Cas9 gene-edited β-cells deficient in C3, or producing only cytosolic C3 from a noncanonical in-frame start codon, we report a protective effect of C3 against IL-1β-induced β-cell death, that is attributed to the cytosolic fraction of C3. Further investigation revealed that intracellular C3 alleviates IL-1β-induced β-cell death, by interaction with and inhibition of Fyn-related kinase (FRK), which is involved in the response of β-cells to cytokines. Furthermore, these data were supported by increased β-cell death in vivo in a β-cell-specific C3 knockout mouse. Our data indicate that a functional, cytoprotective association exists between FRK and cytosolic C3.
Collapse
Affiliation(s)
- Klaudia Kulak
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Katarzyna Kuska
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Lucie Colineau
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Marina Mckay
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Karolina Maziarz
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Julia Slaby
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Anna M Blom
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| | - Ben C King
- Section of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö 214-28, Sweden
| |
Collapse
|
34
|
Bradford BJ, Contreras GA. Adipose Tissue Inflammation: Linking Physiological Stressors to Disease Susceptibility. Annu Rev Anim Biosci 2024; 12:261-281. [PMID: 38064480 DOI: 10.1146/annurev-animal-021122-113212] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
The study of adipose tissue (AT) is enjoying a renaissance. White, brown, and beige adipocytes are being investigated in adult animals, and the critical roles of small depots like perivascular AT are becoming clear. But the most profound revision of the AT dogma has been its cellular composition and regulation. Single-cell transcriptomic studies revealed that adipocytes comprise well under 50% of the cells in white AT, and a substantial portion of the rest are immune cells. Altering the function of AT resident leukocytes can induce or correct metabolic syndrome and, more surprisingly, alter adaptive immune responses to infection. Although the field is dominated by obesity research, conditions such as rapid lipolysis, infection, and heat stress impact AT immune dynamics as well. Recent findings in rodents lead to critical questions that should be explored in domestic livestock as potential avenues for improved animal resilience to stressors, particularly as animals age.
Collapse
Affiliation(s)
- Barry J Bradford
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, Michigan, USA;
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA;
| |
Collapse
|
35
|
Shah R, Zhong J, Massier L, Tanriverdi K, Hwang SJ, Haessler J, Nayor M, Zhao S, Perry AS, Wilkins JT, Shadyab AH, Manson JE, Martin L, Levy D, Kooperberg C, Freedman JE, Rydén M, Murthy VL. Targeted Proteomics Reveals Functional Targets for Early Diabetes Susceptibility in Young Adults. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004192. [PMID: 38323454 PMCID: PMC10940209 DOI: 10.1161/circgen.123.004192] [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: 04/19/2023] [Accepted: 11/05/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND The circulating proteome may encode early pathways of diabetes susceptibility in young adults for surveillance and intervention. Here, we define proteomic correlates of tissue phenotypes and diabetes in young adults. METHODS We used penalized models and principal components analysis to generate parsimonious proteomic signatures of diabetes susceptibility based on phenotypes and on diabetes diagnosis across 184 proteins in >2000 young adults in the CARDIA (Coronary Artery Risk Development in Young Adults study; mean age, 32 years; 44% women; 43% Black; mean body mass index, 25.6±4.9 kg/m2), with validation against diabetes in >1800 individuals in the FHS (Framingham Heart Study) and WHI (Women's Health Initiative). RESULTS In 184 proteins in >2000 young adults in CARDIA, we identified 2 proteotypes of diabetes susceptibility-a proinflammatory fat proteotype (visceral fat, liver fat, inflammatory biomarkers) and a muscularity proteotype (muscle mass), linked to diabetes in CARDIA and WHI/FHS. These proteotypes specified broad mechanisms of early diabetes pathogenesis, including transorgan communication, hepatic and skeletal muscle stress responses, vascular inflammation and hemostasis, fibrosis, and renal injury. Using human adipose tissue single cell/nuclear RNA-seq, we demonstrate expression at transcriptional level for implicated proteins across adipocytes and nonadipocyte cell types (eg, fibroadipogenic precursors, immune and vascular cells). Using functional assays in human adipose tissue, we demonstrate the association of expression of genes encoding these implicated proteins with adipose tissue metabolism, inflammation, and insulin resistance. CONCLUSIONS A multifaceted discovery effort uniting proteomics, underlying clinical susceptibility phenotypes, and tissue expression patterns may uncover potentially novel functional biomarkers of early diabetes susceptibility in young adults for future mechanistic evaluation.
Collapse
Affiliation(s)
- Ravi Shah
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | - Jiawei Zhong
- Dept of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Lucas Massier
- Dept of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Kahraman Tanriverdi
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | - Shih-Jen Hwang
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Matthew Nayor
- Sections of Preventive Medicine & Epidemiology & Cardiovascular Medicine, Dept of Medicine, Dept of Epidemiology, Boston University Schools of Medicine & Public Health, Boston, MA & Framingham Heart Study, Framingham, MA
| | | | - Andrew S. Perry
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | | | - Aladdin H. Shadyab
- Herbert Wertheim School of Public Health & Human Longevity Science, Univ of California, San Diego, La Jolla, CA
| | - JoAnn E. Manson
- Dept of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Lisa Martin
- George Washington Univ School of Medicine & Health Sciences
| | - Daniel Levy
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Jane E. Freedman
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | - Mikael Rydén
- Dept of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
36
|
Gilani A, Stoll L, Homan EA, Lo JC. Adipose Signals Regulating Distal Organ Health and Disease. Diabetes 2024; 73:169-177. [PMID: 38241508 PMCID: PMC10796297 DOI: 10.2337/dbi23-0005] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/03/2023] [Indexed: 01/21/2024]
Abstract
Excessive adiposity in obesity is a significant risk factor for development of type 2 diabetes (T2D), nonalcoholic fatty liver disease, and other cardiometabolic diseases. An unhealthy expansion of adipose tissue (AT) results in reduced adipogenesis, increased adipocyte hypertrophy, adipocyte hypoxia, chronic low-grade inflammation, increased macrophage infiltration, and insulin resistance. This ultimately culminates in AT dysfunction characterized by decreased secretion of antidiabetic adipokines such as adiponectin and adipsin and increased secretion of proinflammatory prodiabetic adipokines including RBP4 and resistin. This imbalance in adipokine secretion alters the physiological state of AT communication with target organs including pancreatic β-cells, heart, and liver. In the pancreatic β-cells, adipokines are known to have a direct effect on insulin secretion, gene expression, cell death, and/or dedifferentiation. For instance, impaired secretion of adipsin, which promotes insulin secretion and β-cell identity, results in β-cell failure and T2D, thus presenting a potential druggable target to improve and/or preserve β-cell function. The cardiac tissue is affected by both the classic white AT-secreted adipokines and the newly recognized brown AT (BAT)-secreted BATokines or lipokines that alter lipid deposition and ventricular function. In the liver, adipokines affect hepatic gluconeogenesis, lipid accumulation, and insulin sensitivity, underscoring the importance of adipose-liver communication in the pathogenesis of nonalcoholic fatty liver disease. In this perspective, we outline what is currently known about the effects of individual adipokines on pancreatic β-cells, liver, and the heart.
Collapse
Affiliation(s)
- Ankit Gilani
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Lisa Stoll
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Edwin A. Homan
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - James C. Lo
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Department of Medicine, Weill Cornell Medicine, New York, NY
| |
Collapse
|
37
|
Wang X, Wang Y, Hou J, Liu H, Zeng R, Li X, Han M, Li Q, Ji L, Pan D, Jia W, Zhong W, Xu T. Plasma proteome profiling reveals the therapeutic effects of the PPAR pan-agonist chiglitazar on insulin sensitivity, lipid metabolism, and inflammation in type 2 diabetes. Sci Rep 2024; 14:638. [PMID: 38182717 PMCID: PMC10770401 DOI: 10.1038/s41598-024-51210-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024] Open
Abstract
Chiglitazar is a novel peroxisome proliferator-activated receptor (PPAR) pan-agonist, which passed phase III clinical trials and was newly approved in China for use as an adjunct to diet and exercise in glycemic control in adult patients with Type 2 Diabetes (T2D). To explore the circulating protein signatures associated with the administration of chiglitazar in T2D patients, we conducted a comparative longitudinal study using plasma proteome profiling. Of the 157 T2D patients included in the study, we administered chiglitazar to a specific group, while the controls were given either placebo or sitagliptin. The plasma proteomes were profiled at baseline and 12 and 24 weeks post-treatment using data-independent acquisition mass spectrometry (DIA-MS). Our study indicated that 13 proteins were associated with chiglitazar treatment in T2D patients, including 10 up-regulated proteins (SHBG, TF, APOA2, APOD, GSN, MBL2, CFD, PGLYRP2, A2M, and APOA1) and 3 down-regulated proteins (PRG4, FETUB, and C2) after treatment, which were implicated in the regulation of insulin sensitivity, lipid metabolism, and inflammation response. Our study provides insight into the response of chiglitazar treatment from a proteome perspective and demonstrates the multi-faceted effects of chiglitazar in T2D patients, which will help the clinical application of chiglitazar and further study of its action mechanism.
Collapse
Affiliation(s)
- Xingyue Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - You Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Junjie Hou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hongyang Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Rong Zeng
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xiangyu Li
- Guangzhou National Laboratory, Guangzhou, China
| | - Mei Han
- Guangzhou National Laboratory, Guangzhou, China
| | - Qingrun Li
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Desi Pan
- Shenzhen Chipscreen Biosciences Co., Ltd, Shenzhen, China
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wen Zhong
- Guangzhou National Laboratory, Guangzhou, China.
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- Guangzhou National Laboratory, Guangzhou, China.
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
38
|
Reed RM, Whyte MB, Goff LM. Cardiometabolic disease in Black African and Caribbean populations: an ethnic divergence in pathophysiology? Proc Nutr Soc 2023:1-11. [PMID: 38230432 DOI: 10.1017/s0029665123004895] [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: 01/18/2024]
Abstract
In the UK, populations of Black African and Caribbean (BAC) ethnicity suffer higher rates of cardiometabolic disease than White Europeans (WE). Obesity, leading to increased visceral adipose tissue (VAT) and intrahepatic lipid (IHL), has long been associated with cardiometabolic risk, driving insulin resistance and defective fatty acid/lipoprotein metabolism. These defects are compounded by a state of chronic low-grade inflammation, driven by dysfunctional adipose tissue. Emerging evidence has highlighted associations between central complement system components and adipose tissue, fatty acid metabolism and inflammation; it may therefore sit at the intersection of various cardiometabolic disease risk factors. However, increasing evidence suggests an ethnic divergence in pathophysiology, whereby current theories fail to explain the high rates of cardiometabolic disease in BAC populations. Lower fasting and postprandial TAG has been reported in BAC, alongside lower VAT and IHL deposition, which are paradoxical to the high rates of cardiometabolic disease exhibited by this ethnic group. Furthermore, BAC have been shown to exhibit a more anti-inflammatory profile, with lower TNF-α and greater IL-10. In contrast, recent evidence has revealed greater complement activation in BAC compared to WE, suggesting its dysregulation may play a greater role in the high rates of cardiometabolic disease experienced by this population. This review outlines the current theories of how obesity is proposed to drive cardiometabolic disease, before discussing evidence for ethnic differences in disease pathophysiology between BAC and WE populations.
Collapse
Affiliation(s)
- Reuben M Reed
- Department of Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London SE1 9NH, UK
| | - Martin B Whyte
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey GU2 7WG, UK
| | - Louise M Goff
- Leicester Diabetes Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
39
|
Khaledian B, Thibes L, Shimono Y. Adipocyte regulation of cancer stem cells. Cancer Sci 2023; 114:4134-4144. [PMID: 37622414 PMCID: PMC10637066 DOI: 10.1111/cas.15940] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 08/26/2023] Open
Abstract
Cancer stem cells (CSCs) are a highly tumorigenic subpopulation of the cancer cells within a tumor that drive tumor initiation, progression, and therapy resistance. In general, stem cell niche provides a specific microenvironment in which stem cells are present in an undifferentiated and self-renewable state. CSC niche is a specialized tumor microenvironment for CSCs which provides cues for their maintenance and propagation. However, molecular mechanisms for the CSC-niche interaction remain to be elucidated. We have revealed that adipsin (complement factor D) and its downstream effector hepatocyte growth factor are secreted from adipocytes and enhance the CSC properties in breast cancers in which tumor initiation and progression are constantly associated with the surrounding adipose tissue. Considering that obesity, characterized by excess adipose tissue, is associated with an increased risk of multiple cancers, it is reasonably speculated that adipocyte-CSC interaction is similarly involved in many types of cancers, such as pancreas, colorectal, and ovarian cancers. In this review, various molecular mechanisms by which adipocytes regulate CSCs, including secretion of adipokines, extracellular matrix production, biosynthesis of estrogen, metabolism, and exosome, are discussed. Uncovering the roles of adipocytes in the CSC niche will propose novel strategies to treat cancers, especially those whose progression is linked to obesity.
Collapse
Affiliation(s)
- Behnoush Khaledian
- Department of BiochemistryFujita Health University School of MedicineToyoakeAichiJapan
| | - Lisa Thibes
- Department of BiochemistryFujita Health University School of MedicineToyoakeAichiJapan
| | - Yohei Shimono
- Department of BiochemistryFujita Health University School of MedicineToyoakeAichiJapan
| |
Collapse
|
40
|
Kawakami E, Saiki N, Yoneyama Y, Moriya C, Maezawa M, Kawamura S, Kinebuchi A, Kono T, Funata M, Sakoda A, Kondo S, Ebihara T, Matsumoto H, Togami Y, Ogura H, Sugihara F, Okuzaki D, Kojima T, Deguchi S, Vallee S, McQuade S, Islam R, Natarajan M, Ishigaki H, Nakayama M, Nguyen CT, Kitagawa Y, Wu Y, Mori K, Hishiki T, Takasaki T, Itoh Y, Takayama K, Nio Y, Takebe T. Complement factor D targeting protects endotheliopathy in organoid and monkey models of COVID-19. Cell Stem Cell 2023; 30:1315-1330.e10. [PMID: 37802037 PMCID: PMC10575686 DOI: 10.1016/j.stem.2023.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 07/04/2023] [Accepted: 09/01/2023] [Indexed: 10/08/2023]
Abstract
COVID-19 is linked to endotheliopathy and coagulopathy, which can result in multi-organ failure. The mechanisms causing endothelial damage due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain elusive. Here, we developed an infection-competent human vascular organoid from pluripotent stem cells for modeling endotheliopathy. Longitudinal serum proteome analysis identified aberrant complement signature in critically ill patients driven by the amplification cycle regulated by complement factor B and D (CFD). This deviant complement pattern initiates endothelial damage, neutrophil activation, and thrombosis specific to organoid-derived human blood vessels, as verified through intravital imaging. We examined a new long-acting, pH-sensitive (acid-switched) antibody targeting CFD. In both human and macaque COVID-19 models, this long-acting anti-CFD monoclonal antibody mitigated abnormal complement activation, protected endothelial cells, and curtailed the innate immune response post-viral exposure. Collectively, our findings suggest that the complement alternative pathway exacerbates endothelial injury and inflammation. This underscores the potential of CFD-targeted therapeutics against severe viral-induced inflammathrombotic outcomes.
Collapse
Affiliation(s)
- Eri Kawakami
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Norikazu Saiki
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Yosuke Yoneyama
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Chiharu Moriya
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Mari Maezawa
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shuntaro Kawamura
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Akiko Kinebuchi
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tamaki Kono
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Masaaki Funata
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Ayaka Sakoda
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Shigeru Kondo
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Takeshi Ebihara
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hisatake Matsumoto
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Yuki Togami
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Fuminori Sugihara
- Core Instrumentation Facility, Immunology Frontier Research Center and Research Institute for Microbial Diseases, Osaka University, 3-3-1, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Disease, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takashi Kojima
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15, Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Sayaka Deguchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Sebastien Vallee
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA
| | - Susan McQuade
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA; BPS Biosciences Inc., 6405 Mira Mesa Blvd. Suite 100, San Diego, CA 92121, USA
| | - Rizwana Islam
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA
| | - Madhusudan Natarajan
- Rare Disease DDU, Takeda Pharmaceutical Company Ltd, 125 Binney Street, Cambridge, MA 02139, USA
| | - Hirohito Ishigaki
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Misako Nakayama
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Cong Thanh Nguyen
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Yoshinori Kitagawa
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Yunheng Wu
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Kensaku Mori
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Information Technology Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Research Center for Medical Bigdata, National Institute of Informatics, Tokyo 100-0003, Japan
| | - Takayuki Hishiki
- Kanagawa Prefectural Institute of Public Health, 1-3-1, Shimomachiya, Chigasaki, Kanagawa 253-0087, Japan; Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Tomohiko Takasaki
- Kanagawa Prefectural Institute of Public Health, 1-3-1, Shimomachiya, Chigasaki, Kanagawa 253-0087, Japan; Advanced Technology and Development Division, BML, INC, 1361-1, Matoba, Kawagoe-shi, Saitama 350-1101, Japan
| | - Yasushi Itoh
- Department of Pathology, Shiga University of Medical Science, Setatsukinowa, Otsu, Shiga 520-2192, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Yasunori Nio
- T-CiRA Discovery & Innovation, Takeda Pharmaceutical Company Ltd, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan.
| | - Takanori Takebe
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Organoid Medicine Project, T-CiRA Joint Program, 2-26-1, Muraoka-higashi, Fujisawa, Kanagawa 251-8555, Japan; Division of Gastroenterology, Hepatology and Nutrition & Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; The Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Communication Design Center, Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe) and Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
41
|
Kim J, Oh CM, Kim H. The Interplay of Adipokines and Pancreatic Beta Cells in Metabolic Regulation and Diabetes. Biomedicines 2023; 11:2589. [PMID: 37761031 PMCID: PMC10526203 DOI: 10.3390/biomedicines11092589] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
The interplay between adipokines and pancreatic beta cells, often referred to as the adipo-insular axis, plays a crucial role in regulating metabolic homeostasis. Adipokines are signaling molecules secreted by adipocytes that have profound effects on several physiological processes. Adipokines such as adiponectin, leptin, resistin, and visfatin influence the function of pancreatic beta cells. The reciprocal communication between adipocytes and beta cells is remarkable. Insulin secreted by beta cells affects adipose tissue metabolism, influencing lipid storage and lipolysis. Conversely, adipokines released from adipocytes can influence beta cell function and survival. Chronic obesity and insulin resistance can lead to the release of excess fatty acids and inflammatory molecules from the adipose tissue, contributing to beta cell dysfunction and apoptosis, which are key factors in developing type 2 diabetes. Understanding the complex interplay of the adipo-insular axis provides insights into the mechanisms underlying metabolic regulation and pathogenesis of metabolic disorders. By elucidating the molecular mediators involved in this interaction, new therapeutic targets and strategies may emerge to reduce the risk and progression of diseases, such as type 2 diabetes and its associated complications. This review summarizes the interactions between adipokines and pancreatic beta cells, and their roles in the pathogenesis of diabetes and metabolic diseases.
Collapse
Affiliation(s)
- Joon Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea;
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea;
| | - Hyeongseok Kim
- Department of Biochemistry, College of Medicine, Chungnam National University, Daejeon 35105, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35105, Republic of Korea
| |
Collapse
|
42
|
Lenz M, Schönbauer R, Stojkovic S, Lichtenauer M, Paar V, Gatterer C, Schukro C, Emich M, Fritzer-Szekeres M, Strametz-Juranek J, Sponder M. Long-term physical activity modulates adipsin and ANGPTL4 serum levels, a potential link to exercise-induced metabolic changes. Panminerva Med 2023; 65:292-302. [PMID: 34309331 DOI: 10.23736/s0031-0808.21.04382-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Within the presented prospective study, we aimed to illuminate the effect of long-term physical exercise on serum levels of adipsin (complement factor D) and angiopoietin-like 4 (ANGPTL4). Although past studies already outlined the effects of acute exercise, our trial design aimed to depict the development under long-term physical activity conditions. METHODS Ninety-eight participants were included in the study and were asked to perform eight months of moderate physical activity for at least 150 minutes/week and/or vigorous-intensity exercise for at least 75 minutes/week. According to initial performance and performance gain throughout the study period, four groups were formed and subsequently compared. Blood sampling for the determination of routine laboratory parameters was done at baseline, after 2, 6, and 8 months. Additionally, adipsin and ANGPTL4 serum levels were concurrently quantified using commercially available ELISA kits. RESULTS The study cohort consisted of 61.2% male participants with an average age of 49.3±6.7 years. Adipsin and ANGPTL4 were found to be strongly increased by long-term physical exercise. Participants displaying a performance gain of >2.9% throughout the study showed significantly increased serum levels of both biomarkers. CONCLUSIONS Serum levels of adipsin and ANGPTL4 were closely tied to the individual performance gain of the participating probands. An association of adipsin levels, initial performance, and serum triglycerides was found at baseline. Interestingly, this interrelationship was not detectable after eight months of physical training. This finding might indicate adipsin's involvement in linking triglyceride-balance to individual performance and energy demands in a homeostatic state.
Collapse
Affiliation(s)
- Max Lenz
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria -
- Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria -
| | - Robert Schönbauer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Stefan Stojkovic
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Michael Lichtenauer
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Vera Paar
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Constantin Gatterer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Christoph Schukro
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Michael Emich
- Austrian Federal Ministry of Defence, Austrian Armed Forces, Vienna, Austria
| | - Monika Fritzer-Szekeres
- Department of Medical-Chemical Laboratory Analysis, Medical University of Vienna, Vienna, Austria
| | | | - Michael Sponder
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
43
|
Pestel J, Blangero F, Watson J, Pirola L, Eljaafari A. Adipokines in obesity and metabolic-related-diseases. Biochimie 2023; 212:48-59. [PMID: 37068579 DOI: 10.1016/j.biochi.2023.04.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/03/2023] [Accepted: 04/13/2023] [Indexed: 04/19/2023]
Abstract
The discovery of leptin in the 1990s led to a reconsideration of adipose tissue (AT) as not only a fatty acid storage organ, but also a proper endocrine tissue. AT is indeed capable of secreting bioactive molecules called adipokines for white AT or batokines for brown/beige AT, which allow communication with numerous organs, especially brain, heart, liver, pancreas, and/or the vascular system. Adipokines exert pro or anti-inflammatory activities. An equilibrated balance between these two sets ensures homeostasis of numerous tissues and organs. During the development of obesity, AT remodelling leads to an alteration of its endocrine activity, with increased secretion of pro-inflammatory adipokines relative to the anti-inflammatory ones, as shown in the graphical abstract. Pro-inflammatory adipokines take part in the initiation of local and systemic inflammation during obesity and contribute to comorbidities associated to obesity, as detailed in the present review.
Collapse
Affiliation(s)
- Julien Pestel
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Ferdinand Blangero
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Julia Watson
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Luciano Pirola
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Assia Eljaafari
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France; Hospices Civils de Lyon: 2 quai des Célestins, 69001 Lyon, France.
| |
Collapse
|
44
|
Eid SA, O’Brien PD, Kretzler KH, Jang DG, Mendelson FE, Hayes JM, Carter A, Zhang H, Pennathur S, Brosius FC, Koubek EJ, Feldman EL. Dietary interventions improve diabetic kidney disease, but not peripheral neuropathy, in a db/db mouse model of type 2 diabetes. FASEB J 2023; 37:e23115. [PMID: 37490006 PMCID: PMC10372884 DOI: 10.1096/fj.202300354r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/05/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023]
Abstract
Patients with type 2 diabetes often develop the microvascular complications of diabetic kidney disease (DKD) and diabetic peripheral neuropathy (DPN), which decrease quality of life and increase mortality. Unfortunately, treatment options for DKD and DPN are limited. Lifestyle interventions, such as changes to diet, have been proposed as non-pharmacological treatment options for preventing or improving DKD and DPN. However, there are no reported studies simultaneously evaluating the therapeutic efficacy of varying dietary interventions in a type 2 diabetes mouse model of both DKD and DPN. Therefore, we compared the efficacy of a 12-week regimen of three dietary interventions, low carbohydrate, caloric restriction, and alternate day fasting, for preventing complications in a db/db type 2 diabetes mouse model by performing metabolic, DKD, and DPN phenotyping. All three dietary interventions promoted weight loss, ameliorated glycemic status, and improved DKD, but did not impact percent fat mass and DPN. Multiple regression analysis identified a negative correlation between fat mass and motor nerve conduction velocity. Collectively, our data indicate that these three dietary interventions improved weight and glycemic status and alleviated DKD but not DPN. Moreover, diets that decrease fat mass may be a promising non-pharmacological approach to improve DPN in type 2 diabetes given the negative correlation between fat mass and motor nerve conduction velocity.
Collapse
Affiliation(s)
- Stephanie A. Eid
- Department of Neurology, University of Michigan, Ann Arbor, MI 48103, USA
| | | | | | - Dae-Gyu Jang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Faye E. Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48103, USA
| | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Andrew Carter
- Department of Neurology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Hongyu Zhang
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48103, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48103, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Frank C. Brosius
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48103, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48103, USA
- Department of Medicine, University of Arizona, Tucson, AZ, 85721 USA
| | - Emily J. Koubek
- Department of Neurology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48103, USA
| |
Collapse
|
45
|
Luo L, Fan W, Qin J, Guo S, Xiao H, Tang Z. Pharmacological and Pathological Effects of Mulberry Leaf Extract on the Treatment of Type 1 Diabetes Mellitus Mice. Curr Issues Mol Biol 2023; 45:5403-5421. [PMID: 37504259 PMCID: PMC10378407 DOI: 10.3390/cimb45070343] [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: 06/07/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023] Open
Abstract
This study investigated the pharmacological and pathological effects of aqueous mulberry leaf extract on type 1 diabetes mellitus mice induced with an intraperitoneal injection of streptozotocin (STZ). Diabetic mice were randomized into six groups: control (normal group), model, metformin-treated mice, and high-dose, medium-dose, and low-dose mulberry. The mulberry-treated mice were divided into high-, medium-, and low-dose groups based on the various doses of aqueous mulberry leaf extract during gavage. The efficacy of the six-week intervention was evaluated by measuring levels of fasting plasma glucose, alkaline phosphatase, alanine aminotransferase, aspartate transaminase, blood urea nitrogen, gamma-glutamyl transferase, glucose, high-density lipoprotein cholesterol, lactate dehydrogenase, and low-density lipoprotein cholesterol and recording body weight. Results revealed that mulberry leaf extract exhibited an ideal hypoglycemic effect, and the high-dose group was the most affected. Histology analysis, glycogen staining and apoptosis detection were used to study the extract's effects on the liver, kidney, and pancreatic cells of diabetic mice, enabling the assessment of its effectiveness and complications on a clinical and theoretical basis. It was shown that a certain concentration of aqueous mulberry leaf extract repaired the islet cells of type 1 diabetes mellitus mice, promoting normal insulin secretion. Herein, it was confirmed that mulberry leaf could be used to develop new hypoglycemic drugs or functional health food with broad applicability.
Collapse
Affiliation(s)
- Liru Luo
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center for Rapeseed Oil Nutrition Health and Deep Development, Changsha 410128, China
| | - Wei Fan
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center for Rapeseed Oil Nutrition Health and Deep Development, Changsha 410128, China
| | - Jingping Qin
- Hunan Engineering Technology Research Center for Rapeseed Oil Nutrition Health and Deep Development, Changsha 410128, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Shiyin Guo
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center for Rapeseed Oil Nutrition Health and Deep Development, Changsha 410128, China
| | - Hang Xiao
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center for Rapeseed Oil Nutrition Health and Deep Development, Changsha 410128, China
| | - Zhonghai Tang
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center for Rapeseed Oil Nutrition Health and Deep Development, Changsha 410128, China
| |
Collapse
|
46
|
Xu ZH, Xiong CW, Miao KS, Yu ZT, Zhang JJ, Yu CL, Huang Y, Zhou XD. Adipokines regulate mesenchymal stem cell osteogenic differentiation. World J Stem Cells 2023; 15:502-513. [PMID: 37424950 PMCID: PMC10324509 DOI: 10.4252/wjsc.v15.i6.502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/26/2023] [Accepted: 04/24/2023] [Indexed: 06/26/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can differentiate into various tissue cell types including bone, adipose, cartilage, and muscle. Among those, osteogenic differentiation of MSCs has been widely explored in many bone tissue engineering studies. Moreover, the conditions and methods of inducing osteogenic differentiation of MSCs are continuously advancing. Recently, with the gradual recognition of adipokines, the research on their involvement in different pathophysiological processes of the body is also deepening including lipid metabolism, inflammation, immune regulation, energy disorders, and bone homeostasis. At the same time, the role of adipokines in the osteogenic differentiation of MSCs has been gradually described more completely. Therefore, this paper reviewed the evidence of the role of adipokines in the osteogenic differentiation of MSCs, emphasizing bone formation and bone regeneration.
Collapse
Affiliation(s)
- Zhong-Hua Xu
- Department of Orthopedics, Jintan Hospital Affiliated to Jiangsu University, Changzhou 213200, Jiangsu Province, China
| | - Chen-Wei Xiong
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
| | - Kai-Song Miao
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
| | - Zhen-Tang Yu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
| | - Jun-Jie Zhang
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
| | - Chang-Lin Yu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
| | - Yong Huang
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
| | - Xin-Die Zhou
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
- Department of Orthopedics, Gonghe County Hospital of Traditional Chinese Medicine, Hainan Tibetan Autonomous Prefecture 811800, Qinghai Province, China
| |
Collapse
|
47
|
Zhang X, Duan Y, Zhang X, Jiang M, Man W, Zhang Y, Wu D, Zhang J, Song X, Li C, Lin J, Sun D. Adipsin alleviates cardiac microvascular injury in diabetic cardiomyopathy through Csk-dependent signaling mechanism. BMC Med 2023; 21:197. [PMID: 37237266 DOI: 10.1186/s12916-023-02887-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Microvascular complications are associated with an overtly increased risk of adverse outcomes in patients with diabetes including coronary microvascular injury which manifested as disruption of adherens junctions between cardiac microvascular endothelial cells (CMECs). However, particular mechanism leading to diabetic coronary microvascular hyperpermeability remains elusive. METHODS Experimental diabetes was induced in mice with adipose tissue-specific Adipsin overexpression (AdipsinLSL/LSL-Cre) and their respective control (AdipsinLSL/LSL). In addition, cultured CMECs were subjected to high glucose/palmitic acid (HG + PA) treatment to simulate diabetes for a mechanistic approach. RESULTS The results showed that Adipsin overexpression significantly reduced cardiac microvascular permeability, preserved coronary microvascular integrity, and increased coronary microvascular density. Adipsin overexpression also attenuated cardiac dysfunction in diabetic mice. E/A ratio, an indicator of cardiac diastolic function, was improved by Adipsin. Adipsin overexpression retarded left ventricular adverse remodeling, enhanced LVEF, and improved cardiac systolic function. Adipsin-enriched exosomes were taken up by CMECs, inhibited CMECs apoptosis, and increased CMECs proliferation under HG + PA treatment. Adipsin-enriched exosomes also accelerated wound healing, rescued cell migration defects, and promoted tube formation in response to HG + PA challenge. Furthermore, Adipsin-enriched exosomes maintained adherens junctions at endothelial cell borders and reversed endothelial hyperpermeability disrupted by HG + PA insult. Mechanistically, Adipsin blocked HG + PA-induced Src phosphorylation (Tyr416), VE-cadherin phosphorylation (Tyr685 and Tyr731), and VE-cadherin internalization, thus maintaining CMECs adherens junctions integrity. LC-MS/MS analysis and co-immunoprecipitation analysis (Co-IP) unveiled Csk as a direct downstream regulator of Adipsin. Csk knockdown increased Src phosphorylation (Tyr416) and VE-cadherin phosphorylation (Tyr685 and Tyr731), while abolishing Adipsin-induced inhibition of VE-cadherin internalization. Furthermore, Csk knockdown counteracted Adipsin-induced protective effects on endothelial hyperpermeability in vitro and endothelial barrier integrity of coronary microvessels in vivo. CONCLUSIONS Together, these findings favor the vital role of Adipsin in the regulation of CMECs adherens junctions integrity, revealing its promises as a treatment target against diabetic coronary microvascular dysfunction. Graphical abstract depicting the mechanisms of action behind Adipsin-induced regulation of diabetic coronary microvascular dysfunction.
Collapse
Affiliation(s)
- Xuebin Zhang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yu Duan
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiao Zhang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wanrong Man
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Dexi Wu
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiye Zhang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xinglong Song
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
48
|
Goff LM, Davies K, Zelek WM, Kodosaki E, Hakim O, Lockhart S, O’Rahilly S, Morgan BP. Ethnic differences in complement system biomarkers and their association with metabolic health in men of Black African and White European ethnicity. Clin Exp Immunol 2023; 212:52-60. [PMID: 36722378 PMCID: PMC10081104 DOI: 10.1093/cei/uxad011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/18/2022] [Accepted: 01/31/2023] [Indexed: 02/02/2023] Open
Abstract
Inflammation plays a fundamental role in the development of several metabolic diseases, including obesity and type 2 diabetes (T2D); the complement system has been implicated in their development. People of Black African (BA) ethnicity are disproportionately affected by T2D and other metabolic diseases but the impact of ethnicity on the complement system has not been explored. We investigated ethnic differences in complement biomarkers and activation status between men of BA and White European (WE) ethnicity and explored their association with parameters of metabolic health. We measured a panel of 15 complement components, regulators, and activation products in fasting plasma from 89 BA and 96 WE men. Ethnic differences were statistically validated. Association of complement biomarkers with metabolic health indices (BMI, waist circumference, insulin resistance, and HbA1c) were assessed in the groups. Plasma levels of the key complement components C3 and C4, the regulators clusterin and properdin and the activation marker iC3b were significantly higher in BA compared to WE men after age adjustment, while FD levels were significantly lower. C3 and C4 levels positively correlated with some or all markers of metabolic dysfunction in both ethnic groups while FD was inversely associated with HbA1c in both groups, and clusterin and properdin were inversely associated with some markers of metabolic dysfunction only in the WE group. Our findings of increased levels of complement components and activation products in BA compared to WE men suggest differences in complement regulation that may impact susceptibility to poor metabolic health.
Collapse
Affiliation(s)
- L M Goff
- Department of Nutritional Sciences, School of Population & Life Course Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, UK
| | - K Davies
- Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff, UK
| | - W M Zelek
- Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff, UK
| | - E Kodosaki
- Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff, UK
| | - O Hakim
- Department of Nutritional Sciences, School of Population & Life Course Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, UK
- School of Life & Health Sciences, University of Roehampton, London, UK
| | - S Lockhart
- MRC Metabolic Diseases Unit & Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - S O’Rahilly
- MRC Metabolic Diseases Unit & Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - B P Morgan
- Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
49
|
Rajamanickam A, Venkataraman A, Kumar NP, Sasidaran R, Pandiarajan AN, Selvaraj N, Mittal R, Gowshika K, Putlibai S, Lakshan Raj S, Ramanan PV, Babu S. Alterations of adipokines, pancreatic hormones and incretins in acute and convalescent COVID-19 children. BMC Pediatr 2023; 23:156. [PMID: 37013538 PMCID: PMC10068212 DOI: 10.1186/s12887-023-03971-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND The Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), accountable for Coronavirus disease 2019 (COVID-19), may cause hyperglycemia and additional systemic complexity in metabolic parameters. It is unsure even if the virus itself causes type 1 or type 2 diabetes mellitus (T1DM or T2DM). Furthermore, it is still unclear whether even recuperating COVID-19 individuals have an increased chance to develop new-onset diabetes. METHODS We wanted to determine the impact of COVID-19 on the levels of adipokines, pancreatic hormones, incretins and cytokines in acute COVID-19, convalescent COVID-19 and control children through an observational study. We performed a multiplex immune assay analysis and compared the plasma levels of adipocytokines, pancreatic hormones, incretins and cytokines of children presenting with acute COVID-19 infection and convalescent COVID-19. RESULTS Acute COVID-19 children had significantly elevated levels of adipsin, leptin, insulin, C-peptide, glucagon and ghrelin in comparison to convalescent COVID-19 and controls. Similarly, convalescent COVID-19 children had elevated levels of adipsin, leptin, insulin, C-peptide, glucagon, ghrelin and Glucagon-like peptide-1 (GLP-1) in comparison to control children. On the other hand, acute COVID-19 children had significantly decreased levels of adiponectin and Gastric Inhibitory Peptide (GIP) in comparison to convalescent COVID-19 and controls. Similarly, convalescent COVID-19 children had decreased levels of adiponectin and GIP in comparison to control children. Acute COVID-19 children had significantly elevated levels of cytokines, (Interferon (IFN)) IFNγ, Interleukins (IL)-2, TNFα, IL-1α, IL-1β, IFNα, IFNβ, IL-6, IL-12, IL-17A and Granulocyte-Colony Stimulating Factors (G-CSF) in comparison to convalescent COVID-19 and controls. Convalescent COVID-19 children had elevated levels of IFNγ, IL-2, TNFα, IL-1α, IL-1β, IFNα, IFNβ, IL-6, IL-12, IL-17A and G-CSF in comparison to control children. Additionally, Principal component Analysis (PCA) analysis distinguishes acute COVID-19 from convalescent COVID-19 and controls. The adipokines exhibited a significant correlation with the levels of pro-inflammatory cytokines. CONCLUSION Children with acute COVID-19 show significant glycometabolic impairment and exaggerated cytokine responses, which is different from convalescent COVID-19 infection and controls.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- National Institutes of Health-National Institute for Research in Tuberculosis - International Center for Excellence in Research, Chennai, India.
| | | | | | - R Sasidaran
- Kanchi Kamakoti CHILDS Trust Hospital, Chennai, India
| | - Arul Nancy Pandiarajan
- National Institutes of Health-National Institute for Research in Tuberculosis - International Center for Excellence in Research, Chennai, India
| | - Nandhini Selvaraj
- National Institutes of Health-National Institute for Research in Tuberculosis - International Center for Excellence in Research, Chennai, India
| | - Ruchi Mittal
- Sri Ramachandra Institute of Higher Education & Research, Chennai, India
| | - K Gowshika
- Sri Ramachandra Institute of Higher Education & Research, Chennai, India
| | | | - S Lakshan Raj
- Kanchi Kamakoti CHILDS Trust Hospital, Chennai, India
| | | | - Subash Babu
- National Institutes of Health-National Institute for Research in Tuberculosis - International Center for Excellence in Research, Chennai, India
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
50
|
Rubio-Navarro A, Gómez-Banoy N, Stoll L, Dündar F, Mawla AM, Ma L, Cortada E, Zumbo P, Li A, Reiterer M, Montoya-Oviedo N, Homan EA, Imai N, Gilani A, Liu C, Naji A, Yang B, Chong ACN, Cohen DE, Chen S, Cao J, Pitt GS, Huising MO, Betel D, Lo JC. A beta cell subset with enhanced insulin secretion and glucose metabolism is reduced in type 2 diabetes. Nat Cell Biol 2023; 25:565-578. [PMID: 36928765 PMCID: PMC10449536 DOI: 10.1038/s41556-023-01103-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 02/02/2023] [Indexed: 03/18/2023]
Abstract
The pancreatic islets are composed of discrete hormone-producing cells that orchestrate systemic glucose homeostasis. Here we identify subsets of beta cells using a single-cell transcriptomic approach. One subset of beta cells marked by high CD63 expression is enriched for the expression of mitochondrial metabolism genes and exhibits higher mitochondrial respiration compared with CD63lo beta cells. Human and murine pseudo-islets derived from CD63hi beta cells demonstrate enhanced glucose-stimulated insulin secretion compared with pseudo-islets from CD63lo beta cells. We show that CD63hi beta cells are diminished in mouse models of and in humans with type 2 diabetes. Finally, transplantation of pseudo-islets generated from CD63hi but not CD63lo beta cells into diabetic mice restores glucose homeostasis. These findings suggest that loss of a specific subset of beta cells may lead to diabetes. Strategies to reconstitute or maintain CD63hi beta cells may represent a potential anti-diabetic therapy.
Collapse
Affiliation(s)
- Alfonso Rubio-Navarro
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Excellence Research Unit "Modeling Nature" (MNat), CTS-963-Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain
| | - Nicolás Gómez-Banoy
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lisa Stoll
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Friederike Dündar
- Department of Physiology and Biophysics, Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Alex M Mawla
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Lunkun Ma
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Eric Cortada
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Paul Zumbo
- Department of Physiology and Biophysics, Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Ang Li
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Moritz Reiterer
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Nathalia Montoya-Oviedo
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Lipids and Diabetes Laboratory, Department of Physiological Sciences, Faculty of Medicine, National University of Colombia, Bogotá, Colombia
| | - Edwin A Homan
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Norihiro Imai
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Ankit Gilani
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Chengyang Liu
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Boris Yang
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - David E Cohen
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Geoffrey S Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
- Department of Physiology and Membrane Biology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Doron Betel
- Department of Physiology and Biophysics, Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Division of Hematology and Medical Oncology, Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - James C Lo
- Weill Center for Metabolic Health, Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|