1
|
Paul S, Chandran R, Vijayan DK, Bhardwaj J, Singh P, Shetty P, Cheruku S, Meleveetil S, Balachandran Krishnamma B. A Cdk5 inhibitor restores cognitive function and alleviates type 2 diabetes in mice. iScience 2025; 28:112200. [PMID: 40224020 PMCID: PMC11986975 DOI: 10.1016/j.isci.2025.112200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/29/2025] [Accepted: 03/06/2025] [Indexed: 04/15/2025] Open
Abstract
Type 2 diabetes (T2D) is a metabolic disorder commonly linked with cognitive decline, increasing patients' susceptibility to dementia. Alzheimer's disease (AD) has a strong connection with hyperglycemia and insulin dysregulation. Interestingly, certain anti-diabetic drugs have shown potential in reducing T2D-induced cognitive impairment. Previous studies, including ours, have highlighted the dysregulation of cyclin-dependent kinase 5 (Cdk5) activity in both T2D and AD, which may contribute to pathological changes in these conditions. Thus, targeting the Cdk5 kinase could offer a therapeutic approach for T2D and cognitive deterioration. Our research identifies Cdk5 as a key link between T2D and cognitive decline. By screening the KINACore library, we discovered two new brain-penetrant Cdk5 inhibitors, BLINK11 and BLINK15. In a high-fat diet-induced T2D model, these inhibitors improved blood glucose levels, obesity, and cognitive function. BLINK11, in particular, shows promise as a therapeutic candidate for treating cognitive impairment associated with T2D.
Collapse
Affiliation(s)
- Sangita Paul
- CSIR Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Remya Chandran
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Dileep K. Vijayan
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Juhi Bhardwaj
- CSIR Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Praveen Singh
- CSIR Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Poornima Shetty
- Srinivasa Engineering College, Mukka, Mangalore 574146, India
| | - Srinivas Cheruku
- Department of Chemistry, Manasa Gangotri, Mysore University, Mysuru 570005, India
| | - Sajith Meleveetil
- Department of Chemistry, SSIT, Sri Siddhartha Academy of Higher Education, Tumkur 572107, Karnataka, India
| | - Binukumar Balachandran Krishnamma
- CSIR Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
2
|
Tilg H, Ianiro G, Gasbarrini A, Adolph TE. Adipokines: masterminds of metabolic inflammation. Nat Rev Immunol 2025; 25:250-265. [PMID: 39511425 DOI: 10.1038/s41577-024-01103-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Adipose tissue is an immunologically active organ that controls host physiology, partly through the release of mediators termed adipokines. In obesity, adipocytes and infiltrating leukocytes produce adipokines, which include the hormones adiponectin and leptin and cytokines such as tumour necrosis factor and IL-1β. These adipokines orchestrate immune responses that are collectively referred to as metabolic inflammation. Consequently, metabolic inflammation characterizes metabolic disorders and promotes distinct disease aspects, such as insulin resistance, metabolic dysfunction-associated liver disease and cardiovascular complications. In this unifying concept, adipokines participate in the immunological cross-talk that occurs between metabolically active organs in metabolic diseases, highlighting the fundamental role of adipokines in obesity and their potential for therapeutic intervention. Here, we summarize how adipokines shape metabolic inflammation in mice and humans, focusing on their contribution to metabolic disorders in the setting of obesity and discussing their value as therapeutic targets.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Gianluca Ianiro
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
3
|
Wan D, Lee JE, Park YK, Maisto S, Agyapong C, Ozato K, Gavrilova O, Ge K. Histone chaperone HIRA facilitates transcription elongation to regulate insulin sensitivity and obesity-associated adipose expansion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644577. [PMID: 40196683 PMCID: PMC11974756 DOI: 10.1101/2025.03.21.644577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Adipose tissue is essential for maintaining glucose and lipid homeostasis in mammals. The histone chaperone HIRA has been reported to play a lineage- and stage-selective role during development. However, its role in adipose tissue development and function as well as its working mechanism remain unknown. Here we show that tissue-specific knockout of histone chaperone HIRA in mice impairs insulin sensitivity and alleviates adipose tissue expansion during high-fat diet-induced obesity, but only moderately affects embryonic development of adipose tissue. Mechanistically, HIRA is selectively required for expression of genes critical for insulin response and lipogenesis, rather than adipogenesis, in adipose tissue. By acute depletion of HIRA protein and by mapping HIRA genomic localization in adipocytes, we demonstrate that HIRA binds to promoters and enhancers of insulin response and lipogenesis genes and regulates their expression by facilitating transcription elongation. Our findings not only identify HIRA as an epigenomic regulator of insulin sensitivity, lipogenesis, and obesity-associated adipose expansion, but also reveal a novel mechanism by which HIRA regulates transcription. Bullet points HIRA depletion reduces expression of insulin response and lipogenesis genes in adipose tissueHIRA depletion alleviates adipose tissue expansion during high-fat diet-induced obesityHIRA targets insulin response and lipogenesis genes in adipocytesAcute depletion of HIRA impairs transcription elongation on target genes.
Collapse
|
4
|
Goli SH, Lim JY, Basaran-Akgul N, Templeton SP. Adiponectin pathway activation dampens inflammation and enhances alveolar macrophage fungal killing via LC3-associated phagocytosis. PLoS Pathog 2025; 21:e1012363. [PMID: 40096083 PMCID: PMC11949351 DOI: 10.1371/journal.ppat.1012363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 03/27/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
Although innate immunity is critical for antifungal host defense against the human opportunistic fungal pathogen Aspergillus fumigatus, potentially damaging inflammation must be controlled. Adiponectin (APN) is an adipokine produced mainly in adipose tissue that exerts anti-inflammatory effects in adipose-distal tissues such as the lung. We observed increased mortality and increased fungal burden and inflammation in neutropenic mice with invasive aspergillosis (IA) that lack APN or the APN receptors AdipoR1 or AdipoR2. Alveolar macrophages (AMs), early immune sentinels that detect and respond to lung infection, express both receptors, and APN-deficient AMs exhibited an inflammatory phenotype that was associated with decreased fungal killing. Pharmacological stimulation of AMs with AdipoR agonist AdipoRon rescued deficient killing in APN-/- AMs and was dependent on the presence of either receptor. Finally, APN-enhanced fungal killing was associated with increased activation of the non-canonical LC3 pathway of autophagy. Thus, our study identifies a novel role for APN in LC3-mediated killing of A.fumigatus.
Collapse
Affiliation(s)
- Sri Harshini Goli
- Department of Microbiology and Immunology, Indiana University School of Medicine-Terre Haute, Terre Haute, Indiana, United States of America
- Department of Biology, Indiana State University, Terre Haute, Indiana, United States of America
| | - Joo-Yeon Lim
- Department of Microbiology and Immunology, Indiana University School of Medicine-Terre Haute, Terre Haute, Indiana, United States of America
| | - Nese Basaran-Akgul
- Department of Microbiology and Immunology, Indiana University School of Medicine-Terre Haute, Terre Haute, Indiana, United States of America
| | - Steven P. Templeton
- Department of Microbiology and Immunology, Indiana University School of Medicine-Terre Haute, Terre Haute, Indiana, United States of America
| |
Collapse
|
5
|
Kim MJ, Kim SW, Ha B, Kim HS, Kwon SH, Jin J, Choi YK, Park KG, Kim JG, Lee IK, Jeon JH. Persistent influence of past obesity on current adiponectin levels and mortality in patients with type 2 diabetes. Korean J Intern Med 2025; 40:299-309. [PMID: 40102712 PMCID: PMC11938665 DOI: 10.3904/kjim.2024.221] [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: 06/22/2024] [Revised: 10/04/2024] [Accepted: 10/28/2024] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND/AIMS Adiponectin, a hormone primarily produced by adipocytes, typically shows an inverse relationship with body mass index (BMI). However, some studies have reported a positive correlation between the two. Thus, this study aimed to examine the relationship between adiponectin level and BMI in diabetic patients, focusing on the impact of past obesity on current adiponectin levels. METHODS We conducted an observational study analyzing data from 323 diabetic patients at Kyungpook National University Hospital. Based on past and current BMIs, participants were categorized into never-obese (nn, n = 106), previously obese (on, n = 43), and persistently obese (oo, n = 73) groups based on a BMI threshold of 25 kg/m2. Adiponectin level and BMI were key variables. Kaplan-Meier analysis assessed their impact on all-cause mortality up to August 2023, with survival differences based on adiponectin quartiles and follow-up starting from patient enrollment (2010-2015). RESULTS The analysis revealed a significant inverse correlation between adiponectin level and past maximum BMI. The on group exhibited approximately 10% lower adiponectin levels compared to the nn group. This association remained significant after adjusting for current BMI, age, and sex, highlighting the lasting influence of previous obesity on adiponectin levels. Furthermore, survival analysis indicated that patients in the lowest adiponectin quartile had reduced survival, with a statistically significant trend (p = 0.062). CONCLUSION Findings of this study suggest that lower adiponectin levels, potentially reflecting past obesity, are associated with decreased survival in diabetic patients, underscoring a critical role of adiponectin in long-term health outcomes.
Collapse
Affiliation(s)
- Min-Ji Kim
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu,
Korea
| | - Sung-Woo Kim
- Department of Internal Medicine, Daegu Catholic University Hospital, Daegu Catholic University School of Medicine, Daegu,
Korea
| | - Bitna Ha
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu,
Korea
| | - Hyang Sook Kim
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu,
Korea
| | - So-Hee Kwon
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu,
Korea
| | - Jonghwa Jin
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu,
Korea
| | - Yeon-Kyung Choi
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu,
Korea
| | - Keun-Gyu Park
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu,
Korea
| | - Jung Guk Kim
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu,
Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu,
Korea
| | - Jae-Han Jeon
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu,
Korea
| |
Collapse
|
6
|
Luo Z, Wu A, Robson S, Alper SL, Yu W. Adiponectin signaling regulates urinary bladder function by blunting smooth muscle purinergic contractility. JCI Insight 2025; 10:e188780. [PMID: 39989457 PMCID: PMC11949013 DOI: 10.1172/jci.insight.188780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/08/2025] [Indexed: 02/25/2025] Open
Abstract
Lower urinary tract symptoms (LUTS) affect approximately 50% of the population over 40 years of age and are strongly associated with obesity and metabolic syndrome. Adipose tissue plays a key role in obesity/metabolic syndrome by releasing adipokines that regulate systemic energy/lipid metabolism, insulin resistance, and inflammation. Adiponectin (ADPN), the most abundant adipokine, modulates energy/metabolism homeostasis through its insulin-sensitizing and antiinflammatory effects. Human plasma ADPN levels are inversely associated with obesity and diabetes. To the best of our knowledge, the role of adipokines such as ADPN in the LUTS associated with obesity/metabolic syndrome remains unknown. We have tested such a possible role in a global ADPN-knockout (Adpn-/-) mouse model. Adpn-/- mice exhibited increased voiding frequency, small voids, and reduced bladder smooth muscle (BSM) contractility, with absence of purinergic contraction. Molecular examination indicated significantly altered metabolic and purinergic pathways. The ADPN receptor agonist AdipoRon was found to abolish acute BSM contraction. Intriguingly, both AMPK activators and inhibitors also abolished BSM purinergic contraction. These data indicate the important contribution of what we believe is a novel ADPN signaling pathway to the regulation of BSM contractility. Dysregulation of this ADPN signaling pathway might be an important mechanism leading to LUTS associated with obesity/metabolic syndrome.
Collapse
Affiliation(s)
| | | | - Simon Robson
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Anesthesia, Harvard Medical School, Boston, Massachusetts, USA
| | - Seth L. Alper
- Division of Nephrology
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Weiqun Yu
- Division of Nephrology
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Braun LM, Giesler S, Andrieux G, Riemer R, Talvard-Balland N, Duquesne S, Rückert T, Unger S, Kreutmair S, Zwick M, Follo M, Hartmann A, Osswald N, Melchinger W, Chapman S, Hutchinson JA, Haferkamp S, Torster L, Kött J, Gebhardt C, Hellwig D, Karantzelis N, Wallrabenstein T, Lowinus T, Yücel M, Brehm N, Rawluk J, Pfeifer D, Bronsert P, Rogg M, Mattern S, Heikenwälder M, Fusco S, Malek NP, Singer S, Schmitt-Graeff A, Ceteci F, Greten FR, Blazar BR, Boerries M, Köhler N, Duyster J, Ihorst G, Lassmann S, Keye P, Minguet S, Schadendorf D, Ugurel S, Rafei-Shamsabadi D, Thimme R, Hasselblatt P, Bengsch B, Schell C, Pearce EL, Meiss F, Becher B, Funke-Lorenz C, Placke JM, Apostolova P, Zeiser R. Adiponectin reduces immune checkpoint inhibitor-induced inflammation without blocking anti-tumor immunity. Cancer Cell 2025; 43:269-291.e19. [PMID: 39933899 DOI: 10.1016/j.ccell.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/30/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025]
Abstract
Immune-related adverse events (irAEs) in cancer patients receiving immune checkpoint inhibitors (ICIs) cause morbidity and necessitate cessation of treatment. Comparing irAE treatments, we find that anti-tumor immunity is preserved in mice after extracorporeal photopheresis (ECP) but reduced with glucocorticosteroids, TNFα blockade, and α4β7-integrin inhibition. Local adiponectin production elicits a tissue-specific effect by reducing pro-inflammatory T cell frequencies in the colon while sparing tumor-specific T cell development. A prospective phase-1b/2 trial (EudraCT-No.2021-002073-26) with 14 patients reveals low ECP-related toxicity. Overall response rate for all irAEs is 92% (95% confidence interval [CI]: 63.97%-99.81%); colitis-specific complete remission rate is 100% (95% CI: 63.06%-100%). Glucocorticosteroid dosages could be reduced for all patients after ECP therapy. The ECP-adiponectin axis reduces intestinal tissue-resident memory T cell activation and CD4+IFN-γ+ T cells in patients with ICI-induced colitis without evidence of loss of anti-tumor immunity. In conclusion, we identify adiponectin as an immunomodulatory molecule that controls ICI-induced irAEs without blocking anti-tumor immunity.
Collapse
Affiliation(s)
- Lukas M Braun
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Sophie Giesler
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Roxane Riemer
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nana Talvard-Balland
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sandra Duquesne
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tamina Rückert
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Susanne Unger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Stefanie Kreutmair
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Melissa Zwick
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Marie Follo
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alina Hartmann
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Natascha Osswald
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang Melchinger
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefanie Chapman
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - James A Hutchinson
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Leopold Torster
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Kött
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dirk Hellwig
- Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Nikolaos Karantzelis
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Till Wallrabenstein
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Theresa Lowinus
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mehtap Yücel
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Niklas Brehm
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Justyna Rawluk
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Bronsert
- Institute of Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Manuel Rogg
- Institute of Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sven Mattern
- Institute of Pathology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; M3 Research Center, Eberhard Karls University Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Stefano Fusco
- Medizinische Klinik I, Uniklinik Tübingen, Tübingen, Germany
| | - Nisar P Malek
- Medizinische Klinik I, Uniklinik Tübingen, Tübingen, Germany
| | - Stephan Singer
- Institute of Pathology, University Hospital Tübingen, 72076 Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | | | - Fatih Ceteci
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt/Main, Germany
| | - Florian R Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt/Main, Germany
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, a Partnership Between DKFZ and Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - Natalie Köhler
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Justus Duyster
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, a Partnership Between DKFZ and Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - Gabriele Ihorst
- Clinical Trials Unit, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Silke Lassmann
- Institute of Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philip Keye
- Eye Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Susana Minguet
- Signalling Research Centres BIOSS and CIBSS, Freiburg. Germany. Department of Synthetic Immunology, Faculty of Biology and Centre for Chronic Immunodeficiency (CCI), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany, and German Cancer Consortium (DKTK), Partner Site Essen/Duesseldorf, Essen, Germany; National Center for Tumor Diseases (NCT)-West, Campus Essen, & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany, and German Cancer Consortium (DKTK), Partner Site Essen/Duesseldorf, Essen, Germany
| | - David Rafei-Shamsabadi
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Thimme
- Department of Internal Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Hasselblatt
- Department of Internal Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bertram Bengsch
- German Cancer Consortium (DKTK), Partner Site Freiburg, a Partnership Between DKFZ and Medical Center - University of Freiburg, Freiburg im Breisgau, Germany; Department of Internal Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Christoph Schell
- Institute of Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Erika L Pearce
- Department of Oncology, The Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Frank Meiss
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Carolin Funke-Lorenz
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany, and German Cancer Consortium (DKTK), Partner Site Essen/Duesseldorf, Essen, Germany
| | - Jan-Malte Placke
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany, and German Cancer Consortium (DKTK), Partner Site Essen/Duesseldorf, Essen, Germany
| | - Petya Apostolova
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, a Partnership Between DKFZ and Medical Center - University of Freiburg, Freiburg im Breisgau, Germany; Department of Oncology, The Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA; Department of Biomedicine, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland; Division of Hematology, University Hospital Basel, Basel, Switzerland.
| | - Robert Zeiser
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, a Partnership Between DKFZ and Medical Center - University of Freiburg, Freiburg im Breisgau, Germany; Signalling Research Centres BIOSS and CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
8
|
Hosoi N, Shiraishi T, Okada T, Osone K, Yokobori T, Sakai M, Ogawa H, Sohda M, Shirabe K, Saeki H. Evaluation of preoperative visceral fat area / psoas muscle area ratio and prognosis in patients with colorectal cancer. Ann Gastroenterol Surg 2025; 9:119-127. [PMID: 39759994 PMCID: PMC11693571 DOI: 10.1002/ags3.12845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 01/07/2025] Open
Abstract
Background Recent research has focused on the prognostic relevance of preoperative sarcopenia and sarcopenic obesity in various cancers. In this study we investigated the relationship between visceral fat area (VFA), psoas muscle area (PMA), and the prognosis of patients undergoing colorectal cancer surgery. Methods Patients with stage III colorectal cancer who underwent surgery between July 2013 and April 2020 were included. The analysis was performed on 151 patients who met the criteria. The VFA and PMA were measured at the level of the third lumbar vertebra on computed tomography (CT) scans, and the ratio of VFA to PMA (V/P ratio) was determined. Results Patients with high V/P ratios were significantly older (p = 0.0213), had a higher body mass index (BMI) (p < 0.0001), a higher percentage of sarcopenic obesity (p < 0.0001), and more diabetes complications (p < 0.0001). Prognostic analysis showed that the overall survival (OS) (p = 0.0154) and relapse-free survival (RFS) (p = 0.0378) were significantly worse in patients with a high V/P ratio. Multivariate analysis revealed that a high V/P ratio was an independent poor prognostic factor for OS. Subgroup analysis was then performed in patients with BMI < 25 kg/m2. OS (p = 0.0259) and RFS (p = 0.0275) were significantly worse in the high V/P ratio group. A high V/P ratio was an independent poor prognostic factor in the multivariate analysis. Conclusion In colorectal cancer, the preoperative V/P ratio is an independent factor for poor prognosis. Preoperative evaluation of the V/P ratio may identify a wide range of high-risk patients because it is an independent poor prognostic factor in patients without obesity.
Collapse
Affiliation(s)
- Nobuhiro Hosoi
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Takuya Shiraishi
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Takuhisa Okada
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Katsuya Osone
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Takehiko Yokobori
- Division of Integrated Oncology ResearchGunma University, Initiative for Advanced Research (GIAR)MaebashiJapan
| | - Makoto Sakai
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Hiroomi Ogawa
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Makoto Sohda
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Ken Shirabe
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Hiroshi Saeki
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| |
Collapse
|
9
|
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
|
10
|
Luo Z, Wu A, Robson S, Alper S, Yu W. Adiponectin Signaling Regulates Urinary Bladder Function by Blunting Smooth Muscle Purinergic Contractility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620328. [PMID: 39554160 PMCID: PMC11565761 DOI: 10.1101/2024.10.25.620328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Lower urinary tract symptoms (LUTS) affect ∼ 50% of the population aged >40 years and are strongly associated with obesity and metabolic syndrome. Adipose tissue plays a key role in obesity/metabolic syndrome by releasing adipokines that regulate systemic energy/lipid metabolism, insulin resistance, and inflammation. Adiponectin (ADPN), the most abundant adipokine, modulates energy/metabolism homeostasis through its insulin-sensitizing and anti-inflammatory effects. Human plasma ADPN levels are inversely associated with obesity and diabetes. The role of adipokines such as ADPN in the LUTS associated with obesity/metabolic syndrome remains unknown. We have tested such a possible role in a global ADPN knockout mouse model ( Adpn -/- ). Adpn -/- mice exhibited increased voiding frequency, small voids, and reduced bladder smooth muscle (BSM) contractility with absence of purinergic contraction. Molecular examination indicated significantly altered metabolic and purinergic pathways. The ADPN receptor agonist AdipoRon was found to abolish acute BSM contraction. Intriguingly, both AMPK activators and inhibitors also abolished BSM purinergic contraction. These data indicate the important contribution of a novel ADPN signaling pathway to the regulation of BSM contractility. Dysregulation of this ADPN signaling pathway might be an important mechanism leading to LUTS associated with obesity/metabolic syndrome. ARTICLE HIGHLIGHTS Lower urinary tract symptom (LUTS) is strongly associated with obesity and metabolic syndrome, however, the underlying molecular mechanisms are missing. Dysregulation of adipokine signaling could be the link for this association.Whether adiponectin, the most abundant adipokine, plays a role in regulating bladder function and dysfunction.Mice null for adiponectin exhibited increased voiding frequency, small voids, and reduced bladder smooth muscle contractility, with corresponding metabolic and purinergic pathway changes.Dysregulation of adiponectin signaling might be an important mechanism leading to LUTS associated with obesity/metabolic syndrome.
Collapse
|
11
|
Moon S, Park Y, Jang S, Kim S, Song DG, Shin DC, Lee CH. Interleukin-2 improves insulin sensitivity through hypothalamic sympathetic activation in obese mice. J Neuroinflammation 2024; 21:250. [PMID: 39367382 PMCID: PMC11453069 DOI: 10.1186/s12974-024-03244-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND IL-2 regulates T cell differentiation: low-dose IL-2 induces immunoregulatory Treg differentiation, while high-dose IL-2 acts as a potent activator of cytotoxic T cells and NK cells. Therefore, high-dose IL-2 has been studied for use in cancer immunotherapy. We aimed to utilize low-dose IL-2 to treat inflammatory diseases such as obesity and insulin resistance, which involve low-grade chronic inflammation. MAIN BODY Systemic administration of low-dose IL-2 increased Treg cells and decreased inflammation in gonadal white adipose tissue (gWAT), leading to improved insulin sensitivity in high-fat diet-fed obese mice. Additionally, central administration of IL-2 significantly enhanced insulin sensitivity through the activation of the sympathetic nervous system. The sympathetic signaling induced by central IL-2 administration not only decreased interferon γ (IFNγ) + Th1 cells and the expression of pro-inflammatory cytokines, including Il-1β, Il-6, and Il-8, but also increased CD4 + CD25 + FoxP3 + Treg cells and Tgfβ expression in the gWAT of obese mice. These phenomena were accompanied by hypothalamic microgliosis and activation of pro-opiomelanocortin neurons. Furthermore, sympathetic denervation in gWAT reversed the enhanced insulin sensitivity and immune cell polarization induced by central IL-2 administration. CONCLUSION Overall, we demonstrated that IL-2 improves insulin sensitivity through two mechanisms: direct action on CD4 + T cells and via the neuro-immune axis triggered by hypothalamic microgliosis.
Collapse
Affiliation(s)
- Subin Moon
- Department of Biomedical Science, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Yejin Park
- Department of Biomedical Science, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Sooyeon Jang
- Department of Biomedical Science, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Saeha Kim
- Department of Biomedical Science, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Dan-Gyeong Song
- Department of Biomedical Science, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Dae-Chul Shin
- Department of Biomedical Science, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Chan Hee Lee
- Department of Biomedical Science, Hallym University, Chuncheon, 24252, Republic of Korea.
- Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, 24252, Republic of Korea.
| |
Collapse
|
12
|
Chen Y, Zhang Y, Jin X, Hong S, Tian H. Exerkines: Benign adaptation for exercise and benefits for non-alcoholic fatty liver disease. Biochem Biophys Res Commun 2024; 726:150305. [PMID: 38917635 DOI: 10.1016/j.bbrc.2024.150305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024]
Abstract
Exercise has multiple beneficial effects on human metabolic health and is regarded as a "polypill" for various diseases. At present, the lack of physical activity usually causes an epidemic of chronic metabolic syndromes, including obesity, cardiovascular diseases, and non-alcoholic fatty liver disease (NAFLD). Remarkably, NAFLD is emerging as a serious public health issue and is associated with the development of cirrhosis and hepatocellular carcinoma. Unfortunately, specific drug therapies for NAFLD and its more severe form, non-alcoholic steatohepatitis (NASH), are currently unavailable. Lifestyle modification is the foundation of treatment recommendations for NAFLD and NASH, especially for exercise. There are under-appreciated organs that crosstalk to the liver during exercise such as muscle-liver crosstalk. Previous studies have reported that certain exerkines, such as FGF21, GDF15, irisin, and adiponectin, are beneficial for liver metabolism and have the potential to be targeted for NAFLD treatment. In addition, some of exerkines can be modified for the new proteins and get enhanced functions, like IL-6/IC7Fc. Another importance of exercise is the physiological adaptation that combats metabolic diseases. Thus, this review aims to summarize the known exerkines and utilize a multi-omics mining tool to identify more exerkines for the future research. Overall, understanding the mechanisms by which exercise-induced exerkines exert their beneficial effects on metabolic health holds promise for the development of novel therapeutic strategies for NAFLD and related diseases.
Collapse
Affiliation(s)
- Yang Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Yan Zhang
- Clinical Laboratory, Suzhou Yong Ding Hospital, Suzhou, 215200, China
| | - Xingsheng Jin
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Shangyu Hong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200032, China.
| | - Haili Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China.
| |
Collapse
|
13
|
Quispe R, Sweeney T, Martin SS, Jones SR, Allison MA, Budoff MJ, Ndumele CE, Elshazly MB, Michos ED. Associations of Adipokine Levels With Levels of Remnant Cholesterol: The Multi-Ethnic Study of Atherosclerosis. J Am Heart Assoc 2024; 13:e030548. [PMID: 39248264 PMCID: PMC11935629 DOI: 10.1161/jaha.123.030548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 03/06/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND The metabolic syndrome phenotype of individuals with obesity is characterized by elevated levels of triglyceride-rich lipoproteins and remnant particles, which have been shown to be significantly atherogenic. Understanding the association between adipokines, endogenous hormones produced by adipose tissue, and remnant cholesterol (RC) would give insight into the link between obesity and atherosclerotic cardiovascular disease. METHODS AND RESULTS We studied 1791 MESA (Multi-Ethnic Study of Atherosclerosis) participants who took part in an ancillary study on body composition with adipokine levels measured (leptin, adiponectin, and resistin) at either visit 2 or visit 3. RC was calculated as non-high-density lipoprotein cholesterol minus low-density lipoprotein cholesterol, measured at the same visit as the adipokines, as well as subsequent visits 4 through 6. Multivariable-adjusted linear mixed-effects models were used to assess the cross-sectional and longitudinal associations between adipokines and log-transformed levels of RC. Mean±SD age was 64.5±9.6 years; mean±SD body mass index was 29.9±5.0 kg/m2; and 52.0% were women. In fully adjusted cross-sectional models that included body mass index, diabetes, low-density lipoprotein cholesterol, and lipid-lowering therapy, for each 1-unit increment in adiponectin, there was 14.6% (95% CI, 12.2-16.9) lower RC. With each 1-unit increment in leptin and resistin, there was 4.8% (95% CI, 2.7-7.0) and 4.0% (95% CI, 0.2-8.1) higher RC, respectively. Lower adiponectin and higher leptin were also associated with longitudinal increases in RC levels over median follow-up of 5 (interquartile range, 4-8) years. CONCLUSIONS Lower adiponectin and higher leptin levels were independently associated with higher levels of RC at baseline and longitudinal RC increase, even after accounting for body mass index and low-density lipoprotein cholesterol.
Collapse
Affiliation(s)
- Renato Quispe
- Ciccarone Center for the Prevention of Cardiovascular DiseaseJohns Hopkins University School of MedicineBaltimoreMD
| | - Ty Sweeney
- Ciccarone Center for the Prevention of Cardiovascular DiseaseJohns Hopkins University School of MedicineBaltimoreMD
| | - Seth S. Martin
- Ciccarone Center for the Prevention of Cardiovascular DiseaseJohns Hopkins University School of MedicineBaltimoreMD
| | - Steven R. Jones
- Ciccarone Center for the Prevention of Cardiovascular DiseaseJohns Hopkins University School of MedicineBaltimoreMD
| | - Matthew A. Allison
- Department of Family MedicineUniversity of California San DiegoSan DiegoCA
| | | | - Chiadi E. Ndumele
- Ciccarone Center for the Prevention of Cardiovascular DiseaseJohns Hopkins University School of MedicineBaltimoreMD
| | - Mohamed B. Elshazly
- Department of Cardiovascular MedicineHeart and Vascular Institute, Cleveland ClinicClevelandOH
| | - Erin D. Michos
- Ciccarone Center for the Prevention of Cardiovascular DiseaseJohns Hopkins University School of MedicineBaltimoreMD
| |
Collapse
|
14
|
Rehman IU, Park JS, Choe K, Park HY, Park TJ, Kim MO. Overview of a novel osmotin abolishes abnormal metabolic-associated adiponectin mechanism in Alzheimer's disease: Peripheral and CNS insights. Ageing Res Rev 2024; 100:102447. [PMID: 39111409 DOI: 10.1016/j.arr.2024.102447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/19/2024] [Accepted: 08/03/2024] [Indexed: 08/16/2024]
Abstract
Alzheimer's disease (AD) is a degenerative brain disease that affects millions of people worldwide. It is caused by abnormalities in cholinergic neurons, oxidative stress, and inflammatory cascades. The illness is accompanied by personality changes, memory issues, and dementia. Metabolic signaling pathways help with fundamental processes like DNA replication and RNA transcription. Being adaptable is essential for both surviving and treating illness. The body's metabolic signaling depends on adipokines, including adiponectin (APN) and other adipokines secreted by adipose tissues. Energy homeostasis is balanced by adipokines, and nutrients. Overconsumption of nutrients messes with irregular signaling of adipokines, such as APN in both peripheral and brain which leads to neurodegeneration, such as AD. Despite the failure of traditional treatments like memantine and cholinesterase inhibitors, natural plant bioactive substances like Osmotin (OSM) have been given a focus as potential therapeutics due to their antioxidant properties, better blood brain barrier (BBB) permeability, excellent cell viability, and especially nanoparticle approaches. The review highlights the published preclinical literature regarding the role of OSM in AD pathology while there is a need for more research to investigate the hidden therapeutic potential of OSM which may open a new gateway and further strengthen its healing role in the pathogenesis of neurodegeneration, especially AD.
Collapse
Affiliation(s)
- Inayat Ur Rehman
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea.
| | - Jun Sung Park
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea.
| | - Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht 6229 ER, the Netherlands.
| | - Hyun Young Park
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht 6229 ER, the Netherlands; Department of Pediatrics, Maastricht University Medical Center (MUMC+), Maastricht 6202 AZ, the Netherlands.
| | - Tae Ju Park
- Haemato-oncology/Systems Medicine Group, Paul O'Gorman Leukemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences (MVLS), University of Glasgow, Glasgow G12 0ZD, United Kingdom.
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Alz-Dementia Korea Co., Jinju 52828, Republic of Korea.
| |
Collapse
|
15
|
Kountouras J, Zavos C, Vardaka E, Kyrailidi F, Mouratidou MC, Tzitiridou-Chatzopoulou M, Orovou E, Touloumtzi M, Papanikolaou IS, Kazakos E. Helicobacter pylori and metabolic syndrome-related adipokines in nonalcoholic fatty liver disease pathophysiology. J Gastroenterol Hepatol 2024; 39:1957-1959. [PMID: 38725326 DOI: 10.1111/jgh.16621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/06/2024] [Accepted: 05/03/2024] [Indexed: 10/01/2024]
Affiliation(s)
- J Kountouras
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Macedonia, Greece
| | - C Zavos
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Macedonia, Greece
| | - E Vardaka
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Macedonia, Greece
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, Alexander Campus, Sindos, Macedonia, Greece
| | - F Kyrailidi
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Macedonia, Greece
| | - M C Mouratidou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Macedonia, Greece
| | - M Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Macedonia, Greece
- School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, Macedonia, Greece
| | - E Orovou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Macedonia, Greece
- School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, Macedonia, Greece
| | - M Touloumtzi
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Macedonia, Greece
| | - I S Papanikolaou
- Hepatogastroenterology Unit, Second Department of Internal Medicine-Propaedeutic, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - E Kazakos
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Macedonia, Greece
- School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, Macedonia, Greece
| |
Collapse
|
16
|
Nunes-Souza V, Alenina N, Qadri F, Mosienko V, Santos RAS, Bader M, Rabelo LA. ACE2 Knockout Mice Are Resistant to High-Fat Diet-Induced Obesity in an Age-Dependent Manner. Int J Mol Sci 2024; 25:9515. [PMID: 39273464 PMCID: PMC11394789 DOI: 10.3390/ijms25179515] [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: 07/27/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) presents pleiotropic actions. It hydrolyzes angiotensin I (AngI) and angiotensin II (AngII) into angiotensin-(1-9) (Ang-(1-9)) and angiotensin-(1-7) (Ang-(1-7)), respectively, as well as participates in tryptophan uptake in the gut and in COVID-19 infection. Our aim was to investigate the metabolic effect of ACE2 deletion in young adults and elderly mice under conditions of high calorie intake. Male C57Bl/6 (WT) and ACE2-deficient (ACE2-/y) mice were analyzed at the age of 6 and 12 months under standard diet (StD) and high-fat diet (HFD). Under StD, ACE2-/y showed lower body weight and fat depots, improved glucose tolerance, enhanced insulin sensitivity, higher adiponectin, and lower leptin levels compared to WT. This difference was even more pronounced after HFD in 6-month-old mice, but, interestingly, it was blunted at the age of 12 months. ACE2-/y presented a decrease in adipocyte diameter and lipolysis, which reflected in the upregulation of lipid metabolism in white adipose tissue through the increased expression of genes involved in lipid regulation. Under HFD, both food intake and total energy expenditure were decreased in 6-month-old ACE2-/y mice, accompanied by an increase in liquid intake, compared to WT mice, fed either StD or HFD. Thus, ACE2-/y mice are less susceptible to HFD-induced obesity in an age-dependent manner, as well as represent an excellent animal model of human lipodystrophy and a tool to investigate new treatments.
Collapse
Affiliation(s)
- Valéria Nunes-Souza
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife 50670-901, Brazil
- National Institute of Science and Technology in Nanobiopharmaceutics (Nanobiofar), Belo Horizonte 31270-901, Brazil;
| | - Natalia Alenina
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
| | - Fatimunnisa Qadri
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
| | - Valentina Mosienko
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Robson Augusto Souza Santos
- National Institute of Science and Technology in Nanobiopharmaceutics (Nanobiofar), Belo Horizonte 31270-901, Brazil;
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, 10117 Berlin, Germany
- Institute for Biology, University of Lübeck, 23562 Lübeck, Germany
| | - Luiza Antas Rabelo
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; (V.N.-S.); (F.Q.); (V.M.); (M.B.)
- National Institute of Science and Technology in Nanobiopharmaceutics (Nanobiofar), Belo Horizonte 31270-901, Brazil;
- Laboratory of Cardiovascular Reactivity, Metabolic Syndrome Center, Institute of Biological Sciences and Health, Federal University of Alagoas, Maceió 57072-900, Brazil
| |
Collapse
|
17
|
Goli SH, Lim JY, Basaran-Akgul N, Templeton SP. Adiponectin pathway activation dampens inflammation and enhances alveolar macrophage fungal killing via LC3-associated phagocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600373. [PMID: 38979340 PMCID: PMC11230297 DOI: 10.1101/2024.06.24.600373] [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
Although innate immunity is critical for antifungal host defense against the human opportunistic fungal pathogen Aspergillus fumigatus, potentially damaging inflammation must be controlled. Adiponectin (APN) is an adipokine produced mainly in adipose tissue that exerts anti-inflammatory effects in adipose-distal tissues such as the lung. We observed 100% mortality and increased fungal burden and inflammation in neutropenic mice with invasive aspergillosis (IA) that lack APN or the APN receptors AdipoR1 or AdipoR2. Alveolar macrophages (AMs), early immune sentinels that detect and respond to lung infection, express both receptors, and APN-/- AMs exhibited an inflammatory/M1 phenotype that was associated with decreased fungal killing. Pharmacological stimulation of AMs with AdipoR agonist AdipoRon partially rescued deficient killing in APN-/- AMs that was dependent on both receptors. Finally, APN-enhanced fungal killing was associated with increased activation of the non-canonical LC3 pathway of autophagy. Thus, our study identifies a novel role for APN in LC3-mediated killing of A. fumigatus.
Collapse
Affiliation(s)
- Sri Harshini Goli
- Department of Microbiology and Immunology, Indiana University School of Medicine-Terre Haute, Terre Haute, IN 47809, USA
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
| | - Joo-Yeon Lim
- Department of Microbiology and Immunology, Indiana University School of Medicine-Terre Haute, Terre Haute, IN 47809, USA
| | - Nese Basaran-Akgul
- Department of Microbiology and Immunology, Indiana University School of Medicine-Terre Haute, Terre Haute, IN 47809, USA
| | - Steven P. Templeton
- Department of Microbiology and Immunology, Indiana University School of Medicine-Terre Haute, Terre Haute, IN 47809, USA
| |
Collapse
|
18
|
Navarro León A, Muñoz M, Iglesias N, Blanco-Vázquez C, Balseiro A, Milhano Santos F, Ciordia S, Corrales FJ, Iglesias T, Casais R. Proteomic Serum Profiling of Holstein Friesian Cows with Different Pathological Forms of Bovine Paratuberculosis Reveals Changes in the Acute-Phase Response and Lipid Metabolism. J Proteome Res 2024; 23:2762-2779. [PMID: 37863471 PMCID: PMC11301775 DOI: 10.1021/acs.jproteome.3c00244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/22/2023]
Abstract
The lack of sensitive diagnostic methods to detect Mycobacterium avium subsp. paratuberculosis (Map) subclinical infections has hindered the control of paratuberculosis (PTB). The serum proteomic profiles of naturally infected cows presenting focal and diffuse pathological forms of PTB and negative controls (n = 4 per group) were analyzed using TMT-6plex quantitative proteomics. Focal and diffuse are the most frequent pathological forms in subclinical and clinical stages of PTB, respectively. One (focal versus (vs.) control), eight (diffuse vs. control), and four (focal vs. diffuse) differentially abundant (DA) proteins (q-value < 0.05) were identified. Ingenuity pathway analysis of the DA proteins revealed changes in the acute-phase response and lipid metabolism. Six candidate biomarkers were selected for further validation by specific ELISA using serum from animals with focal, multifocal, and diffuse PTB-associated lesions (n = 108) and controls (n = 56). Overall, the trends of the serum expression levels of the selected proteins were consistent with the proteomic results. Alpha-1-acid glycoprotein (ORM1)-based ELISA, insulin-like growth factor-binding protein 2 (IGFBP2)-based ELISA, and the anti-Map ELISA had the best diagnostic performance for detection of animals with focal, multifocal, and diffuse lesions, respectively. Our findings identify potential biomarkers that improve diagnostic sensitivity of PTB and help to elucidate the mechanisms involved in PTB pathogenesis.
Collapse
Affiliation(s)
- Alejandra
Isabel Navarro León
- Center
for Animal Biotechnology, Servicio Regional
de Investigación y Desarrollo Agroalimentario [SERIDA], 33394 Deva, Asturias, Spain
| | - Marta Muñoz
- Center
for Animal Biotechnology, Servicio Regional
de Investigación y Desarrollo Agroalimentario [SERIDA], 33394 Deva, Asturias, Spain
| | - Natalia Iglesias
- Center
for Animal Biotechnology, Servicio Regional
de Investigación y Desarrollo Agroalimentario [SERIDA], 33394 Deva, Asturias, Spain
| | - Cristina Blanco-Vázquez
- Center
for Animal Biotechnology, Servicio Regional
de Investigación y Desarrollo Agroalimentario [SERIDA], 33394 Deva, Asturias, Spain
| | - Ana Balseiro
- Departamento
de Sanidad Animal, Facultad de Veterinaria, Universidad de León, 24071 León, Spain
| | - Fátima Milhano Santos
- Functional
Proteomics Laboratory, National Center for Biotechnology, Consejo Superior de Investigaciones Científicas [CSIC],
Proteored-ISCIII, 28049 Madrid, Spain
| | - Sergio Ciordia
- Functional
Proteomics Laboratory, National Center for Biotechnology, Consejo Superior de Investigaciones Científicas [CSIC],
Proteored-ISCIII, 28049 Madrid, Spain
| | - Fernando J. Corrales
- Functional
Proteomics Laboratory, National Center for Biotechnology, Consejo Superior de Investigaciones Científicas [CSIC],
Proteored-ISCIII, 28049 Madrid, Spain
| | - Tania Iglesias
- Unidad
de Consultoría Estadística, Servicios Científico-técnicos, Universidad de Oviedo, Campus de Gijón, 33203 Gijón, Asturias, Spain
| | - Rosa Casais
- Center
for Animal Biotechnology, Servicio Regional
de Investigación y Desarrollo Agroalimentario [SERIDA], 33394 Deva, Asturias, Spain
| |
Collapse
|
19
|
Phuong LDT, Tran Huy T, Huynh Quang T. The Plasma Levels of Protein Adiponectin ( AdipoQ) and Meteorin-Like (Metrnl) in Newly Diagnosed Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2024; 17:2903-2909. [PMID: 39100966 PMCID: PMC11298186 DOI: 10.2147/dmso.s471954] [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: 04/02/2024] [Accepted: 07/23/2024] [Indexed: 08/06/2024] Open
Abstract
Purpose This study aimed to measure the concentrations of the Adiponectin and Meteorin - Like (Metrnl) in newly diagnosed type 2 diabetes patients. Patients and Methods A comparative cross-sectional study contained two groups: Group 1 (86 newly diagnosed diabetes mellitus type 2 patients) and group 2 (71 healthy persons). The plasma concentrations of Adiponectin and Metrnl were measured by Enzyme Link Immunosorbent Assay (ELISA). Results The plasma level of Adiponectin of the newly diagnosed diabetes mellitus type 2 group and the healthy group were 1219.82 ng/mL (1132.43-2772.50) and 1187.25 ng/mL (1160.66-3807.50) respectively. The plasma level of Metrnl of two groups were 757.60 pg/mL (564.15-994.00) and 697.60 pg/mL (538.50-986.10) respectively. There were no significant difference between two groups. Metrnl had no correlation with glucose, HbA1c, lipid profile, BMI. Adiponectin had correlation with Metrnl and HDL-cholesterol. Adiponectin had no correlation to glucose, HbA1c, LDL-cholesterol, total cholesterol, triglyceride, BMI. People with the lower Adiponectin concentration had the higher risk of diabetes (OR=6.52; 95% CI: 2.43 -17.55). Conclusion Adiponectin and Metrnl were not significantly different in newly diagnosed type 2 diabetes and healthy people. The lower concentration of Adiponectin might increase the risk of type 2 diabetes.
Collapse
Affiliation(s)
- Lan Dam Thi Phuong
- Biochemistry Department, Hanoi Medical University, Hanoi, Vietnam
- Department of Biochemistry, 103 Military Hospital, Vietnam Military Medical University, Hanoi, Vietnam
| | - Thinh Tran Huy
- Biochemistry Department, Hanoi Medical University, Hanoi, Vietnam
| | - Thuan Huynh Quang
- Department of Biochemistry, 103 Military Hospital, Vietnam Military Medical University, Hanoi, Vietnam
| |
Collapse
|
20
|
Lai HC, Chen PH, Tang CH, Chen LW. IL-10 Enhances the Inhibitory Effect of Adipose-Derived Stromal Cells on Insulin Resistance/Liver Gluconeogenesis by Treg Cell Induction. Int J Mol Sci 2024; 25:8088. [PMID: 39125659 PMCID: PMC11311376 DOI: 10.3390/ijms25158088] [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: 06/10/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
The modulation of cellular phenotypes within adipose tissue provides a potential means for therapeutic intervention for diabetes. Endogenous interleukin-10 (IL-10) protects against diet-induced insulin resistance. We examined the effects and mechanisms of action of IL-10-treated adipose-derived stromal cells on diabetes-induced insulin resistance and liver gluconeogenesis. We harvested stromal vascular fractions (SVFs) from the adipose tissue of diabetic (Leprdb/db) mice and treated them with IL-10 in vitro. SVFs treated with 10 or 100 ng of IL-10 were injected into the inguinal adipose tissue of Leprdb/db mice. IL-10 treatment suppressed the mRNA expression of IL-6, IL-33, CCL2, TNF-α, and IL-1β. Additionally, it suppressed the protein expression of IL-6, pmTOR, pJNK, and pNF-κB but enhanced Foxp3 mRNA expression in SVFs from diabetic mice. Meanwhile, IL-10 treatment repressed CCL2 and PDGFRα expression in adipose tissue macrophages (ATMs) and IL-6 expression in non-ATMs but increased the Foxp3 and IL-10 mRNA expression of ATMs from diabetic mice. Injection of IL-10-treated SVFs decreased the IL-6, IL-33, CCL2, IL-1β, and CCL2 but enhanced the Foxp3 and IL-10 mRNA expression of adipose tissue from Leprdb/db mice. Furthermore, injection of IL-10-treated SVFs increased CD4+ regulatory T cells (Tregs) in SVFs and adipose IL-10 levels and suppressed plasma adiponectin levels and DPP4 activity in diabetic mice. Injection of IL-10-treated SVFs decreased hepatic G6PC and PCK1 mRNA expression and increased Akt activation, STAT3 phosphorylation in the liver, and glucose tolerance in diabetic mice. Our data suggest that IL-10 treatment decreases inflammation in adipose SVFs of diabetic mice. Injection of IL-10-treated SVFs into the adipose tissue decreased diabetes-induced gluconeogenesis gene expression, DPP4 activity, and insulin resistance by enhancing Treg cells in diabetic mice. These data suggest that IL-10-treated adipose stromal vascular cells could be a promising therapeutic strategy for diabetes mellitus.
Collapse
Affiliation(s)
- Hsiao-Chi Lai
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan; (H.-C.L.); (P.-H.C.); (C.-H.T.)
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Pei-Hsuan Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan; (H.-C.L.); (P.-H.C.); (C.-H.T.)
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chia-Hua Tang
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan; (H.-C.L.); (P.-H.C.); (C.-H.T.)
| | - Lee-Wei Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan; (H.-C.L.); (P.-H.C.); (C.-H.T.)
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| |
Collapse
|
21
|
DeBari MK, Johnston EK, Scott JV, Ilzuka E, Sun W, Webster-Wood VA, Abbott RD. A Preliminary Study on Factors That Drive Patient Variability in Human Subcutaneous Adipose Tissues. Cells 2024; 13:1240. [PMID: 39120271 PMCID: PMC11311805 DOI: 10.3390/cells13151240] [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: 06/10/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Adipose tissue is a dynamic regulatory organ that has profound effects on the overall health of patients. Unfortunately, inconsistencies in human adipose tissues are extensive and multifactorial, including large variability in cellular sizes, lipid content, inflammation, extracellular matrix components, mechanics, and cytokines secreted. Given the high human variability, and since much of what is known about adipose tissue is from animal models, we sought to establish correlations and patterns between biological, mechanical, and epidemiological properties of human adipose tissues. To do this, twenty-six independent variables were cataloged for twenty patients, which included patient demographics and factors that drive health, obesity, and fibrosis. A factorial analysis for mixed data (FAMD) was used to analyze patterns in the dataset (with BMI > 25), and a correlation matrix was used to identify interactions between quantitative variables. Vascular endothelial growth factor A (VEGFA) and actin alpha 2, smooth muscle (ACTA2) gene expression were the highest loadings in the first two dimensions of the FAMD. The number of adipocytes was also a key driver of patient-related differences, where a decrease in the density of adipocytes was associated with aging. Aging was also correlated with a decrease in overall lipid percentage of subcutaneous tissue, with lipid deposition being favored extracellularly, an increase in transforming growth factor-β1 (TGFβ1), and an increase in M1 macrophage polarization. An important finding was that self-identified race contributed to variance between patients in this study, where Black patients had significantly lower gene expression levels of TGFβ1 and ACTA2. This finding supports the urgent need to account for patient ancestry in biomedical research to develop better therapeutic strategies for all patients. Another important finding was that TGFβ induced factor homeobox 1 (TGIF1), an understudied signaling molecule, which is highly correlated with leptin signaling, was correlated with metabolic inflammation. Furthermore, this study draws attention to what we define as "extracellular lipid droplets", which were consistently found in collagen-rich regions of the obese adipose tissues evaluated here. Reduced levels of TGIF1 were correlated with higher numbers of extracellular lipid droplets and an inability to suppress fibrotic changes in adipose tissue. Finally, this study indicated that M1 and M2 macrophage markers were correlated with each other and leptin in patients with a BMI > 25. This finding supports growing evidence that macrophage polarization in obesity involves a complex, interconnecting network system rather than a full switch in activation patterns from M2 to M1 with increasing body mass. Overall, this study reinforces key findings in animal studies and identifies important areas for future research, where human and animal studies are divergent. Understanding key drivers of human patient variability is required to unravel the complex metabolic health of unique patients.
Collapse
Affiliation(s)
- Megan K. DeBari
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Elizabeth K. Johnston
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Jacqueline V. Scott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Erica Ilzuka
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Wenhuan Sun
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Victoria A. Webster-Wood
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Rosalyn D. Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| |
Collapse
|
22
|
Gan HW, Cerbone M, Dattani MT. Appetite- and Weight-Regulating Neuroendocrine Circuitry in Hypothalamic Obesity. Endocr Rev 2024; 45:309-342. [PMID: 38019584 PMCID: PMC11074800 DOI: 10.1210/endrev/bnad033] [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: 02/04/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Since hypothalamic obesity (HyOb) was first described over 120 years ago by Joseph Babinski and Alfred Fröhlich, advances in molecular genetic laboratory techniques have allowed us to elucidate various components of the intricate neurocircuitry governing appetite and weight regulation connecting the hypothalamus, pituitary gland, brainstem, adipose tissue, pancreas, and gastrointestinal tract. On a background of an increasing prevalence of population-level common obesity, the number of survivors of congenital (eg, septo-optic dysplasia, Prader-Willi syndrome) and acquired (eg, central nervous system tumors) hypothalamic disorders is increasing, thanks to earlier diagnosis and management as well as better oncological therapies. Although to date the discovery of several appetite-regulating peptides has led to the development of a range of targeted molecular therapies for monogenic obesity syndromes, outside of these disorders these discoveries have not translated into the development of efficacious treatments for other forms of HyOb. This review aims to summarize our current understanding of the neuroendocrine physiology of appetite and weight regulation, and explore our current understanding of the pathophysiology of HyOb.
Collapse
Affiliation(s)
- Hoong-Wei Gan
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Manuela Cerbone
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul Tulsidas Dattani
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
23
|
Kountouras J, Boziki M, Kazakos E, Theotokis P, Kesidou E, Nella M, Bakirtzis C, Karafoulidou E, Vardaka E, Mouratidou MC, Kyrailidi F, Tzitiridou-Chatzopoulou M, Orovou E, Giartza-Taxidou E, Deretzi G, Grigoriadis N, Doulberis M. Impact of Helicobacter pylori and metabolic syndrome on mast cell activation-related pathophysiology and neurodegeneration. Neurochem Int 2024; 175:105724. [PMID: 38508416 DOI: 10.1016/j.neuint.2024.105724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/03/2024] [Accepted: 03/17/2024] [Indexed: 03/22/2024]
Abstract
Both Helicobacter pylori (H. pylori) infection and metabolic syndrome (MetS) are highly prevalent worldwide. The emergence of relevant research suggesting a pathogenic linkage between H. pylori infection and MetS-related cardio-cerebrovascular diseases and neurodegenerative disorders, particularly through mechanisms involving brain pericyte deficiency, hyperhomocysteinemia, hyperfibrinogenemia, elevated lipoprotein-a, galectin-3 overexpression, atrial fibrillation, and gut dysbiosis, has raised stimulating questions regarding their pathophysiology and its translational implications for clinicians. An additional stimulating aspect refers to H. pylori and MetS-related activation of innate immune cells, mast cells (MC), which is an important, often early, event in systemic inflammatory pathologies and related brain disorders. Synoptically, MC degranulation may play a role in the pathogenesis of H. pylori and MetS-related obesity, adipokine effects, dyslipidemia, diabetes mellitus, insulin resistance, arterial hypertension, vascular dysfunction and arterial stiffness, an early indicator of atherosclerosis associated with cardio-cerebrovascular and neurodegenerative disorders. Meningeal MC can be activated by triggers including stress and toxins resulting in vascular changes and neurodegeneration. Likewise, H.pylori and MetS-related MC activation is linked with: (a) vasculitis and thromboembolic events that increase the risk of cardio-cerebrovascular and neurodegenerative disorders, and (b) gut dysbiosis-associated neurodegeneration, whereas modulation of gut microbiota and MC activation may promote neuroprotection. This narrative review investigates the intricate relationship between H. pylori infection, MetS, MC activation, and their collective impact on pathophysiological processes linked to neurodegeneration. Through a comprehensive search of current literature, we elucidate the mechanisms through which H. pylori and MetS contribute to MC activation, subsequently triggering cascades of inflammatory responses. This highlights the role of MC as key mediators in the pathogenesis of cardio-cerebrovascular and neurodegenerative disorders, emphasizing their involvement in neuroinflammation, vascular dysfunction and, ultimately, neuronal damage. Although further research is warranted, we provide a novel perspective on the pathophysiology and management of brain disorders by exploring potential therapeutic strategies targeting H. pylori eradication, MetS management, and modulation of MC to mitigate neurodegeneration risk while promoting neuroprotection.
Collapse
Affiliation(s)
- Jannis Kountouras
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece.
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Evangelos Kazakos
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, 50100, Macedonia, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Maria Nella
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Christos Bakirtzis
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Eleni Karafoulidou
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Elisabeth Vardaka
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, Alexander Campus, 57400, Macedonia, Greece
| | - Maria C Mouratidou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece
| | - Foteini Kyrailidi
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, 50100, Macedonia, Greece
| | - Eirini Orovou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, 50100, Macedonia, Greece
| | - Evaggelia Giartza-Taxidou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece
| | - Georgia Deretzi
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; Department of Neurology, Papageorgiou General Hospital, Thessaloniki, Macedonia, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; Gastroklinik, Private Gastroenterological Practice, 8810, Horgen, Switzerland; Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001, Aarau, Switzerland
| |
Collapse
|
24
|
Zhou N, Gong L, Zhang E, Wang X. Exploring exercise-driven exerkines: unraveling the regulation of metabolism and inflammation. PeerJ 2024; 12:e17267. [PMID: 38699186 PMCID: PMC11064867 DOI: 10.7717/peerj.17267] [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: 11/01/2023] [Accepted: 03/28/2024] [Indexed: 05/05/2024] Open
Abstract
Exercise has many beneficial effects that provide health and metabolic benefits. Signaling molecules are released from organs and tissues in response to exercise stimuli and are widely termed exerkines, which exert influence on a multitude of intricate multi-tissue processes, such as muscle, adipose tissue, pancreas, liver, cardiovascular tissue, kidney, and bone. For the metabolic effect, exerkines regulate the metabolic homeostasis of organisms by increasing glucose uptake and improving fat synthesis. For the anti-inflammatory effect, exerkines positively influence various chronic inflammation-related diseases, such as type 2 diabetes and atherosclerosis. This review highlights the prospective contribution of exerkines in regulating metabolism, augmenting the anti-inflammatory effects, and providing additional advantages associated with exercise. Moreover, a comprehensive overview and analysis of recent advancements are provided in this review, in addition to predicting future applications used as a potential biomarker or therapeutic target to benefit patients with chronic diseases.
Collapse
Affiliation(s)
- Nihong Zhou
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
- School of Sport Science, Beijing Sport University, Beijing, China
| | - Lijing Gong
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
- Key Laboratory for Performance Training & Recovery of General Administration of Sport, Beijing Sport University, Beijing, China
| | - Enming Zhang
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, Sweden
- NanoLund Center for NanoScience, Lund University, Lund, Sweden
| | - Xintang Wang
- Key Laboratory for Performance Training & Recovery of General Administration of Sport, Beijing Sport University, Beijing, China
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| |
Collapse
|
25
|
Meyer M, Schwärzler J, Jukic A, Tilg H. Innate Immunity and MASLD. Biomolecules 2024; 14:476. [PMID: 38672492 PMCID: PMC11048298 DOI: 10.3390/biom14040476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has emerged as the most common liver disease worldwide in recent years. MASLD commonly presents as simple hepatic steatosis, but ~25% of patients develop liver inflammation, progressive fibrosis, liver cirrhosis and related hepatocellular carcinoma. Liver inflammation and the degree of fibrosis are key determinants of the prognosis. The pathophysiology of liver inflammation is incompletely understood and involves diverse factors and specifically innate and adaptive immune responses. More specifically, diverse mediators of innate immunity such as proinflammatory cytokines, adipokines, inflammasomes and various cell types like mononuclear cells, macrophages and natural killer cells are involved in directing the inflammatory process in MASLD. The activation of innate immunity is driven by various factors including excess lipids and lipotoxicity, insulin resistance and molecular patterns derived from gut commensals. Targeting pathways of innate immunity might therefore appear as an attractive therapeutic strategy in the future management of MASLD and possibly its complications.
Collapse
Affiliation(s)
| | | | | | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.M.); (A.J.)
| |
Collapse
|
26
|
Dedinská I, Kleinová P, Macháleková K, Graňák K, Vnučák M, Beliančinová M. The role of hyperleptinaemia and low values of interleukin 10 in de novo DSA production after kidney transplantation. Transpl Immunol 2024; 83:101982. [PMID: 38218229 DOI: 10.1016/j.trim.2023.101982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 12/20/2023] [Accepted: 12/31/2023] [Indexed: 01/15/2024]
Abstract
BACKGROUND White adipose tissue secretes a number of peptide hormones. The aim of this paper was to determine the role of leptin, adiponectin and interleukin-10 and interleukin-6 on the development of graft rejection in protocol biopsy after kidney transplantation. METHODS In a prospective analysis (n = 104), we monitored the values of leptin, adiponectin, IL-6, and IL-10 prior to the transplantation and in the 3rd month after the transplantation. The protocol biopsy of the graft was performed in the 3rd month after the transplantation. The group was divided into the following according to the biopsy result: negative result, IFTA 1, borderline, and DSA positive. RESULTS After adjusting for the differences in the baseline recipient and donor characteristics, we identified the hyperleptinaemia baseline (HR = 2.0444, P = 0.0341) and month 3 (HR = 49.8043, P < 0.0001) as independent risk factors for borderline changes in the protocol biopsy. The hyperleptinaemia baseline (HR = 7.4979, P = 0.0071) and month 3 (HR = 9.7432, P = 0.0057) are independent risk factors for de novo DSA positivity. A low value of IL-10 month 3 is a risk factor for de novo DSA positivity (HR = 3.0746, P = 0.0388). CONCLUSIONS Higher leptin levels and low values of IL-10 might play a role in rejection and de novo DSA production.
Collapse
Affiliation(s)
- Ivana Dedinská
- Transplant Center, University Hospital Martin and Jessenius Faculty of Medicine Comenius University, Slovak Republic; Department of Internal Diseases I, University Hospital Martin and Jessenius Faculty of Medicine Comenius University, Slovak Republic.
| | - Patrícia Kleinová
- Transplant Center, University Hospital Martin and Jessenius Faculty of Medicine Comenius University, Slovak Republic; Department of Internal Diseases I, University Hospital Martin and Jessenius Faculty of Medicine Comenius University, Slovak Republic
| | - Katarína Macháleková
- Department od Pathology, St. Elizabeth Cancer Institute, Bratislava, Slovak Republic
| | - Karol Graňák
- Transplant Center, University Hospital Martin and Jessenius Faculty of Medicine Comenius University, Slovak Republic; Department of Internal Diseases I, University Hospital Martin and Jessenius Faculty of Medicine Comenius University, Slovak Republic
| | - Matej Vnučák
- Transplant Center, University Hospital Martin and Jessenius Faculty of Medicine Comenius University, Slovak Republic; Department of Internal Diseases I, University Hospital Martin and Jessenius Faculty of Medicine Comenius University, Slovak Republic
| | - Monika Beliančinová
- Transplant Center, University Hospital Martin and Jessenius Faculty of Medicine Comenius University, Slovak Republic; Department of Internal Diseases I, University Hospital Martin and Jessenius Faculty of Medicine Comenius University, Slovak Republic
| |
Collapse
|
27
|
Bellissimo CA, Gandhi S, Castellani LN, Murugathasan M, Delfinis LJ, Thuhan A, Garibotti MC, Seo Y, Rebalka IA, Hsu HH, Sweeney G, Hawke TJ, Abdul-Sater AA, Perry CGR. The slow-release adiponectin analog ALY688-SR modifies early-stage disease development in the D2. mdx mouse model of Duchenne muscular dystrophy. Am J Physiol Cell Physiol 2024; 326:C1011-C1026. [PMID: 38145301 DOI: 10.1152/ajpcell.00638.2023] [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: 12/05/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Fibrosis is associated with respiratory and limb muscle atrophy in Duchenne muscular dystrophy (DMD). Current standard of care partially delays the progression of this myopathy but there remains an unmet need to develop additional therapies. Adiponectin receptor agonism has emerged as a possible therapeutic target to lower inflammation and improve metabolism in mdx mouse models of DMD but the degree to which fibrosis and atrophy are prevented remain unknown. Here, we demonstrate that the recently developed slow-release peptidomimetic adiponectin analog, ALY688-SR, remodels the diaphragm of murine model of DMD on DBA background (D2.mdx) mice treated from days 7-28 of age during early stages of disease. ALY688-SR also lowered interleukin-6 (IL-6) mRNA but increased IL-6 and transforming growth factor-β1 (TGF-β1) protein contents in diaphragm, suggesting dynamic inflammatory remodeling. ALY688-SR alleviated mitochondrial redox stress by decreasing complex I-stimulated H2O2 emission. Treatment also attenuated fibrosis, fiber type-specific atrophy, and in vitro diaphragm force production in diaphragm suggesting a complex relationship between adiponectin receptor activity, muscle remodeling, and force-generating properties during the very early stages of disease progression in murine model of DMD on DBA background (D2.mdx) mice. In tibialis anterior, the modest fibrosis at this young age was not altered by treatment, and atrophy was not apparent at this young age. These results demonstrate that short-term treatment of ALY688-SR in young D2.mdx mice partially prevents fibrosis and fiber type-specific atrophy and lowers force production in the more disease-apparent diaphragm in relation to lower mitochondrial redox stress and heterogeneous responses in certain inflammatory markers. These diverse muscle responses to adiponectin receptor agonism in early stages of DMD serve as a foundation for further mechanistic investigations.NEW & NOTEWORTHY There are limited therapies for the treatment of Duchenne muscular dystrophy. As fibrosis involves an accumulation of collagen that replaces muscle fibers, antifibrotics may help preserve muscle function. We report that the novel adiponectin receptor agonist ALY688-SR prevents fibrosis in the diaphragm of D2.mdx mice with short-term treatment early in disease progression. These responses were related to altered inflammation and mitochondrial functions and serve as a foundation for the development of this class of therapy.
Collapse
MESH Headings
- Animals
- Mice
- Mice, Inbred mdx
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Adiponectin/genetics
- Disease Models, Animal
- Interleukin-6/metabolism
- Mice, Inbred C57BL
- Hydrogen Peroxide/metabolism
- Receptors, Adiponectin/genetics
- Receptors, Adiponectin/metabolism
- Mice, Inbred DBA
- Muscle, Skeletal/metabolism
- Diaphragm/metabolism
- Fibrosis
- Inflammation/metabolism
- Disease Progression
- Atrophy/metabolism
- Atrophy/pathology
Collapse
Affiliation(s)
- Catherine A Bellissimo
- School of Kinesiology & Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Shivam Gandhi
- School of Kinesiology & Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Laura N Castellani
- School of Kinesiology & Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Mayoorey Murugathasan
- School of Kinesiology & Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Luca J Delfinis
- School of Kinesiology & Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Arshdeep Thuhan
- School of Kinesiology & Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Madison C Garibotti
- School of Kinesiology & Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Yeji Seo
- School of Kinesiology & Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Henry H Hsu
- Allysta Pharmaceuticals Inc, Bellevue, Washington, United States
| | - Gary Sweeney
- Department of Biology, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ali A Abdul-Sater
- School of Kinesiology & Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Christopher G R Perry
- School of Kinesiology & Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Liu Y, Qian SW, Tang Y, Tang QQ. The secretory function of adipose tissues in metabolic regulation. LIFE METABOLISM 2024; 3:loae003. [PMID: 39872218 PMCID: PMC11748999 DOI: 10.1093/lifemeta/loae003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 01/30/2025]
Abstract
In addition to their pivotal roles in energy storage and expenditure, adipose tissues play a crucial part in the secretion of bioactive molecules, including peptides, lipids, metabolites, and extracellular vesicles, in response to physiological stimulation and metabolic stress. These secretory factors, through autocrine and paracrine mechanisms, regulate various processes within adipose tissues. These processes include adipogenesis, glucose and lipid metabolism, inflammation, and adaptive thermogenesis, all of which are essential for the maintenance of the balance and functionality of the adipose tissue micro-environment. A subset of these adipose-derived secretory factors can enter the circulation and target the distant tissues to regulate appetite, cognitive function, energy expenditure, insulin secretion and sensitivity, gluconeogenesis, cardiovascular remodeling, and exercise capacity. In this review, we highlight the role of adipose-derived secretory factors and their signaling pathways in modulating metabolic homeostasis. Furthermore, we delve into the alterations in both the content and secretion processes of these factors under various physiological and pathological conditions, shedding light on potential pharmacological treatment strategies for related diseases.
Collapse
Affiliation(s)
- Yang Liu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shu-Wen Qian
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
29
|
Chaurasiya V, Nidhina Haridas PA, Olkkonen VM. Adipocyte-endothelial cell interplay in adipose tissue physiology. Biochem Pharmacol 2024; 222:116081. [PMID: 38408682 DOI: 10.1016/j.bcp.2024.116081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/02/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Adipose tissue (AT) expansion through hyperplasia or hypertrophy requires vascular remodeling that involves angiogenesis. There is quite some evidence that obese white AT (WAT) displays altered vasculature. Some studies suggest that this is associated with hypoxia, which is thought to play a role in inducing inflammatory activation of the excessively expanding WAT. Increasing evidence, based on genetic manipulations or treatments with inhibitory or activator pharmaceuticals, demonstrates that AT angiogenesis is crucial for AT metabolic function, and thereby for whole body metabolism and metabolic health. Despite some contradiction between studies, disturbance of WAT angiogenesis in obesity could be an important factor driving WAT dysfunction and the comorbidities of obesity. Endothelial cells (ECs) contribute to healthy WAT metabolism via transport of fatty acids and other plasma components, secretory signaling molecules, and extracellular vesicles (EVs). This communication is crucial for adipocyte metabolism and underscores the key role that the AT endothelium plays in systemic energy homeostasis and healthy metabolism. Adipocytes communicate towards the neighboring endothelium through several mechanisms. The pro-inflammatory status of hypertrophic adipocytes in obesity is reflected in ECs activation, which promotes chronic inflammation. On the other hand, adiponectin secreted by the adipocytes is important for healthy endothelial function, and adipocytes also secrete other pro- or anti-angiogenic effector molecules and a wealth of EVs - however, their detailed roles in signaling towards the endothelium are yet poorly understood. To conclude, targeting AT angiogenesis and promoting the healthy communication between adipocytes and ECs represent potentially promising strategies to treat obesity and its comorbidities.
Collapse
Affiliation(s)
- Vaishali Chaurasiya
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland
| | - P A Nidhina Haridas
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland.
| |
Collapse
|
30
|
Chen P, Li Y, Dai Y, Wang Z, Zhou Y, Wang Y, Li G. Advances in the Pathogenesis of Metabolic Liver Disease-Related Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:581-594. [PMID: 38525158 PMCID: PMC10960512 DOI: 10.2147/jhc.s450460] [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: 11/17/2023] [Accepted: 03/13/2024] [Indexed: 03/26/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer globally and the primary cause of death in cancer cases, with significant public health concern worldwide. Despite the overall decline in the incidence and mortality rates of HCC in recent years in recent years, the emergence of metabolic liver disease-related HCC is causing heightened concern, especially in countries like the United States, the United Kingdom, and P.R. China. The escalation of metabolic liver disease-related HCC is attributed to a combination of factors, including genetic predisposition, lifestyle choices, and changes in the living environment. However, the pathogenesis of metabolic liver disease-associated HCC remains imperfect. In this review, we encapsulate the latest advances and essential aspects of the pathogenesis of metabolic liver disease-associated HCC, including alcoholic liver disease (ALD), metabolic dysfunction-associated steatotic liver disease (MASLD), and inherited metabolic liver diseases.
Collapse
Affiliation(s)
- Pinggui Chen
- Department of Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, People’s Republic of China
| | - Yaoxuan Li
- Department of School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Yunyan Dai
- Department of Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, People’s Republic of China
| | - Zhiming Wang
- Department of Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, People’s Republic of China
| | - Yunpeng Zhou
- Department of Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, People’s Republic of China
| | - Yi Wang
- Department of Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, People’s Republic of China
| | - Gaopeng Li
- Department of Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, People’s Republic of China
| |
Collapse
|
31
|
Sung HK, Tang J, Jahng JWS, Song E, Chan YK, Lone AH, Peterson J, Abdul‐Sater A, Sweeney G. Ischemia-induced cardiac dysfunction is exacerbated in adiponectin-knockout mice due to impaired autophagy flux. Clin Transl Sci 2024; 17:e13758. [PMID: 38515365 PMCID: PMC10958170 DOI: 10.1111/cts.13758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 01/23/2024] [Accepted: 02/14/2024] [Indexed: 03/23/2024] Open
Abstract
Strategies to enhance autophagy flux have been suggested to improve outcomes in cardiac ischemic models. We explored the role of adiponectin in mediating cardiac autophagy under ischemic conditions induced by permanent coronary artery ligation. We studied the molecular mechanisms underlying adiponectin's cardio-protective effects in adiponectin knockout (Ad-KO) compared with wild-type (WT) mice subjected to ischemia by coronary artery ligation and H9c2 cardiomyocyte cell line exposed to hypoxia. Systemic infusion of a cathepsin-B activatable near-infrared probe as a biomarker for autophagy and detection via noninvasive three-dimensional fluorescence molecular tomography combined with computerized tomography to quantitate temporal changes, indicated increased activity in the myocardium of WT mice after myocardial infarction which was attenuated in Ad-KO. Seven days of ischemia increased myocardial adiponectin accumulation and elevated ULK1/AMPK phosphorylation and autophagy assessed by Western blotting for LC3 and p62, an outcome not observed in Ad-KO mice. Cell death, assessed by TUNEL analysis and the ratio of Bcl-2:Bax, plus cardiac dysfunction, measured using echocardiography with strain analysis, were exacerbated in Ad-KO mice. Using cellular models, we observed that adiponectin stimulated autophagy flux in isolated primary adult cardiomyocytes and increased basal and hypoxia-induced autophagy in H9c2 cells. Real-time temporal analysis of caspase-3/7 activation and caspase-3 Western blot indicated that adiponectin suppressed activation by hypoxia. Hypoxia-induced mitochondrial reactive oxygen species production and cell death were also attenuated by adiponectin. Importantly, the ability of adiponectin to reduce caspase-3/7 activation and cell death was not observed in autophagy-deficient cells generated by CRISPR-mediated deletion of Atg7. Collectively, our data indicate that adiponectin acts in an autophagy-dependent manner to attenuate cardiomyocyte caspase-3/7 activation and cell death in response to hypoxia in vitro and ischemia in mice.
Collapse
Affiliation(s)
| | - Jialing Tang
- Department of BiologyYork UniversityTorontoOntarioCanada
| | | | - Erfei Song
- Department of BiologyYork UniversityTorontoOntarioCanada
| | - Yee Kwan Chan
- Department of BiologyYork UniversityTorontoOntarioCanada
| | | | | | - Ali Abdul‐Sater
- School of Kinesiology and Health ScienceYork UniversityTorontoOntarioCanada
| | - Gary Sweeney
- Department of BiologyYork UniversityTorontoOntarioCanada
| |
Collapse
|
32
|
Jin L, Diaz-Canestro C, Wang Y, Tse MA, Xu A. Exerkines and cardiometabolic benefits of exercise: from bench to clinic. EMBO Mol Med 2024; 16:432-444. [PMID: 38321233 PMCID: PMC10940599 DOI: 10.1038/s44321-024-00027-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Regular exercise has both immediate and long-lasting benefits on cardiometabolic health, and has been recommended as a cornerstone of treatment in the management of diabetes and cardiovascular conditions. Exerkines, which are defined as humoral factors responsive to acute or chronic exercise, have emerged as important players conferring some of the multiple cardiometabolic benefits of exercise. Over the past decades, hundreds of exerkines released from skeletal muscle, heart, liver, adipose tissue, brain, and gut have been identified, and several exerkines (such as FGF21, IL-6, and adiponectin) have been exploited therapeutically as exercise mimetics for the treatment of various metabolic and cardiovascular diseases. Recent advances in metagenomics have led to the identification of gut microbiota, a so-called "hidden" metabolic organ, as an additional class of exerkines determining the efficacy of exercise in diabetes prevention, cardiac protection, and exercise performance. Furthermore, multiomics-based studies have shown the feasibility of using baseline exerkine signatures to predict individual responses to exercise with respect to metabolic and cardiorespiratory health. This review aims to explore the molecular pathways whereby exerkine networks mediate the cardiometabolic adaptations to exercise by fine-tuning inter-organ crosstalk, and discuss the roadmaps for translating exerkine-based discovery into the therapeutic application and personalized medicine in the management of the cardiometabolic disease.
Collapse
Affiliation(s)
- Leigang Jin
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Candela Diaz-Canestro
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Michael Andrew Tse
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Centre for Sports and Exercise, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.
- Department of Medicine, The University of Hong Kong, Hong Kong, China.
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
33
|
Pries R, Kosyna FK, Depping R, Plötze-Martin K, Lange C, Meyhöfer S, Meyhöfer SM, Marquardt JU, Bruchhage KL, Steffen A. Distinguishing the impact of distinct obstructive sleep apnea syndrome (OSAS) and obesity related factors on human monocyte subsets. Sci Rep 2024; 14:340. [PMID: 38172514 PMCID: PMC10764945 DOI: 10.1038/s41598-023-49921-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) and obesity go hand in hand in the majority of patients and both are associated with a systemic inflammation, immune disturbance and comorbidities such as cardiovascular disease. However, the unambiguous impact of OSAS and obesity on the individual inflammatory microenvironment and the immunological consequences of human monocytes has not been distinguished yet. Therefore, aim of this study was to investigate the impact of OSAS and obesity related factors on the inflammatory microenvironment by performing flow cytometric whole blood measurements of CD14/CD16 monocyte subsets in normal weight OSAS patients, patients with obesity but without OSAS, and patients with OSAS and obesity, compared to healthy donors. Moreover, explicitly OSAS and obesity related plasma levels of inflammatory mediators adiponectin, leptin, lipocalin and metalloproteinase-9 were determined and the influence of different OSAS and obesity related factors on cytokine secretion and expression of different adhesion molecules by THP-1 monocytes was analysed. Our data revealed a significant redistribution of circulating classical and intermediate monocytes in all three patient cohorts, but differential effects in terms of monocytic adhesion molecules CD11a, CD11b, CD11c, CX3CR1, CD29, CD49d, and plasma cytokine levels. These data were reflected by differential effects of OSAS and obesity related factors leptin, TNFα and hypoxia on THP-1 cytokine secretion patterns and expression of adhesion molecules CD11b and CD49d. In summary, our data revealed differential effects of OSAS and obesity, which underlines the need for a customized therapeutic regimen with respect to the individual weighting of these overlapping diseases.
Collapse
Affiliation(s)
- Ralph Pries
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| | - Friederike Katharina Kosyna
- Institute of Physiology, Working Group Hypoxia, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Reinhard Depping
- Institute of Physiology, Working Group Hypoxia, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Kirstin Plötze-Martin
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Christian Lange
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Svenja Meyhöfer
- Department of Medicine 1, University Hospital of Schleswig-Holstein, Lübeck, Germany
- Institute for Endocrinology & Diabetes, University Hospital of Schleswig-Holstein, Lübeck, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sebastian M Meyhöfer
- Institute for Endocrinology & Diabetes, University Hospital of Schleswig-Holstein, Lübeck, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jens U Marquardt
- Department of Medicine 1, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Armin Steffen
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| |
Collapse
|
34
|
Al Zein M, Zein O, Diab R, Dimachkie L, Sahebkar A, Al-Asmakh M, Kobeissy F, Eid AH. Intermittent fasting favorably modulates adipokines and potentially attenuates atherosclerosis. Biochem Pharmacol 2023; 218:115876. [PMID: 37871879 DOI: 10.1016/j.bcp.2023.115876] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
Adipose tissue is now recognized as an endocrine organ that secretes bioactive molecules called adipokines. These biomolecules regulate key physiological functions, including insulin sensitivity, energy metabolism, appetite regulation, endothelial function and immunity. Dysregulated secretion of adipokines is intimately associated with obesity, and translates into increased risk of obesity-related cardiovasculo-metabolic diseases. In particular, emerging evidence suggests that adipokine imbalance contributes to the pathogenesis of atherosclerosis. One of the promising diet regimens that is beneficial in the fight against obesity and cardiometabolic disorders is intermittent fasting (IF). Indeed, IF robustly suppresses inflammation, meditates weight loss and mitigates many aspects of the cardiometabolic syndrome. In this paper, we review the main adipokines and their role in atherosclerosis, which remains a major contributor to cardiovascular-associated morbidity and mortality. We further discuss how IF can be employed as an effective management modality for obesity-associated atherosclerosis. By exploring a plethora of the beneficial effects of IF, particularly on inflammatory markers, we present IF as a possible intervention to help prevent atherosclerosis.
Collapse
Affiliation(s)
- Mohammad Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Omar Zein
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rawan Diab
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Lina Dimachkie
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maha Al-Asmakh
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar
| | - Firas Kobeissy
- Department of Neurobiology and Neuroscience, Morehouse School of Medicine, Atlanta, GA, USA
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
35
|
Choubey M, Tirumalasetty MB, Bora NS, Bora PS. Linking Adiponectin and Its Receptors to Age-Related Macular Degeneration (AMD). Biomedicines 2023; 11:3044. [PMID: 38002042 PMCID: PMC10668948 DOI: 10.3390/biomedicines11113044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/26/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
In recent years, there has been a captivating focus of interest in elucidating the intricate crosstalk between adiponectin (APN), a versatile fat-associated adipokine and ocular pathologies. Unveiling the intricate relationship between adipocytokine APN and its receptors (AdipoRs) with aging eye disorders has emerged as a fascinating frontier in medical research. This review article delves into this connection, illuminating the hidden influence of APN on retinal health. This comprehensive review critically examines the latest findings and breakthroughs that underscore the pivotal roles of APN/AdipoRs signaling in maintaining ocular homeostasis and protecting against eye ailments. Here, we meticulously explore the intriguing mechanisms by which APN protein influences retinal function and overall visual acuity. Drawing from an extensive array of cutting-edge studies, the article highlights APN's multifaceted functions, ranging from anti-inflammatory properties and oxidative stress reduction to angiogenic regulation within retinal and macula tissues. The involvement of APN/AdipoRs in mediating these effects opens up novel avenues for potential therapeutic interventions targeting prevalent aging eye conditions. Moreover, this review unravels the interplay between APN signaling pathways and age-related macular degeneration (AMD). The single-cell RNA-seq results validate the expression of both the receptor isoforms (AdipoR1/R2) in retinal cells. The transcriptomic analysis showed lower expression of AdipoR1/2 in dry AMD pathogenesis compared to healthy subjects. The inhibitory adiponectin peptide (APN1) demonstrated over 75% suppression of CNV, whereas the control peptide did not exert any inhibitory effect on choroidal neovascularization (CNV). The elucidation of these relationships fosters a deeper understanding of adipose tissue's profound influence on ocular health, presenting new prospects for personalized treatments and preventative measures. Because APN1 inhibits CNV and leakage, it can be used to treat human AMD, although the possibility to treat human AMD is in the early stage and more clinical research is needed. In conclusion, this review provides a captivating journey into the enthralling world of APN, intertwining the realms of adipose biology and ophthalmology in aging.
Collapse
Affiliation(s)
- Mayank Choubey
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (M.C.); (M.B.T.)
| | - Munichandra B. Tirumalasetty
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (M.C.); (M.B.T.)
| | - Nalini S. Bora
- Department of Ophthalmology, Jones Eye Institute, Pat & Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR 72205, USA;
| | - Puran S. Bora
- Department of Ophthalmology, Jones Eye Institute, Pat & Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR 72205, USA;
| |
Collapse
|
36
|
Lai HC, Chen PH, Tang CH, Chen LW. Dipeptidyl Peptidase 4 Stimulation Induces Adipogenesis-Related Gene Expression of Adipose Stromal Cells. Int J Mol Sci 2023; 24:16101. [PMID: 38003291 PMCID: PMC10671339 DOI: 10.3390/ijms242216101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/01/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Adipogenesis has emerged as a new therapeutic target for regulating metabolism and achieving anti-inflammatory and anti-atherosclerotic effects via the release of adiponectin. However, at present, the effects and mechanism of action of dipeptidyl peptidase 4 (DPP4) stimulation on adiponectin production and adipogenesis have not been clarified. Here, we investigated the effects of DPP4 stimulation with monocyte chemoattractant protein-1 (MCP-1) on platelet-derived growth factor receptor alpha (PDGFRα) expression in adipose tissue and blood adiponectin levels. Stromal vascular fractions (SVFs) purified from human subcutaneous adipose tissue and inguinal adipose tissue of obese and diabetic (Leprdb/db) mice were treated with 50 ng of MCP-1 and plasma from control (Lepr+/+) mice supplemented with 10 ng or 50 ng of MCP-1. Treatment of SVFs from human subcutaneous adipose tissues with 50 ng of MCP-1 significantly increased AdipoQ, DPP4, peroxisome proliferator-activated receptor gamma (PPARγ), fatty-acid-binding protein (FABP4), and SERBF1 mRNA expression. MCP-1-supplemented plasma increased adiponectin, CCAAT-Enhancer-binding protein alpha (C/EBPα), DPP4, IL-33, and PDGFRα mRNA expression and adiponectin and DPP4 protein expression, while decreasing the expression of IL-10 mRNA in SVFs compared with the levels in the plasma treatment group. MCP-1-supplemented plasma was shown to increase PPARγ, PPARγ2, adiponectin, DPP4, and FABP4 and decrease IL-10 mRNA expression in PDGFRα cells from adipose tissue. Meanwhile, MCP-1-supplemented plasma increased MCP-1, PDGFRα, TNFα, adiponectin, and IL-1β and decreased IL-10 and FOXP3 mRNA expression in DPP4 cells. Moreover, the injection of MCP-1-supplemented plasma into adipose tissue increased the proportion of DPP4+ cells among PDGFRα+ cells from adipose tissue and plasma adiponectin levels of Leprdb/db mice compared with the levels in the plasma injection group. Our results demonstrate that DPP4+ cells are important adipose progenitor cells. Stimulation of DPP4 with MCP-1 increases adipogenesis-related gene expression and the population of DPP4+ cells among PDGFRα+ cells in SVFs and blood adiponectin levels. DPP4 stimulation could be a novel therapy to increase local adipogenesis and systemic adiponectin levels.
Collapse
Affiliation(s)
- Hsiao-Chi Lai
- Department of Surgery, Kaohsiung Veterans General Hospital, No. 386, Ta-Chung 1st Road, Kaohsiung 813, Taiwan
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei 112, Taiwan
| | - Pei-Hsuan Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, No. 386, Ta-Chung 1st Road, Kaohsiung 813, Taiwan
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei 112, Taiwan
| | - Chia-Hua Tang
- Department of Surgery, Kaohsiung Veterans General Hospital, No. 386, Ta-Chung 1st Road, Kaohsiung 813, Taiwan
| | - Lee-Wei Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, No. 386, Ta-Chung 1st Road, Kaohsiung 813, Taiwan
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei 112, Taiwan
- Department of Biological Sciences, National Sun Yat-Sen University, No. 70, Lien-Hai Road, Kaohsiung 804, Taiwan
| |
Collapse
|
37
|
Wu X, Tao Y, Ren Y, Zhang Z, Zhao Y, Tian Y, Li Y, Hou M, Guo Y, Gong Y, Zhang Y, Li D, Li H, Jiang R, Li G, Liu X, Kang X, Tian Y. Adiponectin inhibits GnRH secretion via activating AMPK and PI3K signaling pathways in chicken hypothalamic neuron cells. Poult Sci 2023; 102:103028. [PMID: 37660449 PMCID: PMC10491727 DOI: 10.1016/j.psj.2023.103028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023] Open
Abstract
It has been reported that adiponectin (AdipoQ), an adipokine secreted by white adipose tissue, plays an important role in the control of animal reproduction in addition to its function in energy homeostasis by binding to its receptors AdipoR1/2. However, the molecular mechanisms of AdipoQ in the regulation of animal reproduction remain elusive. In this study, we investigated the effects of AdipoQ on hypothalamic reproductive hormone (GnRH) secretion and reproduction-related receptor gene (estrogen receptor [ER] and progesterone receptor [PR]) expression in hypothalamic neuronal cells (HNCs) of chickens by using real-time fluorescent quantitative PCR (RT-qPCR), enzyme-linked immunosorbent assay (ELISA), Western blot (WB) and cell counting kit-8 (CCK-8) assays and found that overexpression of AdipoQ could increase the expression levels of AdipoR1/2 and reproduction-related receptor genes (P < 0.05) while decreasing the expression level of GnRH. In contrast, interference with AdipoQ mRNA showed the opposite results in HNCs. Furthermore, we demonstrated that AdipoQ exerts its functions through the AMPK and PI3K signaling pathways. Finally, our in vitro experiments found that AdipoRon (a synthetic substitute for AdipoQ) treatment and AdipoR1/2 RNAi interference co-treatment resulted in no effect on GnRH secretion, suggesting that the inhibition of GnRH secretion by AdipoQ is mediated by the AdipoR1/2 signaling axis. In summary, we uncovered, for the first time, the molecular mechanism of AdipoQ in the regulation of reproductive hormone secretion in hypothalamic neurons in chickens.
Collapse
Affiliation(s)
- Xing Wu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yiqing Tao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yangguang Ren
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Zihao Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yudian Zhao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yixiang Tian
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Yijie Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Meng Hou
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yulong Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yujie Gong
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yanhua Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Donghua Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Hong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Ruirui Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Guoxi Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xiaojun Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China.
| |
Collapse
|
38
|
An Y, Cao B, Li K, Xu Y, Zhao W, Zhao D, Ke J. A Prediction Model for Sight-Threatening Diabetic Retinopathy Based on Plasma Adipokines among Patients with Mild Diabetic Retinopathy. J Diabetes Res 2023; 2023:8831609. [PMID: 37920605 PMCID: PMC10620016 DOI: 10.1155/2023/8831609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/13/2023] [Accepted: 08/24/2023] [Indexed: 11/04/2023] Open
Abstract
Background Accumulating evidence has suggested a link between adipokines and diabetic retinopathy (DR). This study is aimed at investigating the risk factors for sight-threatening DR (STDR) and establishing a prognostic model for predicting STDR among a high-risk population of patients with type 2 diabetes mellitus (T2DM). Methods Plasma concentrations of adipokines were determined by enzyme-linked immunosorbent assay. In the case-control set, principal component analysis (PCA) was performed to select optimal predictive cytokines for STDR, involving severe nonproliferative DR (NPDR) and proliferative DR. Support vector machine (SVM) was used to examine the possible combination of baseline plasma adipokines to discriminate the patients with mild NPDR who will later develop STDR. An individual prospective cohort with a follow-up period of 3 years was used for the external validation. Results In both training and testing sets, involving 306 patients with T2DM, median levels of plasma adiponectin (APN), leptin, and fatty acid-binding protein 4 (FABP4) were significantly higher in the STDR group than those in mild NPDR. Except for adipsin, the other three adipokines, FABP4, APN, and leptin, were selected by PCA and integrated into SVM. The accuracy of the multivariate SVM classification model was acceptable in both the training set (AUC = 0.81, sensitivity = 71%, and specificity = 91%) and the testing set (AUC = 0.77, sensitivity = 61%, and specificity = 92%). 110 T2DM patients with mild NPDR, the high-risk population of STDR, were enrolled for external validation. Based on the SVM, the risk of each patient was calculated. More STDR occurred in the high-risk group than in the low-risk group, which were grouped by the median value of APN, FABP4, and leptin, respectively. The model was validated in an individual cohort using SVM with the AUC, sensitivity, and specificity reaching 0.77, 64%, and 91%, respectively. Conclusions Adiponectin, leptin, and FABP4 were demonstrated to be associated with the severity of DR and maybe good predictors for STDR, suggesting that adipokines may play an important role in the pathophysiology of DR development.
Collapse
Affiliation(s)
- Yaxin An
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Bin Cao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Kun Li
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - Yongsong Xu
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - Wenying Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| |
Collapse
|
39
|
Fagian Pansani V, Dolfini Celim LB, Amorim Oliveira G, Rosa Degasperi G. Adiponectin: A "Friendly adipokine" in Diabetic Retinopathy? Semin Ophthalmol 2023; 38:602-609. [PMID: 37157861 DOI: 10.1080/08820538.2023.2205929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 05/10/2023]
Abstract
PURPOSE Adiponectin has also been associated with diabetic retinopathy, a diabetic microvascular complication. However, the mechanism of action of adiponectin in retinopathy is still under investigation. This review summarizes emerging evidence on the association with diabetic retinopathy in type 2 diabetes. METHODS We reviwed papers from 2004 to 2022 and included studies related to retinopathy and its association with blood and intraocular adiponectin in type 2 diabetes. RESULTS Most of the studies analyzed in this review suggested an association between the diabetic retinopathy progression and intraocular, serum, or plasma adiponectin levels. Increased levels of adiponectin contributed to the development of the disease in diabetic patients. In a minority of studies, it was indicated an inversely proportional relationship between adiponectin concentration and diabetic retinopathy severity. CONCLUSION The high levels of adiponectin in diabetic patients may be related to the decrease in renal clearance. Under this situation, if the predominant isoform is globular adiponectin, this may explain the retinopathy progression, considering a pro-inflammatory response induced by this isoform. However, the actions of adiponectin in diabetic retinopathy pathophysiology are still controversial.
Collapse
Affiliation(s)
- Victor Fagian Pansani
- Centro de Ciências da Saúde, Faculdade de Medicina, Pontifícia Universidade Católica de Campinas (PUC-Campinas), Campinas, SP, Brazil
| | | | | | | |
Collapse
|
40
|
Sasaki T, Nishimoto Y, Hirata T, Abe Y, Hirose N, Takayama M, Takebayashi T, Okano H, Arai Y. Status and physiological significance of circulating adiponectin in the very old and centenarians: an observational study. eLife 2023; 12:e86309. [PMID: 37768324 PMCID: PMC10564453 DOI: 10.7554/elife.86309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 09/28/2023] [Indexed: 09/29/2023] Open
Abstract
Background High levels of circulating adiponectin are associated with increased insulin sensitivity, low prevalence of diabetes, and low body mass index (BMI); however, high levels of circulating adiponectin are also associated with increased mortality in the 60-70 age group. In this study, we aimed to clarify factors associated with circulating high-molecular-weight (cHMW) adiponectin levels and their association with mortality in the very old (85-89 years of age) and centenarians. Methods The study included 812 (women: 84.4%) for centenarians and 1498 (women: 51.7%) for the very old. The genomic DNA sequence data were obtained by whole-genome sequencing or DNA microarray-imputation methods. Least Absolute Shrinkage and Selection Operator (LASSO) and multivariate regression analyses were used to evaluate cHMW adiponectin characteristics and associated factors. All-cause mortality was analyzed in three quantile groups of cHMW adiponectin levels using Cox regression. Results The cHMW adiponectin levels were increased significantly beyond 100 years of age, were negatively associated with diabetes prevalence, and were associated with SNVs in CDH13 (p=2.21 × 10-22) and ADIPOQ (p=5.72 × 10-7). Multivariate regression analysis revealed that genetic variants, BMI, and high-density lipoprotein cholesterol (HDLC) were the main factors associated with cHMW adiponectin levels in the very old, whereas the BMI showed no association in centenarians. The hazard ratios for all-cause mortality in the intermediate and high cHMW adiponectin groups in very old men were significantly higher rather than those for all-cause mortality in the low-level cHMW adiponectin group, even after adjustment with BMI. In contrast, the hazard ratios for all-cause mortality were significantly higher for high cHMW adiponectin groups in very old women, but were not significant after adjustment with BMI. Conclusions cHMW adiponectin levels increased with age until centenarians, and the contribution of known major factors associated with cHMW adiponectin levels, including BMI and HDLC, varies with age, suggesting that its physiological significance also varies with age in the oldest old. Funding This study was supported by grants from the Ministry of Health, Welfare, and Labour for the Scientific Research Projects for Longevity; a Grant-in-Aid for Scientific Research (No 21590775, 24590898, 15KT0009, 18H03055, 20K20409, 20K07792, 23H03337) from the Japan Society for the Promotion of Science; Keio University Global Research Institute (KGRI), Kanagawa Institute of Industrial Science and Technology (KISTEC), Japan Science and Technology Agency (JST) Research Complex Program "Tonomachi Research Complex" Wellbeing Research Campus: Creating new values through technological and social innovation (JP15667051), the Program for an Integrated Database of Clinical and Genomic Information from the Japan Agency for Medical Research and Development (No. 16kk0205009h001, 17jm0210051h0001, 19dk0207045h0001); the medical-welfare-food-agriculture collaborative consortium project from the Japan Ministry of Agriculture, Forestry, and Fisheries; and the Biobank Japan Program from the Ministry of Education, Culture, Sports, and Technology.
Collapse
Affiliation(s)
- Takashi Sasaki
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Yoshinori Nishimoto
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo, Japan
- Department of Neurology, Keio University, School of Medicine, Tokyo, Japan
| | - Takumi Hirata
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo, Japan
- Institute for Clinical and Translational Science, Nara Medical University, Nara, Japan
| | - Yukiko Abe
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo, Japan
| | | | - Michiyo Takayama
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo, Japan
- Center for Preventive Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toru Takebayashi
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Yasumichi Arai
- Faculty of Nursing and Medical Care, Keio University, Tokyo, Japan
| |
Collapse
|
41
|
Kou R, Wang J, Li A, Wang Y, Zhang B, Liu J, Sun Y, Wang S. Ameliorating Effects of Bifidobacterium longum subsp. infantis FB3-14 against High-Fat-Diet-Induced Obesity and Gut Microbiota Disorder. Nutrients 2023; 15:4104. [PMID: 37836387 PMCID: PMC10574542 DOI: 10.3390/nu15194104] [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: 08/30/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Obesity has emerged as one of the most prevalent chronic diseases worldwide. Our study was conducted to investigate the anti-obese potential of novel probiotic Bifidobacterium longum subsp. infantis FB3-14 (FB3-14) and the underlying molecular mechanisms in high-fat diet (HFD)-fed mice. The results demonstrated that an 8-week FB3-14 intervention significantly suppressed the HFD-induced body and fat weight gain and abnormal alterations of the serum lipid parameter, restoring the levels of cholesterol (4.29 mmol/L) and low-density lipoprotein cholesterol (3.42 mmol/L). FB3-14 treatment also attenuated adipocyte expansion, hepatic injury, and low-grade systemic inflammation and restored the expressions of lipid-metabolism-related genes, including Hsl, Leptin, and Adiponectin. Furthermore, FB3-14 was observed to reduce the Firmicutes/Bacteroidetes ratio in obese mice; increase the abundance of Akkermansia muciniphila, unclassified_Muribaculaceae, Lachnospiraceae_NK4A136_group, and Bifidobacterim; and upregulate G protein-coupled receptor41 associated with higher levels of butyric acid. These results indicate the protective effectiveness of FB3-14 in HFD-driven obesity and gut microbiota disorders, highlighting the promising potential of FB3-14 as a functional nutrition supplement.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300350, China; (R.K.); (J.W.); (A.L.); (Y.W.); (B.Z.); (J.L.); (Y.S.)
| |
Collapse
|
42
|
He J, Li X. Relationship between chronic obstructive pulmonary disease and adiponectin concentrations: An updated meta-analysis and single-cell RNA sequencing. Medicine (Baltimore) 2023; 102:e34825. [PMID: 37603523 PMCID: PMC10443756 DOI: 10.1097/md.0000000000034825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/28/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Adipose tissue, being an organ of the endocrine system, can influence the severity of chronic obstructive pulmonary disease (COPD). Even though several inflammatory markers can potentially significantly influence lung function, the precise function of adipokines, like adiponectin, in COPD is still disputed. To analyze the association of COPD with adiponectin concentrations, a meta-analysis of the most recent literature and single-cell sequencing data were conducted. METHODS Studies in Embase, PubMed, Cochrane Library, and Web of Science were browsed to obtain relevant data, which were then assessed with the aid of R 4.1.3 and STATA 11.0 software. Standardized mean differences and correlation coefficients aided the analysis of effect values. Moreover, a single-cell sequencing GSE136831 dataset was retrieved to ascertain the mRNA expression of adiponectin gene (ADIPOQ) in the lung tissue of COPD patients to confirm the difference in the expression of adiponectin between the case and control groups. RESULTS This meta-analysis comprised 18 publications involving 24 studies. The overall combined data established the concentration of plasma/serum adiponectin as significantly higher in patients with COPD compared to healthy subjects. Subgroup analyses based on disease status, specimen type, ethnicity, study design method, measurement method, and age of COPD patients demonstrated that all patients with COPD had elevated levels of adiponectin compared to healthy controls. When subgroup analysis was performed for gender alone, the results depicted that male COPD patients had significantly higher adiponectin than healthy males, while female patients of COPD had elevated adiponectin compared to healthy females. Furthermore, it was found that plasma/serum adiponectin appeared to be positively correlated with tumor necrosis factor-α, and it was negatively correlated with FEV1% and FEV1/FVC. The results of single-cell sequencing data suggested that ADIPOQ mRNA was mainly expressed in alveolar epithelial cells, and the level of ADIPOQ mRNA was higher in lung tissues of patients with COPD than in lung tissues of healthy subjects. CONCLUSION This meta-analysis suggests that the levels of plasma/serum adiponectin are significantly elevated in patients with COPD versus controls. Tumor necrosis factor-α, FEV1/FVC, and FEV1% may all be associated with the concentrations of adiponectin.
Collapse
Affiliation(s)
- Jie He
- Clinical Medical College of Chengdu Medical College, Chengdu, China
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Key Laboratory of Geriatric Respiratory Diseases of Sichuan Higher Education Institutes, Chengdu, China
| | - Xuemei Li
- Clinical Medical College of Chengdu Medical College, Chengdu, China
- Neurosurgery department, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
43
|
Choubey M, Bora P. Emerging Role of Adiponectin/AdipoRs Signaling in Choroidal Neovascularization, Age-Related Macular Degeneration, and Diabetic Retinopathy. Biomolecules 2023; 13:982. [PMID: 37371562 DOI: 10.3390/biom13060982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/15/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Age-related macular degeneration (AMD), a leading cause of irreversible blindness in adults, may result in poor central vision, making it difficult to see, read, and drive. AMD is generally classified in either dry or wet types. Milder cases of dry AMD may progress to geographic atrophy (GA), leading to significant visual disability; wet, or neovascular AMD, which involves choroidal neovascularization (CNV), can lead to complete loss of central vision. Adiponectin (APN) discovery in the mid-1990's and, subsequently, its two cognate receptors (AdipoRs) in the early 2000s have led to a remarkable progress in better understanding metabolic disorders, as well as metabolism-associated ocular pathology. APN/AdipoRs signaling plays a central role in a variety of molecular and cellular physiological events, including glucose and lipid metabolism, whole-body energy regulation, immune and inflammation responses, insulin sensitivity and retinal cell biological functions. This review is an amalgamation of recent information related to APN/AdipoRs in the pathophysiology of retinal diseases and furthers its association with AMD and diabetic retinopathy. Additionally, we present our original research, where we designed control peptide and CNV inhibitory peptide from the globular region of APN to see the effect of these peptides on the mouse model of laser-induced CNV. The inhibitory peptide (APN1) inhibited CNV by more than 75% while the control peptide did not inhibit CNV.
Collapse
Affiliation(s)
- Mayank Choubey
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
| | - Puran Bora
- Pat & Willard Walker Eye Research Center, Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR 72205, USA
| |
Collapse
|
44
|
Quispe R, Sweeney T, Martin SS, Jones SR, Allison MA, Budoff MJ, Ndumele CE, Elshazly MB, Michos ED. Associations of Adipokine Levels with Levels of Remnant Cholesterol: the Multi-Ethnic Study of Atherosclerosis (MESA). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.24.23289072. [PMID: 37162928 PMCID: PMC10168480 DOI: 10.1101/2023.04.24.23289072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Background The metabolic syndrome phenotype of individuals with obesity is characterized by elevated levels of triglyceride (TG)-rich lipoproteins and remnant particles, which have been shown to be significantly atherogenic. Understanding the association between adipokines, endogenous hormones produced by adipose tissue, and remnant cholesterol (RC) would give insight into the link between obesity and atherosclerotic cardiovascular disease. Methods We studied 1,791 MESA participants of an ancillary study on body composition who had adipokine levels measured (leptin, adiponectin, resistin) at either visit 2 or 3. RC was calculated as non-high density lipoprotein cholesterol minus low-density lipoprotein cholesterol (LDL-C), measured at the same visit as the adipokines, as well as subsequent visits 4 through 6. Multivariable-adjusted linear mixed effects models were used to assess the cross-sectional and longitudinal associations between adipokines and levels of RC. Results Mean (SD) age was 64.5±9.6 years and for body mass index (BMI) was 29.9±5.0 kg/m2; 52.0% were women. In fully adjusted models that included BMI, LDL-C and lipid-lowering therapy, for each 1-unit increment in adiponectin, there was 14.4% (12.0, 16.8) lower RC. With each 1-unit increment in leptin and resistin, there was 4.5% (2.3, 6.6) and 5.1% (1.2, 9.2) higher RC, respectively. Lower adiponectin and higher leptin were also associated with longitudinal increases in RC levels over median follow-up of 5(4-8) years. Conclusions Lower adiponectin and higher leptin levels were independently associated with higher levels of RC at baseline and longitudinal RC increase, even after accounting for BMI and LDL-C. CLINICAL PERSPECTIVE What is new?: - Among individuals without history of cardiovascular disease, adiponectin is inversely associated with cross-sectional levels of remnant cholesterol, whereas leptin and resistin are directly associated.- Adiponectin had an inverse association with progression of remnant cholesterol levels over time.What are the clinical implications?: - Adiponectin levels were not associated with LDL-C levels but with levels of triglyceride-rich lipoproteins, particularly remnant cholesterol.-Incrementing adiponectin via lifestyle modification and/or pharmacological therapies (i.e. GLP-1 agonists) could be a mechanism to reduce remnant cholesterol levels and ultimately cardiovascular risk.
Collapse
|
45
|
Michel LYM. Extracellular Vesicles in Adipose Tissue Communication with the Healthy and Pathological Heart. Int J Mol Sci 2023; 24:ijms24097745. [PMID: 37175451 PMCID: PMC10177965 DOI: 10.3390/ijms24097745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/11/2023] [Accepted: 04/16/2023] [Indexed: 05/15/2023] Open
Abstract
Adipose tissue and its diverse cell types constitute one of the largest endocrine organs. With multiple depot locations, adipose tissue plays an important regulatory role through paracrine and endocrine communication, particularly through the secretion of a wide range of bioactive molecules, such as nucleic acids, proteins, lipids or adipocytokines. Over the past several years, research has uncovered a myriad of interorgan communication signals mediated by small lipid-derived nanovesicles known as extracellular vesicles (EVs), in which secreted bioactive molecules are stably transported as cargo molecules and delivered to adjacent cells or remote organs. EVs constitute an essential part of the human adipose secretome, and there is a growing body of evidence showing the crucial implications of adipose-derived EVs in the regulation of heart function and its adaptative capacity. The adipose tissue modifications and dysfunction observed in obesity and aging tremendously affect the adipose-EV secretome, with important consequences for the myocardium. The present review presents a comprehensive analysis of the findings in this novel area of research, reports the key roles played by adipose-derived EVs in interorgan cross-talk with the heart and discusses their implications in physiological and pathological conditions affecting adipose tissue and/or the heart (pressure overload, ischemia, diabetic cardiomyopathy, etc.).
Collapse
Affiliation(s)
- Lauriane Y M Michel
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 57 Avenue Hippocrate, 1200 Brussels, Belgium
| |
Collapse
|
46
|
Munhoz AC, Serna JDC, Vilas-Boas EA, Caldeira da Silva CC, Santos TG, Mosele FC, Felisbino SL, Martins VR, Kowaltowski AJ. Adiponectin reverses β-Cell damage and impaired insulin secretion induced by obesity. Aging Cell 2023:e13827. [PMID: 37060190 DOI: 10.1111/acel.13827] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 04/16/2023] Open
Abstract
Obesity significantly decreases life expectancy and increases the incidence of age-related dysfunctions, including β-cell dysregulation leading to inadequate insulin secretion. Here, we show that diluted plasma from obese human donors acutely impairs β-cell integrity and insulin secretion relative to plasma from lean subjects. Similar results were observed with diluted sera from obese rats fed ad libitum, when compared to sera from lean, calorically restricted, animals. The damaging effects of obese circulating factors on β-cells occurs in the absence of nutrient overload, and mechanistically involves mitochondrial dysfunction, limiting glucose-supported oxidative phosphorylation and ATP production. We demonstrate that increased levels of adiponectin, as found in lean plasma, are the protective characteristic preserving β-cell function; indeed, sera from adiponectin knockout mice limits β-cell metabolic fluxes relative to controls. Furthermore, oxidative phosphorylation and glucose-sensitive insulin secretion, which are completely abrogated in the absence of this hormone, are restored by the presence of adiponectin alone, surprisingly even in the absence of other serological components, for both the insulin-secreting INS1 cell line and primary islets. The addition of adiponectin to cells treated with plasma from obese donors completely restored β-cell functional integrity, indicating the lack of this hormone was causative of the dysfunction. Overall, our results demonstrate that low circulating adiponectin is a key damaging element for β-cells, and suggest strong therapeutic potential for the modulation of the adiponectin signaling pathway in the prevention of age-related β-cell dysfunction.
Collapse
Affiliation(s)
- Ana Cláudia Munhoz
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Julian D C Serna
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Tiago G Santos
- Centro Internacional de Pesquisa (CIPE), A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Francielle C Mosele
- Instituto de Biociências de Botucatu (IBB), Universidade Estadual Paulista (UNESP), Botucatu, Brazil
| | - Sergio L Felisbino
- Instituto de Biociências de Botucatu (IBB), Universidade Estadual Paulista (UNESP), Botucatu, Brazil
| | - Vilma Regina Martins
- Centro Internacional de Pesquisa (CIPE), A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
47
|
Zhong Y, Chen X, Huang C, Chen Y, Zhao F, Hao R, Wang N, Liao W, Xia H, Yang L, Wang S, Sun G. The effects of a low carbohydrate diet combined with partial meal replacement on obese individuals. Nutr Metab (Lond) 2023; 20:18. [PMID: 36997952 PMCID: PMC10064565 DOI: 10.1186/s12986-023-00740-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/20/2023] [Indexed: 04/01/2023] Open
Abstract
OBJECTIVE We explored the dietary effects of replacing normal dietary staple foods with supplementary nutritional protein powder, dietary fiber, and fish oil on several metabolic parameters. We examined weight loss, glucose and lipid metabolism, and intestinal flora in obese individuals when compared with individuals on a reduced staple food low carbohydrate diet. METHODS From inclusion and exclusion criteria, 99 participants (28 kg/m2 ≤ body mass index (BMI) ≤ 35 kg/m2) were recruited and randomly assigned to control and intervention 1 and 2 groups. Physical examinations and biochemical indices were performed/gathered before the intervention and at 4 and 13 weeks post intervention. After 13 weeks, feces was collected and 16s rDNA sequenced. RESULTS After 13 weeks, when compared with controls, body weight, BMI, waist circumference, hip circumference, systolic blood pressure, and diastolic blood pressure values in intervention group 1 were significantly reduced. In intervention group 2, body weight, BMI, waist circumference, and hip circumference were significantly reduced. Triglyceride (TG) levels in both intervention groups were significantly reduced. Fasting blood glucose, glycosylated hemoglobin, glycosylated albumin, total cholesterol, and apolipoprotein B levels in intervention group 1 were decreased, while high density lipoprotein cholesterol (HDL-c) decreased slightly. Glycosylated albumin, TG, and total cholesterol levels in intervention group 2 decreased, while HDL-c decreased slightly, High sensitive C-reactive protein, MPO, Ox-LDL, LEP, TGF-β1, IL-6, GPLD1, pro NT, GPC-4, and LPS levels in both intervention groups were lower when compared with controls. Adiponectin (ADPN) levels in intervention groups were higher when compared with controls. Tumor necrosis factor-α (TNF-α) levels in intervention group 1 were lower when compared with controls. There is no obvious difference in α diversity and β diversity between intestinal flora of 3 groups. Among the first 10 species of Phylum, only the control group and the intervention group 2 had significantly higher Patescibacteria than the intervention group 1. Among the first 10 species of Genus, only the number of Agathobacter in intervention group 2 was significantly higher than that in control group and intervention group 1. CONCLUSIONS We showed that an LCD, where nutritional protein powder replaced some staple foods and dietary fiber and fish oil were simultaneously supplemented, significantly reduced weight and improved carbohydrate and lipid metabolism in obese individuals when compared with an LCD which reduced staple food intake.
Collapse
Affiliation(s)
- Yulian Zhong
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Ximin Chen
- Beijing Institute of Nutritional Resources, Beijing, 100069, China
| | - Chao Huang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yuexiao Chen
- Institute of Biotechnology and Health, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Fengyi Zhao
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Runhua Hao
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Niannian Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Wang Liao
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Hui Xia
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Ligang Yang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Shaokang Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China.
| | - Guiju Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| |
Collapse
|
48
|
Sheng C, Liu T, Chen S, Liao M, Yang P. The neglected association between central obesity markers and abdominal aortic aneurysm presence: A systematic review and meta-analysis. Front Cardiovasc Med 2023; 10:1044560. [PMID: 36844737 PMCID: PMC9947524 DOI: 10.3389/fcvm.2023.1044560] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/27/2023] [Indexed: 02/11/2023] Open
Abstract
Purpose To review the association between central obesity and abdominal aortic aneurysm (AAA). Materials and methods The PubMed, Web of Sciences, Embase, The China national knowledge infrastructure (CNKI), and Cochrane Library were searched up to April 30, 2022. Researches includes investigation of the relationship between central obesity markers and AAA. Included studies must use recognized measures of central obesity, i.e., waist circumference (WC) and waist-to-hip ratio (WHR), or use imaging techniques to calculate abdominal fat distribution, such as computed tomography (CT) imaging. Results Eleven clinical researches were identified of which eight discussed the association between physical examination and AAA, and three studies mainly focused on abdominal fat volume (AFV). Seven researches concluded that there was a positive correlation between markers of central obesity and AAA. Three studies found no significant link between markers of central obesity and AAA. One of the remaining studies reported different results for each sex. Three studies pooled in a meta-analysis identified correlation between central obesity and AAA presence (RR = 1.29; 95% confidence interval, 1.14-1.46). Conclusion Central obesity plays a role in the risk of AAA. Standardized central obesity markers may be predictors of AAA. However, there was no association between abdominal fat volume and AAA. Additional relevant evidence and specific mechanisms warrant further study. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?IDCRD42022332519, identifier CRD42022332519.
Collapse
Affiliation(s)
- Chang Sheng
- Department of Vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tinghua Liu
- Department of Vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shen Chen
- Department of Vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mingmei Liao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China,Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China,*Correspondence: Mingmei Liao,
| | - Pu Yang
- Department of Vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China,Pu Yang,
| |
Collapse
|
49
|
Vliora M, Ravelli C, Grillo E, Corsini M, Flouris AD, Mitola S. The impact of adipokines on vascular networks in adipose tissue. Cytokine Growth Factor Rev 2023; 69:61-72. [PMID: 35953434 DOI: 10.1016/j.cytogfr.2022.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 02/07/2023]
Abstract
Adipose tissue (AT) is a highly active and plastic endocrine organ. It secretes numerous soluble molecules known as adipokines, which act locally to AT control the remodel and homeostasis or exert pleiotropic functions in different peripheral organs. Aberrant production or loss of certain adipokines contributes to AT dysfunction associated with metabolic disorders, including obesity. The AT plasticity is strictly related to tissue vascularization. Angiogenesis supports the AT expansion, while regression of blood vessels is associated with AT hypoxia, which in turn mediates tissue inflammation, fibrosis and metabolic dysfunction. Several adipokines can regulate endothelial cell functions and are endowed with either pro- or anti-angiogenic properties. Here we address the role of adipokines in the regulation of angiogenesis. A better understanding of the link between adipokines and angiogenesis will open the way for novel therapeutic approaches to treat obesity and metabolic diseases.
Collapse
Affiliation(s)
- Maria Vliora
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece; Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Andreas D Flouris
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy.
| |
Collapse
|
50
|
Zhou XH, Cheng ZP, Lu M, Lin WY, Luo LL, Ming ZY, Hu Y. Adiponectin receptor agonist AdipoRon modulates human and mouse platelet function. Acta Pharmacol Sin 2023; 44:356-366. [PMID: 35918410 PMCID: PMC9889809 DOI: 10.1038/s41401-022-00943-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/14/2022] [Indexed: 02/04/2023]
Abstract
Adiponectin, an adipokine secreted by adipocytes, has anti-atherosclerotic and antithrombotic activities. AdipoRon is synthetic small molecule adiponectin receptor agonist. In this study, we investigated the effect of AdipoRon on platelet activation and thrombus formation. Washed human platelets were prepared from the peripheral blood of healthy donors. In a series of in vitro platelet functional assays, pre-treatment with AdipoRon (10, 20, 40 µg/mL) dose-dependently inhibited the aggregation, granule secretion and spreading of washed human platelets. We showed that AdipoRon (20, 40 µg/mL) significantly inhibited AMPK, Syk, PLCγ2, PI3K, Akt, p38-MAPK and ERK1/2 signalling pathways in washed human platelets. In addition, we demonstrated that the phosphorylation of CKII at Tyr255 was an important mechanism of the integrin αIIbβ3-mediated platelet activation. Meanwhile, AdipoR1 deficiency impaired the inhibitory effect of AdipoRon on mouse platelets. In ferric chloride-induced carotid injury model, injection of AdipoRon (5 or 12.5 mg/kg, iv) significantly attenuated arterial thrombosis. In conclusion, AdipoRon attenuates platelet function via the AdipoR1/AMPK/CKII/PI3K/AKT signalling pathways, while exerting a protective effect against arterial thrombosis. This study offers new insights into the fields of cardiovascular disease and antiplatelet drug discovery.Schematic model of AdipoRon regulating platelet activation. (BioRender.com).
Collapse
Affiliation(s)
- Xiang-Hui Zhou
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhi-Peng Cheng
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meng Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen-Yi Lin
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Li Luo
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhang-Yin Ming
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, China
- Tongji-Rongcheng Center for Biomedicine, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China.
| |
Collapse
|