1
|
Kumari S, Peela S, Srilatha M, Girish BP, Nagaraju GP. Adiponectin: its role in diabetic and pancreatic cancer. Mol Aspects Med 2025; 103:101370. [PMID: 40403652 DOI: 10.1016/j.mam.2025.101370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/24/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
Adiponectin (ApN) is an antidiabetic and anti-inflammatory protein synthesized by adipose tissue. It is essential in regulating insulin sensitivity, glucose, and lipid metabolism by controlling AMPK, PPARα, and MAPK signals. It is an anti-inflammatory property that protects pancreatic β-cells. Often, low levels of ApN are linked to obesity, type II diabetes and the development of PDAC. However, changes in lifestyle and the use of certain drugs can improve ApN function and insulin sensitivity. PDAC is a highly aggressive cancer linked to obesity, type II diabetes, and insulin resistance. ApN plays a complex role in PDAC progression and can suppress PDAC development by weakening β-catenin signaling. Decreases in ApN levels are associated with increased PDAC risk in diabetic patients. PDAC and diabetes are interconnected through the development of insulin resistance, islet dysfunction, change in immunological response, inflammation, oxidative stress, and altered hormone secretion. Genetic studies highlight specific genes like HNF4G and PDX1 that influence both conditions and miRNAs such as miR-19a promote tumor progression through the PI3K/AKT pathway. This review discusses the role of ApN in diabetes and PDAC and the interrelation between diabetes and PDAC.
Collapse
Affiliation(s)
- Seema Kumari
- Department of Biotechnology, Dr.B.R. Ambedkar University, Srikakulam, 532410, AP, India
| | - Sujatha Peela
- Department of Biotechnology, Dr.B.R. Ambedkar University, Srikakulam, 532410, AP, India
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh, 517502, India
| | - Bala Prabhakar Girish
- Regional Agricultural Research Station, Institute of Frontier Technology, Acharya N G Ranga Agricultural University, Tirupati, India
| | - Ganji Purnachandra Nagaraju
- School of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
2
|
Naidu D, Althaf Umar KP, Muhsina K, Augustine S, Jeengar MK, S K K. Zingiberaceae in Cardiovascular Health: A review of adipokine modulation and endothelial protection via adipocyte-endothelial crosstalk mechanism. Curr Nutr Rep 2025; 14:66. [PMID: 40366476 DOI: 10.1007/s13668-025-00656-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2025] [Indexed: 05/15/2025]
Abstract
PURPOSE OF THE REVIEW Although adipose tissue controls metabolism and protects vital organs, its importance to general health is being highlighted by the rise in type 2 diabetes and cardiovascular disease. Adipokines produced by adipose cells are essential regulators of metabolism, glucose homeostasis, and inflammatory response. It also protects vascular endothelial cells for its potential implications for cardiovascular protection. Understanding its intricate involvement in adipose tissue-endothelial communication is critical in developing targeted therapeutics to treat cardiovascular conditions linked with obesity and metabolic dysregulation. Spices from the Zingiberaceae family, such as cardamom, turmeric, and ginger, have anti-inflammatory and anti-oxidant properties that help reduce oxidative stress, vascular dysfunction, and adipocyte-endothelial crosstalk which are all linked to the etiology of CVD. Comprehensive molecular insights into how they modulate adipokine signalling, inflammatory pathways, and ROS-induced adipocyte-vascular interactions remain unexplored, demanding additional translational and clinical validation. With an emphasis on patients with obesity and metabolic dysregulation, the investigation aims to elucidate the mechanisms by which the spice as whole/bioactive constituents of the Zingiberaceae family may provide protection against CVD by integrating previous studies. RECENT FINDINGS Current research continues to support the use of spices from the Zingiberaceae family, such as ginger, turmeric, cardamom, and pepper, as potential therapeutic agents for addressing metabolic complications like obesity, type II diabetes, and CVDs. These natural remedies may modulate adipocyte-endothelial crosstalk and inflammation by modulating important signalling pathways such as AMPK, AKT, PPAR, and NF-κB.. CONCLUSION This review provides a complete summary of existing knowledge, opening the way for future research and prospective therapeutic applications of Zingiberaceae spices in cardiovascular health management.
Collapse
Affiliation(s)
- Disha Naidu
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - K P Althaf Umar
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - K Muhsina
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Sanu Augustine
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Manish Kumar Jeengar
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India.
| | - Kanthlal S K
- Department of Pharmacology, Sree Krishna College of Pharmacy and Research Centre, Parassala, Thiruvananthapuram, Kerala, 695502, India.
| |
Collapse
|
3
|
Saini S, Panchal SS. Role of Diabetes and its metabolic pathways in Epilepsy: An insight to various target approaches. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04245-1. [PMID: 40347278 DOI: 10.1007/s00210-025-04245-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/28/2025] [Indexed: 05/12/2025]
Abstract
The human brain acts as a crucial organ that requires a high glucose metabolic content. However, abnormal glucose levels act as a major factor for frequent epileptic foci. Thus, it has come to attention in the recent past that epilepsy is a metabolic problem in addition to a neurological condition. However, several studies have postulated a link between epilepsy and diabetes mellitus, but very few have emphasized the exact molecular mechanism behind it and its related specific targets. Hence, this article mainly outlines in-depth knowledge about the molecular mechanisms involved and its associated target approaches. Data from several publications, such as meta-analysis, systematic and narrative reviews, and research papers obtained from electronic databases, have been used for postulating a strong evidence in order to establish a comprehensive article addressing this problem in depth. The data discussed here have revealed how adiponectin levels and mitochondrial activity impact obesity, type 2 diabetes mellitus (T2DM), and epilepsy. We have also tried to give a brief idea about the possible theories that would also impact the severity of these two conditions, including adequate exercise and the impact of commonly used AEDs. Furthermore, one of the factors causing genetic predisposition to seizures due to glucose metabolism, such as GLUT-1 deficiency, has also been described briefly. It has to be mentioned that researchers and clinical practitioners might need to take these factors into account while discovering and evaluating a suitable novel therapeutic in the future.
Collapse
Affiliation(s)
- Sakshi Saini
- Department of Pharmacology, Institute of Pharmacy, Nirma University, S.G Highway, Ahmedabad, 382 481, Gujarat, India
| | - Shital S Panchal
- Department of Pharmacology, Institute of Pharmacy, Nirma University, S.G Highway, Ahmedabad, 382 481, Gujarat, India.
| |
Collapse
|
4
|
Solsona‐Vilarrasa E, Vousden KH. Obesity, white adipose tissue and cancer. FEBS J 2025; 292:2189-2207. [PMID: 39496581 PMCID: PMC12062788 DOI: 10.1111/febs.17312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024]
Abstract
White adipose tissue (WAT) is crucial for whole-body energy homeostasis and plays an important role in metabolic and hormonal regulation. While healthy WAT undergoes controlled expansion and contraction to meet the body's requirements, dysfunctional WAT in conditions like obesity is characterized by excessive tissue expansion, alterations in lipid homeostasis, inflammation, hypoxia, and fibrosis. Obesity is strongly associated with an increased risk of numerous cancers, with obesity-induced WAT dysfunction influencing cancer development through various mechanisms involving both systemic and local interactions between adipose tissue and tumors. Unhealthy obese WAT affects circulating levels of free fatty acids and factors like leptin, adiponectin, and insulin, altering systemic lipid metabolism and inducing inflammation that supports tumor growth. Similar mechanisms are observed locally in an adipose-rich tumor microenvironment (TME), where WAT cells can also trigger extracellular matrix remodeling, thereby enhancing the TME's ability to promote tumor growth. Moreover, tumors reciprocally interact with WAT, creating a bidirectional communication that further enhances tumorigenesis. This review focuses on the complex interplay between obesity, WAT dysfunction, and primary tumor growth, highlighting potential targets for therapeutic intervention.
Collapse
|
5
|
Mazurkiewicz M, Bodnar P, Blachut D, Chwalba T, Wagner W, Barczyk E, Romuk E, Jacheć W, Wojciechowska C. Adipokines and Adipose Tissue: The Role and Use of Sodium-Glucose Co-Transporter-2 (SGLT-2) Inhibitors in Patients with Diabetes or Heart Failure. Biomedicines 2025; 13:1098. [PMID: 40426925 PMCID: PMC12108695 DOI: 10.3390/biomedicines13051098] [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: 03/16/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Sodium-glucose co-transporter-2 (SGLT-2) inhibitors have become integral in treating both diabetes mellitus and heart failure, independent of left ventricular ejection fraction. Their pleiotropic effect influences multiple mechanisms, enhancing the function of various systems within the body. They exhibit nephroprotective and cardioprotective effects by improving cell metabolism, endothelial function, and slowing the fibrosis of the cardiac muscle, and they also have a beneficial impact on other organs. At the cellular level, they protect against the harmful effects of free radicals both by lowering glucose levels and by supporting the function of the antioxidant system. Moreover, SGLT-2 inhibitors can modify the metabolism of adipocytes by affecting the production of cytokines such as adiponectin-which increases insulin sensitivity, leading to weight loss and improved glycemic control.
Collapse
Affiliation(s)
- Michalina Mazurkiewicz
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Specialistic Hospital in Zabrze, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (D.B.); (T.C.); (W.J.); (C.W.)
| | - Patryk Bodnar
- Department of Anaesthesiology and Intensive Care, Clinical Hospital in Czeladź, Szpitalna 40 Street, 41-250 Czeladź, Poland;
| | - Dominika Blachut
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Specialistic Hospital in Zabrze, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (D.B.); (T.C.); (W.J.); (C.W.)
| | - Tomasz Chwalba
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Specialistic Hospital in Zabrze, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (D.B.); (T.C.); (W.J.); (C.W.)
| | - Wiktor Wagner
- Student Research Team at the Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (W.W.); (E.B.)
| | - Eliza Barczyk
- Student Research Team at the Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (W.W.); (E.B.)
| | - Ewa Romuk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 19 Street, 41-808 Zabrze, Poland;
| | - Wojciech Jacheć
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Specialistic Hospital in Zabrze, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (D.B.); (T.C.); (W.J.); (C.W.)
| | - Celina Wojciechowska
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Specialistic Hospital in Zabrze, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (D.B.); (T.C.); (W.J.); (C.W.)
| |
Collapse
|
6
|
McMahon JE, Graves JL, Tovar AP, Peloquin M, Greenwood K, Chen FL, Nelson M, McCandless EE, Halioua-Haubold CL, Juarez-Salinas D. Translational immune and metabolic markers of aging in dogs. Sci Rep 2025; 15:14460. [PMID: 40281285 PMCID: PMC12032292 DOI: 10.1038/s41598-025-99349-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Dogs serve as a promising aging model due to their genetic diversity, condensed lifespan, and shared living environment with humans. Alterations in the immune and metabolic parameters are hallmarks of aging in humans, but few studies have investigated these changes in dogs. We investigated the association of whole blood parameters with aging in a cross-sectional field study with a population of 451 companion dogs. Additionally, we measured total lymphocytes, total T-cells, CD4 T-cells, CD8 T-cells, B-cells, CBC, insulin and adiponectin in a cross-sectional study of 74 laboratory research beagles. In companion dogs, we report total lymphocytes and RBCs decrease significantly with age while platelets increase significantly. In lab beagles, total lymphocytes, T-cells, CD4 T-cells, CD8 T-cells, and B cells are significantly lower in Aged and Geriatric beagles. Furthermore, the CD4/CD8 ratio is significantly lower in Geriatric beagles. We also found that Geriatric beagles experience hyperinsulinemia, while plasma adiponectin is significantly lower in both Aged and Geriatric beagles. These results align with the age-related immune and metabolic alterations seen in humans and provide additional evidence that dogs serve as a relevant translational model of aging.
Collapse
|
7
|
Zhi Y, Xie S, Wei B. Electrochemical biosensors for enhanced detection of diabetes mellitus. Clin Chim Acta 2025; 571:120221. [PMID: 40024276 DOI: 10.1016/j.cca.2025.120221] [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: 01/21/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
The worldwide incidence of diabetes mellitus (DM), as a long-term metabolic condition, continues to rise, creating an urgent need for accurate and efficient diagnostic methods to identify and treat the disease early. Among various analytical technologies, electrochemical biosensors stand out for their exceptional attributes, including precise detection, selective response, quick results, and affordable implementation. The current review study examines the latest developments in electrochemical biosensor technology designed specifically for diabetes detection, emphasizing novel approaches in blood sugar monitoring and tracking key diabetes indicators, including HbA1c, insulin levels, and ketones. The discussion encompasses cutting-edge developments such as sensors incorporating nanomaterials, non-enzymatic detection systems, and portable monitoring devices, emphasizing how these innovations improve both technical capabilities and patient experience. This review also demonstrates how next-generation electrochemical biosensors could fundamentally change diabetes care and monitoring, leading to more widely available and accurate disease tracking methods.
Collapse
Affiliation(s)
- Yong Zhi
- Xinjiang 474 Hospital, Urumqi, Xinjiang 830011, China; College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang 830017, China.
| | - Shanshan Xie
- Xinjiang Key Laboratory of Mental Development and Learning Science, Xinjiang Normal University, Urumqi, Xinjiang 830000, China
| | - Bowen Wei
- Independent Research, Jilin 132100, China.
| |
Collapse
|
8
|
Lupu A, Mihai CM, Dragan F, Tarnita I, Alecsa M, Chisnoiu T, Morariu ID, Cuciureanu M, Nedelcu AH, Salaru DL, Anton E, Danielescu C, Fotea S, Stoleriu G, Beser OF, Lupu VV. Antioxidant Supplementation in Childhood Obesity: A Path to Improved Metabolic Health? Antioxidants (Basel) 2025; 14:466. [PMID: 40298814 PMCID: PMC12024302 DOI: 10.3390/antiox14040466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Childhood obesity is linked to heightened oxidative stress, a key driver of endothelial dysfunction, inflammation, and metabolic complications. Antioxidants, including Vitamins C and E, are vital in neutralizing free radicals and mitigating oxidative damage. This non-systematic review examines the potential advantages of antioxidant supplementation in pediatric obesity, focusing on its effects on vascular health, insulin sensitivity, and inflammatory processes. Emerging data suggest that antioxidants may improve endothelial function, reduce blood pressure, and enhance metabolic homeostasis in obese children. However, the long-term efficacy and safety of antioxidant supplementation remain uncertain, necessitating further rigorous randomized controlled trials. A deeper understanding of antioxidants' role in pediatric obesity could unlock novel therapeutic approaches for managing obesity-related complications and improving children's overall health outcomes.
Collapse
Affiliation(s)
- Ancuta Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (M.A.); (V.V.L.)
| | - Cristina Maria Mihai
- Pediatrics, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania; (C.M.M.); (T.C.)
| | - Felicia Dragan
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Irina Tarnita
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (M.A.); (V.V.L.)
| | - Mirabela Alecsa
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (M.A.); (V.V.L.)
| | - Tatiana Chisnoiu
- Pediatrics, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania; (C.M.M.); (T.C.)
| | - Ionela Daniela Morariu
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Magdalena Cuciureanu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (A.H.N.); (D.L.S.); (E.A.); (C.D.)
| | - Alin Horatiu Nedelcu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (A.H.N.); (D.L.S.); (E.A.); (C.D.)
| | - Delia Lidia Salaru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (A.H.N.); (D.L.S.); (E.A.); (C.D.)
| | - Emil Anton
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (A.H.N.); (D.L.S.); (E.A.); (C.D.)
| | - Ciprian Danielescu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (A.H.N.); (D.L.S.); (E.A.); (C.D.)
| | - Silvia Fotea
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (S.F.); (G.S.)
| | - Gabriela Stoleriu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (S.F.); (G.S.)
| | - Omer Faruk Beser
- Department of Pediatric Gastroenterology, Hepatology & Nutrition, Cerrahpasa Medical Faculty, Istanbul University Cerrahpasa, 34776 Istanbul, Turkey;
| | - Vasile Valeriu Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (M.A.); (V.V.L.)
| |
Collapse
|
9
|
Siriphorn SV, Thorsuwan S, Thongam J, Ruangklai S, Hussarin P, Rungruang T, Srisuma S. Alterations in Adiponectin Expression in Models of Cigarette Smoke Extract-Induced Mouse Pulmonary Emphysema and Alveolar Epithelial Cell Injury. COPD 2025; 22:2477235. [PMID: 40079477 DOI: 10.1080/15412555.2025.2477235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
PURPOSE Cigarette smoke activates lung inflammation and destruction and the development of COPD. Among various factors influenced by lung inflammation, adiponectin produced by lung epithelial cells is thought to play a significant role in regulating inflammation and maintaining tissue integrity. This study aims to examine adiponectin expression in a mouse model of cigarette smoke extract (CSE)-induced emphysema and explore the effects of adiponectin on cell survival and cytokine gene expression in CSE-induced lung epithelial cell damage. METHODS CSE was prepared by passing cigarette smoke through a glass tube containing solvent. PBS or CSE was intraperitoneally administered to C57BL/6 mice. Inflammatory cells, cytokines, adiponectin expression in lung, bronchoalveolar lavage fluid (BALF) and adipose tissue were assessed. CSE and adiponectin were administered to A549 cells to determine cell viability and cytokine gene expression. RESULTS Intraperitoneal CSE injection significantly increased the mean alveolar linear intercept by 23.11%. CSE significantly increased total cells, macrophages, neutrophils, eosinophils, TNFα, IL-1β levels in BALF. CSE enhanced lung adiponectin protein expression. Treatment of A549 cells with CSE reduced cell survival and adiponectin gene expression. Furthermore, adiponectin treatment enhanced MCP-1 and IL-8 gene expression in A549 cells post-CSE exposure. CONCLUSION Intraperitoneal CSE treatment induced lung inflammation, airspace enlargement, and increased adiponectin expression in mice. CSE-exposed A549 cells showed reduced cell viability, upregulated proinflammatory genes, downregulated adiponectin genes. Adiponectin treatment further intensified these genes expressions, aligning with in vivo findings. Elevated adiponectin expression in alveolar epithelial cells suggests its potential role in the development of COPD by enhancing lung inflammation.
Collapse
Affiliation(s)
- Siriporn Vongsaiyat Siriphorn
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Physical Therapy and Sports Medicine, Rangsit University, Pathumtani, Thailand
| | - Supitsara Thorsuwan
- Princess Agrarajakumari College of Nursing, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Julalux Thongam
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sukpattaraporn Ruangklai
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Poungpetch Hussarin
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Thanaporn Rungruang
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sorachai Srisuma
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
10
|
Cillo U, Lonati C, Bertacco A, Magnini L, Battistin M, Borsetto L, Dazzi F, Al-Adra D, Gringeri E, Bacci ML, Schlegel A, Dondossola D. A proof-of-concept study in small and large animal models for coupling liver normothermic machine perfusion with mesenchymal stromal cell bioreactors. Nat Commun 2025; 16:283. [PMID: 39746966 PMCID: PMC11697227 DOI: 10.1038/s41467-024-55217-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
To fully harness mesenchymal-stromal-cells (MSCs)' benefits during Normothermic Machine Perfusion (NMP), we developed an advanced NMP platform coupled with a MSC-bioreactor and investigated its bio-molecular effects and clinical feasibility using rat and porcine models. The study involved three work packages: 1) Development (n = 5): MSC-bioreactors were subjected to 4 h-liverless perfusion; 2) Rat model (n = 10): livers were perfused for 4 h on the MSC-bioreactor-circuit or with the standard platform; 3) Porcine model (n = 6): livers were perfused using a clinical device integrated with a MSC-bioreactor or in its standard setup. MSCs showed intact stem-core properties after liverless-NMP. Liver NMP induced specific, liver-tailored, changes in MSCs' secretome. Rat livers exposed to bioreactor-based perfusion produced more bile, released less damage and pro-inflammatory biomarkers, and showed improved mithocondrial function than those subjected to standard NMP. MSC-bioreactor integration into a clinical device resulted in no machine failure and perfusion-related injury. This proof-of-concept study presents a novel MSC-based liver NMP platform that could reduce the deleterious effects of ischemia/reperfusion before transplantation.
Collapse
Affiliation(s)
- Umberto Cillo
- Hepato-Biliary-Pancreatic Surgery and Liver Transplant Unit, General Surgery 2, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Caterina Lonati
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20100, Milan, Italy.
| | - Alessandra Bertacco
- Hepato-Biliary-Pancreatic Surgery and Liver Transplant Unit, General Surgery 2, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Lucrezia Magnini
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20100, Milan, Italy
| | - Michele Battistin
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20100, Milan, Italy
| | - Lara Borsetto
- Hepato-Biliary-Pancreatic Surgery and Liver Transplant Unit, General Surgery 2, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Francesco Dazzi
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
| | - David Al-Adra
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Enrico Gringeri
- Hepato-Biliary-Pancreatic Surgery and Liver Transplant Unit, General Surgery 2, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Maria Laura Bacci
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Andrea Schlegel
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20100, Milan, Italy
- Transplantation Center, Digestive Disease and Surgery Institute, Department of Immunity and Inflammation, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Daniele Dondossola
- General and Liver Transplant Surgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20100, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza 35, 20100, Milan, Italy
| |
Collapse
|
11
|
Duan F, Wu J, Chang J, Peng H, Liu Z, Liu P, Han X, Sun T, Shang D, Yang Y, Li Z, Li P, Liu Y, Zhu Y, Lv Y, Guo X, Zhao Y, An Y. Deciphering endocrine function of adipose tissue and its significant influences in obesity-related diseases caused by its dysfunction. Differentiation 2025; 141:100832. [PMID: 39709882 DOI: 10.1016/j.diff.2024.100832] [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: 10/29/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Current research has found that adipose tissue is not only involved in energy metabolism, but also a highly active endocrine organ that secretes various adipokines, including adiponectin, leptin, resistin and apelin, which are involved in the regulation of physiology and pathology of tissues and organs throughout the body. With the yearly increasing incidence, obesity has become a risk factor for a variety of pathological changes, including inflammation and metabolic syndrome in various system (endocrine, circulatory, locomotor and central nervous system). Thus these symptoms lead to multi-organ dysfunctions, including the heart, liver, kidneys, brain and joints. An in-depth summary of the roles of adipokines in the regulation of other tissues and organs can help to provide more effective therapeutic strategies for obesity-related diseases and explore potential therapeutic targets. Therefore, this review has retrospected the endocrine function of adipose tissue under obesity and the role of dysregulated adipokine secretion in related diseases and the underlying mechanisms, in order to provide a theoretical basis for targeting adipokine-mediated systemic dysregulation.
Collapse
Affiliation(s)
- Feiyi Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiaoyan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiayi Chang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Haoyuan Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Zitao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengfei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Dandan Shang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yutian Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengkun Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yixuan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yonghao Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yunzhi Lv
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Xiumei Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Ying Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
12
|
Chen TY, Marín-López A, Raduwan H, Fikrig E. Aedes aegypti adiponectin receptor-like protein signaling facilitates Zika virus infection. mBio 2024; 15:e0243324. [PMID: 39373507 PMCID: PMC11559040 DOI: 10.1128/mbio.02433-24] [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: 08/07/2024] [Accepted: 09/18/2024] [Indexed: 10/08/2024] Open
Abstract
The Aedes aegypti mosquito plays a critical role in the transmission of viral diseases, including Zika virus, which poses significant public health challenges. Understanding the complex interactions between mosquitoes and viruses is paramount for the development of effective control strategies. In this study, we demonstrate that silencing the A. aegypti adiponectin receptor-like protein (AaARLP) results in a reduction of Zika virus infection. Transcriptomic analysis identified alterations in several trypsin genes and further revealed that AaARLP-knockdown mosquitoes had diminished trypsin activity. Moreover, silencing of selected trypsins resulted in a similar delay in Zika virus infection in mosquitoes, further highlighting the connection between the AaARLP and trypsin. Overall, our findings demonstrate that AaARLP signaling is important for Zika virus infection of A. aegypti. IMPORTANCE Arboviruses pose a significant threat to public health, with mosquitoes, especially Aedes aegypti, being a major vector for their transmission. Gaining insight into the complex interaction between mosquitoes and viruses is essential to build successful control strategies. In this study, we identified a novel pathway connecting the A. aegypti adiponectin receptor-like protein and its association with trypsin, key enzymes involved in blood digestion. Furthermore, we demonstrated the significance of signaling via the adiponectin receptor-like protein in virus infection within the mosquito. Together, our discoveries illuminate mosquito metabolic pathways essential in viral infection.
Collapse
Affiliation(s)
- Tse-Yu Chen
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Alejandro Marín-López
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Hamidah Raduwan
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
13
|
Liu YJ, Lee CW, Liao YC, Huang JJT, Kuo HC, Jih KY, Lee YC, Chern Y. The role of adiponectin-AMPK axis in TDP-43 mislocalization and disease severity in ALS. Neurobiol Dis 2024; 202:106715. [PMID: 39490684 DOI: 10.1016/j.nbd.2024.106715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
Hypermetabolism is a prominent characteristic of ALS patients. Aberrant activation of AMPK, an energy sensor regulated by adiponectin, is known to cause TDP-43 mislocalization, an early event in ALS pathogenesis. This study aims to evaluate the association between key energy mediators and clinical severity in ALS patients. We found that plasma adiponectin levels were significantly higher in ALS patients with ALSFRS-R scores below 38 compared to controls (p = 0.047). Additionally, adiponectin concentration was inversely correlated with ALSFRS-R scores (p = 0.021). Immunofluorescence staining of PBMCs revealed negative associations between AMPK activation, TDP-43 mislocalization, and ALSFRS-R scores. We then examined the hypothesis that adiponectin may activate the AMPK-TDP-43 axis in motor neurons. Our results demonstrated that adiponectin treatment of NSC34 cells and HiPSC-MNs induced AMPK activation and TDP-43 mislocalization in an adiponectin receptor-dependent manner. Collectively, these findings suggest that elevated plasma adiponectin may enhance AMPK activation, leading to TDP-43 mislocalization in both PBMCs and motor neurons of ALS patients. This highlights the potential involvement of the adiponectin-AMPK-TDP-43 axis in the dysregulated energy balance observed in ALS.
Collapse
Affiliation(s)
- Yu-Ju Liu
- Division of Neuroscience, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chia-Wei Lee
- Division of Neuroscience, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Chu Liao
- Department of Neurology, Taipei Veterans General Hospital, and Department of Neurology and Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | | | - Hung-Chih Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Kang-Yang Jih
- Department of Neurology, Taipei Veterans General Hospital, and Department of Neurology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Chung Lee
- Department of Neurology, Taipei Veterans General Hospital, and Department of Neurology and Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Yijuang Chern
- Division of Neuroscience, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
14
|
Torres-Torres J, Monroy-Muñoz IE, Perez-Duran J, Solis-Paredes JM, Camacho-Martinez ZA, Baca D, Espino-Y-Sosa S, Martinez-Portilla R, Rojas-Zepeda L, Borboa-Olivares H, Reyes-Muñoz E. Cellular and Molecular Pathophysiology of Gestational Diabetes. Int J Mol Sci 2024; 25:11641. [PMID: 39519193 PMCID: PMC11546748 DOI: 10.3390/ijms252111641] [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: 09/13/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Gestational diabetes (GD) is a metabolic disorder characterized by glucose intolerance during pregnancy, significantly impacting maternal and fetal health. Its global prevalence is approximately 14%, with risk factors including obesity, family history of diabetes, advanced maternal age, and ethnicity, which are linked to cellular and molecular disruptions in glucose regulation and insulin resistance. GD is associated with short- and long-term complications for both the mother and the newborn. For mothers, GD increases the risk of developing type 2 diabetes, cardiovascular diseases, and metabolic syndrome. In the offspring, exposure to GD in utero predisposes them to obesity, glucose intolerance, and metabolic disorders later in life. This review aims to elucidate the complex cellular and molecular mechanisms underlying GD to inform the development of effective therapeutic strategies. A systematic review was conducted using medical subject headings (MeSH) terms related to GD's cellular and molecular pathophysiology. Inclusion criteria encompassed original studies, systematic reviews, and meta-analyses focusing on GD's impact on maternal and fetal health, adhering to PRISMA guidelines. Data extraction captured study characteristics, maternal and fetal outcomes, key findings, and conclusions. GD disrupts insulin signaling pathways, leading to impaired glucose uptake and insulin resistance. Mitochondrial dysfunction reduces ATP production and increases reactive oxygen species, exacerbating oxidative stress. Hormonal influences, chronic inflammation, and dysregulation of the mammalian target of rapamycin (mTOR) pathway further impair insulin signaling. Gut microbiota alterations, gene expression, and epigenetic modifications play significant roles in GD. Ferroptosis and placental dysfunction primarily contribute to intrauterine growth restriction. Conversely, fetal macrosomia arises from maternal hyperglycemia and subsequent fetal hyperinsulinemia, resulting in excessive fetal growth. The chronic inflammatory state and oxidative stress associated with GD exacerbate these complications, creating a hostile intrauterine environment. GD's complex pathophysiology involves multiple disruptions in insulin signaling, mitochondrial function, inflammation, and oxidative stress. Effective management requires early detection, preventive strategies, and international collaboration to standardize care and improve outcomes for mothers and babies.
Collapse
Affiliation(s)
- Johnatan Torres-Torres
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Irma Eloisa Monroy-Muñoz
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Javier Perez-Duran
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Juan Mario Solis-Paredes
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | | | - Deyanira Baca
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Salvador Espino-Y-Sosa
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
- Centro de Investigacion en Ciencias de la Salud, Universidad Anahuac Mexico, Campus Norte, Huixquilucan 52786, Mexico
| | - Raigam Martinez-Portilla
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Lourdes Rojas-Zepeda
- Maternal-Fetal Department, Instituto Materno Infantil del Estado de Mexico, Toluca 50170, Mexico
| | - Hector Borboa-Olivares
- Community Interventions Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Enrique Reyes-Muñoz
- Research Division, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| |
Collapse
|
15
|
Hu F, Yu Y, Xu H. How does exosome cause diabetes? Hormones (Athens) 2024; 23:385-393. [PMID: 38233729 DOI: 10.1007/s42000-024-00525-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 01/08/2024] [Indexed: 01/19/2024]
Abstract
Exosomes are extracellular vesicles that are widely distributed in multiple cell types and circulating body fluids. They have a specific effect on the target cells by releasing different vesicle contents. They have recently been recognized as important means of intercellular communication, being involved, for example, in the development of diabetes by increasing β-cell apoptosis, activating autoimmunity, and regulating cytokines to affect islet β-cell function and insulin sensitivity. An in-depth study of the role of exosome in the pathogenesis of diabetes may therefore provide a novel means of diagnosing and treating diabetes. In this review, we detail how exosome is involved in the development of diabetes.
Collapse
Affiliation(s)
- Fei Hu
- Cixi Biomedical Research Institute, Wenzhou Medical University, Cixi, Ningbo, China
| | - Yicong Yu
- Zhejiang Center of Animal Disease Control, Hangzhou, China
| | - Hongming Xu
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital, Wenzhou Medical University, No. 999, South Second Ring Road, Hushan Street, Cixi, Ningbo, 315300, China.
| |
Collapse
|
16
|
Shen X, Ma C, Yang Y, Liu X, Wang B, Wang Y, Zhang G, Bian X, Zhang N. The Role and Mechanism of Probiotics Supplementation in Blood Glucose Regulation: A Review. Foods 2024; 13:2719. [PMID: 39272484 PMCID: PMC11394447 DOI: 10.3390/foods13172719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
With economic growth and improved living standards, the incidence of metabolic diseases such as diabetes mellitus caused by over-nutrition has risen sharply worldwide. Elevated blood glucose and complications in patients seriously affect the quality of life and increase the economic burden. There are limitations and side effects of current hypoglycemic drugs, while probiotics, which are safe, economical, and effective, have good application prospects in disease prevention and remodeling of intestinal microecological health and are gradually becoming a research hotspot for diabetes prevention and treatment, capable of lowering blood glucose and alleviating complications, among other things. Probiotic supplementation is a microbiologically based approach to the treatment of type 2 diabetes mellitus (T2DM), which can achieve anti-diabetic efficacy through the regulation of different tissues and metabolic pathways. In this study, we summarize recent findings that probiotic intake can achieve blood glucose regulation by modulating intestinal flora, decreasing chronic low-grade inflammation, modulating glucagon-like peptide-1 (GLP-1), decreasing oxidative stress, ameliorating insulin resistance, and increasing short-chain fatty acids (SCFAs) content. Moreover, the mechanism, application, development prospect, and challenges of probiotics regulating blood glucose were discussed to provide theoretical references and a guiding basis for the development of probiotic preparations and related functional foods regulating blood glucose.
Collapse
Affiliation(s)
- Xinyu Shen
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Chunmin Ma
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Yang Yang
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Xiaofei Liu
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Bing Wang
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Yan Wang
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Guang Zhang
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Xin Bian
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| | - Na Zhang
- College of Food Engineering, Harbin University of Commerce, Harbin 150028, China
| |
Collapse
|
17
|
Abdollahi A, Szramowski M, Tomoo K, Henderson GC. Metabolic responses to albumin deficiency differ distinctly between partial and full ablation of albumin expression in mice. Lipids Health Dis 2024; 23:242. [PMID: 39123208 PMCID: PMC11312229 DOI: 10.1186/s12944-024-02229-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
It had been observed that homozygous albumin knockout mice (Alb-/-) exhibit low plasma free fatty acid (FFA) concentration and improved blood glucose regulation. However, it was not yet known to what extent heterozygous albumin knockout (Alb+/-) mice would display a similar phenotype. Alb-/-, Alb+/-, and wild-type (WT) female mice were studied on a low-fat diet (LFD) or high-fat diet (HFD). On both diets, decreased plasma FFA concentration, and improved glucose tolerance test were observed in Alb-/-, but not in Alb+/-, compared to WT. Plasma adiponectin concentration showed greater elevation in Alb-/- than Alb+/-. Consistent with that, adiponectin gene expression was significantly higher in Alb-/- mice than in Alb+/- and WT mice. A dose-dependent response was observed for hepatic Acadl gene expression showing higher Acadl gene expression in Alb-/- mice than in Alb+/- and WT mice. In conclusion, although female Alb+/- mice exhibited some slight differences from WT mice (e.g., increased plasma adiponectin and hepatic Acadl gene expression), Alb+/- mice did not exhibit improved glucoregulation in comparison to WT mice, indicating that a minor suppression of albumin expression is not sufficient to improve glucoregulation. Furthermore, it is now clear that although the response of female mice to HFD might be unique from how males generally respond, still the complete albumin deficiency in Alb-/- mice and the associated FFA reduction is capable of improving glucoregulation in females on this diet. The present results have implications for the role of albumin and FFA in the regulation of metabolism.
Collapse
Affiliation(s)
- Afsoun Abdollahi
- Department of Nutrition Science, Purdue University, STON 208, 700 Mitch Daniels Blvd, West Lafayette, IN, 47907, USA
| | - Mirandia Szramowski
- Department of Nutrition Science, Purdue University, STON 208, 700 Mitch Daniels Blvd, West Lafayette, IN, 47907, USA
| | - Keigo Tomoo
- Department of Nutrition Science, Purdue University, STON 208, 700 Mitch Daniels Blvd, West Lafayette, IN, 47907, USA
| | - Gregory C Henderson
- Department of Nutrition Science, Purdue University, STON 208, 700 Mitch Daniels Blvd, West Lafayette, IN, 47907, USA.
| |
Collapse
|
18
|
Barbosa S, Pedrosa MB, Ferreira R, Moreira-Gonçalves D, Santos LL. The impact of chemotherapy on adipose tissue remodeling: The molecular players involved in this tissue wasting. Biochimie 2024; 223:1-12. [PMID: 38537739 DOI: 10.1016/j.biochi.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/05/2024]
Abstract
The depletion of visceral and subcutaneous adipose tissue (AT) during chemotherapy significantly correlates with diminished overall survival and progression-free survival. Despite its clinical significance, the intricate molecular mechanisms governing this AT loss and its chemotherapy-triggered initiation remain poorly understood. Notably, the evaluation of AT remodeling in most clinical trials has predominantly relied on computerized tomography scans or bioimpedance, with molecular studies often conducted using animal or in vitro models. To address this knowledge gap, a comprehensive narrative review was conducted. The findings underscore that chemotherapy serves as a key factor in inducing AT loss, exacerbating cachexia, a paraneoplastic syndrome that significantly compromises patient quality of life and survival. The mechanism driving AT loss appears intricately linked to alterations in AT metabolic remodeling, marked by heightened lipolysis and fatty acid oxidation, coupled with diminished lipogenesis. However, adipocyte stem cells' lost ability to divide due to chemotherapy also appears to be at the root of the loss of AT. Notably, chemotherapy seems to deactivate the mitochondrial antioxidant system by reducing key regulatory enzymes responsible for neutralizing reactive oxygen species (ROS), thereby impeding lipogenesis. Despite FDG-PET evidence of AT browning, no molecular evidence of thermogenesis was reported. Prospective investigations unraveling the molecular mechanisms modulated in AT by chemotherapy, along with therapeutic strategies aimed at preventing AT loss, promise to refine treatment paradigms and enhance patient outcomes.
Collapse
Affiliation(s)
- Samuel Barbosa
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450, Porto, Portugal; Experimental Pathology and Therapeutics Group, Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (P.CCC), 4200-072, Porto, Portugal.
| | - Mafalda Barbosa Pedrosa
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450, Porto, Portugal; Experimental Pathology and Therapeutics Group, Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (P.CCC), 4200-072, Porto, Portugal; Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (LAQV-REQUIMTE), Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rita Ferreira
- Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (LAQV-REQUIMTE), Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Daniel Moreira-Gonçalves
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450, Porto, Portugal; Laboratory for Integrative and Translational Research in Population Health (ITR), 4050-600, Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (P.CCC), 4200-072, Porto, Portugal
| |
Collapse
|
19
|
Aslam M, Li L, Nürnberger S, Niemann B, Rohrbach S. CTRP13-Mediated Effects on Endothelial Cell Function and Their Potential Role in Obesity. Cells 2024; 13:1291. [PMID: 39120321 PMCID: PMC11311976 DOI: 10.3390/cells13151291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Obesity, a major component of cardiometabolic syndrome, contributes to the imbalance between pro- and anti-atherosclerotic factors via dysregulation of adipocytokine secretion. Among these adipocytokines, the C1q/TNF-related proteins (CTRPs) play a role in the modulation of atherosclerosis development and progression. Here, we investigated the vascular effects of CTRP13. RESULTS CTRP13 is not only expressed in adipose tissue but also in vessels/endothelial cells (ECs) of mice, rats, and humans. Obese individuals (mice, rats, and humans) showed higher vascular CTRP13 expression. Human Umbilical Vein Endothelial Cells (HUVECs), cultured in the presence of serum from obese mice, mimicked this obesity-associated effect on CTRP13 protein expression. Similarly, high glucose conditions and TNF-alpha, but not insulin, resulted in a strong increase in CTRP13 in these cells. Recombinant CTRP13 induced a reduction in EC proliferation via AMPK. In addition, CTRP13 reduced cell cycle progression and increased p53 phosphorylation and p21 protein expression, but reduced Rb phosphorylation, with the effects largely depending on alpha-2 AMPK as suggested by adenoviral overexpression of dominant-negative (DN) or wild-type (WT) alpha 1/alpha 2 AMPK. CONCLUSION The present study demonstrates that CTRP13 expression is induced in ECs under diabetic conditions and that CTRP13 possesses significant vaso-modulatory properties which may have an impact on vascular disease progression in patients.
Collapse
Affiliation(s)
- Muhammad Aslam
- Experimental Cardiology, Department of Internal Medicine I, Justus Liebig University Giessen, 35390 Giessen, Germany;
| | - Ling Li
- Institute of Physiology, Justus Liebig University Giessen, 35390 Giessen, Germany; (L.L.); (S.N.)
| | - Sina Nürnberger
- Institute of Physiology, Justus Liebig University Giessen, 35390 Giessen, Germany; (L.L.); (S.N.)
| | - Bernd Niemann
- Department of Cardiovascular Surgery Giessen, University-Hospital Giessen and Marburg, Justus Liebig University Giessen, 35390 Giessen, Germany;
| | - Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, 35390 Giessen, Germany; (L.L.); (S.N.)
| |
Collapse
|
20
|
Baldelli S, Aiello G, Mansilla Di Martino E, Campaci D, Muthanna FMS, Lombardo M. The Role of Adipose Tissue and Nutrition in the Regulation of Adiponectin. Nutrients 2024; 16:2436. [PMID: 39125318 PMCID: PMC11313710 DOI: 10.3390/nu16152436] [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: 06/14/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Adipose tissue (AT), composed mainly of adipocytes, plays a critical role in lipid control, metabolism, and energy storage. Once considered metabolically inert, AT is now recognized as a dynamic endocrine organ that regulates food intake, energy homeostasis, insulin sensitivity, thermoregulation, and immune responses. This review examines the multifaceted role of adiponectin, a predominant adipokine released by AT, in glucose and fatty acid metabolism. We explore the regulatory mechanisms of adiponectin, its physiological effects and its potential as a therapeutic target for metabolic diseases such as type 2 diabetes, cardiovascular disease and fatty liver disease. Furthermore, we analyze the impact of various dietary patterns, specific nutrients, and physical activities on adiponectin levels, highlighting strategies to improve metabolic health. Our comprehensive review provides insights into the critical functions of adiponectin and its importance in maintaining systemic metabolic homeostasis.
Collapse
Affiliation(s)
- Sara Baldelli
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy (E.M.D.M.)
- IRCCS San Raffaele Roma, 00166 Rome, Italy
| | - Gilda Aiello
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy (E.M.D.M.)
| | - Eliana Mansilla Di Martino
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy (E.M.D.M.)
| | - Diego Campaci
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy (E.M.D.M.)
| | - Fares M. S. Muthanna
- Pharmacy Department, Faculty of Medicine and Health Sciences, University of Science and Technology-Aden, Alshaab Street, Enmaa City 22003, Yemen
| | - Mauro Lombardo
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy (E.M.D.M.)
| |
Collapse
|
21
|
Pan S, Wang Y, Zhang Y, Ma X, Peng J, Li F, Qian W, Zong J. The Relationship Between the Serum NLRP3 and Adiponectin Levels and Coronary Lesions in Patients with Unstable Angina with Type 2 Diabetes. Clin Interv Aging 2024; 19:1301-1308. [PMID: 39050520 PMCID: PMC11268749 DOI: 10.2147/cia.s467291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
Objective To investigate the Levels of Nucleotide-binding, leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) and Adiponectin (APN) and their relationship with the severity of coronary artery disease in patients with Unstable Angina (UA) and Type 2 Diabetes (T2D). Methods Two hundred and thirty-one patients with UA were diagnosed by CAG in the Department of Cardiology of the Affiliated Hospital of Xuzhou Medical University from July 2022 to May 2023 were included, and 74 healthy subjects were included as the control group. The levels of NLRP3 and APN in each group were detected by ELISA and the Gensini score in each patient according to the results of CAG. The correlations between NLRP3, APN, and Gensini score were analyzed. According to whether complicated with T2D or not, we further analyze the effect of NLRP3 and APN levels of patients with UA and T2D on the severity of coronary artery stenosis. Results The levels of NLRP3 in UA with T2D group were the highest, followed by simple UA group, and the lowest in the control group, and the level of APN was the opposite. Spearman Correlation analysis showed that the level of NLRP3 was positively correlated with Gensini score (ρ1=0.688, P<0.05) and the level of APN was negatively associated with Gensini score (ρ2= -0.515, P<0.05). There was a negative correlation between NLRP3 and the level of APN (ρ3= -0.366, P<0.05). High NLRP3 and low APN levels are the risk factors for atherosclerosis. Conclusion The NLRP3 and APN were abnormally expressed in patients with UA complicated with T2D. With the aggravation of atherosclerosis, the level of NLRP3 increased and the level of APN decreased.
Collapse
Affiliation(s)
- Siyu Pan
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Yixiao Wang
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Yuchen Zhang
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Xiaoyu Ma
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Jingfeng Peng
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Fangfang Li
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Wenhao Qian
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Jing Zong
- Institute of Cardiovascular Diseases, Xuzhou Medical University, Xuzhou, People’s Republic of China
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| |
Collapse
|
22
|
Fu L, Du J, Furkert D, Shipton ML, Liu X, Aguirre T, Chin AC, Riley AM, Potter BVL, Fiedler D, Zhang X, Zhu Y, Fu C. Depleting inositol pyrophosphate 5-InsP7 protected the heart against ischaemia-reperfusion injury by elevating plasma adiponectin. Cardiovasc Res 2024; 120:954-970. [PMID: 38252884 PMCID: PMC11218692 DOI: 10.1093/cvr/cvae017] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 01/24/2024] Open
Abstract
AIMS Adiponectin is an adipocyte-derived circulating protein that exerts cardiovascular and metabolic protection. Due to the futile degradation of endogenous adiponectin and the challenges of exogenous administration, regulatory mechanisms of adiponectin biosynthesis are of significant pharmacological interest. METHODS AND RESULTS Here, we report that 5-diphosphoinositol 1,2,3,4,6-pentakisphosphate (5-InsP7) generated by inositol hexakisphosphate kinase 1 (IP6K1) governed circulating adiponectin levels via thiol-mediated protein quality control in the secretory pathway. IP6K1 bound to adiponectin and DsbA-L and generated 5-InsP7 to stabilize adiponectin/ERp44 and DsbA-L/Ero1-Lα interactions, driving adiponectin intracellular degradation. Depleting 5-InsP7 by either IP6K1 deletion or pharmacological inhibition blocked intracellular adiponectin degradation. Whole-body and adipocyte-specific deletion of IP6K1 boosted plasma adiponectin levels, especially its high molecular weight forms, and activated AMPK-mediated protection against myocardial ischaemia-reperfusion injury. Pharmacological inhibition of 5-InsP7 biosynthesis in wild-type but not adiponectin knockout mice attenuated myocardial ischaemia-reperfusion injury. CONCLUSION Our findings revealed that 5-InsP7 is a physiological regulator of adiponectin biosynthesis that is amenable to pharmacological intervention for cardioprotection.
Collapse
Affiliation(s)
- Lin Fu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Jimin Du
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - David Furkert
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Megan L Shipton
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Xiaoqi Liu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Tim Aguirre
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Alfred C Chin
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Andrew M Riley
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Barry V L Potter
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Dorothea Fiedler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Chenglai Fu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Yangpu District, Shanghai 200092, China
| |
Collapse
|
23
|
McDowell JA, Kosmacek EA, Baine MJ, Adebisi O, Zheng C, Bierman MM, Myers MS, Chatterjee A, Liermann-Wooldrik KT, Lim A, Dickinson KA, Oberley-Deegan RE. Exogenous APN protects normal tissues from radiation-induced oxidative damage and fibrosis in mice and prostate cancer patients with higher levels of APN have less radiation-induced toxicities. Redox Biol 2024; 73:103219. [PMID: 38851001 PMCID: PMC11201354 DOI: 10.1016/j.redox.2024.103219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024] Open
Abstract
Radiation causes damage to normal tissues that leads to increased oxidative stress, inflammation, and fibrosis, highlighting the need for the selective radioprotection of healthy tissues without hindering radiotherapy effectiveness in cancer. This study shows that adiponectin, an adipokine secreted by adipocytes, protects normal tissues from radiation damage invitro and invivo. Specifically, adiponectin (APN) reduces chronic oxidative stress and fibrosis in irradiated mice. Importantly, APN also conferred no protection from radiation to prostate cancer cells. Adipose tissue is the primary source of circulating endogenous adiponectin. However, this study shows that adipose tissue is sensitive to radiation exposure exhibiting morphological changes and persistent oxidative damage. In addition, radiation results in a significant and chronic reduction in blood APN levels from adipose tissue in mice and human prostate cancer patients exposed to pelvic irradiation. APN levels negatively correlated with bowel toxicity and overall toxicities associated with radiotherapy in prostate cancer patients. Thus, protecting, or modulating APN signaling may improve outcomes for prostate cancer patients undergoing radiotherapy.
Collapse
Affiliation(s)
- Joshua A McDowell
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Elizabeth A Kosmacek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Michael J Baine
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Oluwaseun Adebisi
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Cheng Zheng
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Madison M Bierman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Molly S Myers
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Arpita Chatterjee
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kia T Liermann-Wooldrik
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Andrew Lim
- College of Nursing, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kristin A Dickinson
- College of Nursing, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
24
|
Czerewaty M, Łączna M, Kiełbowski K, Bakinowska E, Dec P, Modrzejewski A, Kotrych D, Burszewski P, Safranow K, Pawlik A. The effect of plasma cytokines on the expression of adiponectin and its receptors in the synovial membrane of joints and the infrapatellar fat pad in patients with rheumatoid arthritis and osteoarthritis. Prostaglandins Other Lipid Mediat 2024; 172:106824. [PMID: 38438104 DOI: 10.1016/j.prostaglandins.2024.106824] [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: 10/10/2023] [Revised: 12/16/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune inflammatory disease that leads to joint destruction. Numerous pro-inflammatory mediators, including adipokines, play an important role in the pathogenesis of RA. OBJECTIVE The aim of the study was to investigate the relationships between selected plasma cytokines and expression of adiponectin and its receptors in the synovium and the infrapatellar fat pad in patients with RA and osteoarthritis (OA). METHODS Blood, synovium and fat pad samples from 18 patients with RA and 18 with OA were collected during joint replacement surgery. Spearman rank correlations between plasma concentrations of selected cytokines (IL-1β, IL-2, IL-4, IL-6, IL-7, IL-8, IL-10, IL-12 p40, IL-13, IL-17, G-CSF and GM-CSF) and the expression of adiponectin and its receptors were determined. Plasma levels of cytokines were determined using a magnetic bead-based multiplex assay, mRNA expression of adiponectin and its receptors were determined by real-time PCR. RESULTS In OA patients, there were significant positive correlations between adiponectin expression in the synovial membrane and plasma levels of IL-1β, IL-4, G-CSF and GM-CSF, as well as a significant positive correlation between adiponectin expression in the fat pad and plasma levels of GM-CSF. In addition, OA patients showed significant negative correlations between AdipoR1 and AdipoR2 expression in the synovial membrane and plasma IL-6 levels, as well as between AdipoR2 expression in the synovial membrane and plasma MCP-1 and TNF-α levels. In patients with RA, there were no significant correlations between adiponectin expression in the synovial membrane and infrapatellar fat pad and plasma levels of the cytokines studied. In addition, RA patients showed a statistically significant negative correlation between AdipoR1 expression in the synovial membrane and plasma levels of TNF-α, IL-7, IL-12 and IL-13, and a significant negative correlation between AdipoR1 expression in the infrapatellar fat pad and plasma levels of IL-1β. CONCLUSIONS Adiponectin and its receptors showed the correlations with several plasma cytokines, however, a thorough understanding of the role of adiponectin in RA and OA requires further investigation.
Collapse
Affiliation(s)
- Michał Czerewaty
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Małgorzata Łączna
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Paweł Dec
- Department of Plastic and Reconstructive Surgery, 109 Military Hospital, Szczecin, Poland
| | | | - Daniel Kotrych
- Department of Orthopedics, Traumatology and Orthopedic Oncology, Pomeranian Medical University, Szczecin, Poland
| | - Piotr Burszewski
- Department of Plastic and Reconstructive Surgery, 109 Military Hospital, Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland.
| |
Collapse
|
25
|
Bastard JP, Dridi-Brahimi I, Vatier C, Fellahi S, Fève B. Biological markers of adipose tissue: Adipokines. ANNALES D'ENDOCRINOLOGIE 2024; 85:171-172. [PMID: 38614158 DOI: 10.1016/j.ando.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2024]
Abstract
We currently have a large sum of clinical and experimental data documenting the involvement of numerous adipokines in the maintenance of energy homeostasis in healthy individuals and their dysregulation in diseases such as obesity, metabolic syndrome or type 2 diabetes. Despite the impressive discoveries made in this field over many years, much remains to be done before understanding all the physiological and pathological implications, and hoping for the development of other effective and safe therapeutic strategies. Two original adipokines will be taken as examples to illustrate these remarks, chemerin and neuregulin 4.
Collapse
Affiliation(s)
- Jean-Philippe Bastard
- Département de biochimie-pharmacologie, Assistance publique-Hôpitaux de Paris, hôpitaux universitaires Henri-Mondor, Créteil, France; FHU-SENEC, Inserm U955 and université Paris Est (UPEC), UMR U955, faculté de santé, Créteil, France.
| | - Imane Dridi-Brahimi
- Département de biochimie-pharmacologie, Assistance publique-Hôpitaux de Paris, hôpitaux universitaires Henri-Mondor, Créteil, France
| | - Camille Vatier
- Sorbonne université-Inserm, centre de recherche Saint-Antoine UMR S_938, 75012 Paris, France; Institut hospitalo-universitaire de cardio-métabolisme et nutrition (ICAN), Paris, France; Service d'endocrinologie-diabétologie, centre de référence des maladies rares de l'insulino-sécrétion et de l'insulino-sensibilité (PRISIS), hôpital Saint-Antoine, Assistance publique-Hôpitaux de Paris, 75012 Paris, France
| | - Soraya Fellahi
- Département de biochimie-pharmacologie, Assistance publique-Hôpitaux de Paris, hôpitaux universitaires Henri-Mondor, Créteil, France; Sorbonne université-Inserm, centre de recherche Saint-Antoine UMR S_938, 75012 Paris, France; Institut hospitalo-universitaire de cardio-métabolisme et nutrition (ICAN), Paris, France
| | - Bruno Fève
- Sorbonne université-Inserm, centre de recherche Saint-Antoine UMR S_938, 75012 Paris, France; Institut hospitalo-universitaire de cardio-métabolisme et nutrition (ICAN), Paris, France; Service d'endocrinologie-diabétologie, centre de référence des maladies rares de l'insulino-sécrétion et de l'insulino-sensibilité (PRISIS), hôpital Saint-Antoine, Assistance publique-Hôpitaux de Paris, 75012 Paris, France
| |
Collapse
|
26
|
Balejko EB, Bogacka A, Lichota J, Pawlus J. Effects of Bioactive Dietary Components on Changes in Lipid and Liver Parameters in Women after Bariatric Surgery and Procedures. Nutrients 2024; 16:1379. [PMID: 38732625 PMCID: PMC11085392 DOI: 10.3390/nu16091379] [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: 03/11/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Excess adipose tissue, as well as its distribution, correlates strongly with disorders of lipid and liver parameters and chronic inflammation. The pathophysiology of metabolic diseases caused by obesity is associated with the dysfunction of visceral adipose tissue. Effective and alternative interventions such as the Bioenteric Intragastric Balloon and bariatric surgeries such as the Roux-en-Y gastric bypass. The aim of this study was to assess the effect of modifying the recommended standard weight loss diet after bariatric surgery and procedures on reducing chronic inflammation in overweight patients. In the study, bioactive anti-inflammatory dietary components were used supportively. Changes in the concentrations of lipid parameters, liver parameters, antioxidant enzymes, cytokines, and chemokines were demonstrated. The enrichment of the diet, after bariatric surgery, with the addition of n-3 EFAs(Essential Fatty Acids), bioflavonoids, vitamins, and synbiotics resulted in higher weight losses in the patients in the study with a simultaneous reduction in parameters indicating liver dysfunction.
Collapse
Affiliation(s)
- Edyta Barbara Balejko
- Department of Commodity Science, Quality Assessment, Process Engineering and Human Nutrition, West Pomeranian University of Technology in Szczecin, 71-459 Szczecin, Poland
| | - Anna Bogacka
- Department of Commodity Science, Quality Assessment, Process Engineering and Human Nutrition, West Pomeranian University of Technology in Szczecin, 71-459 Szczecin, Poland
| | - Jarosław Lichota
- Unii Lubelskiej 1, Department of General, Minimally Invasive and Gastroenterological Surgery, Independent Public Clinical Hospital No. 1 of Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Jan Pawlus
- Unii Lubelskiej 1, Department of General, Minimally Invasive and Gastroenterological Surgery, Independent Public Clinical Hospital No. 1 of Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| |
Collapse
|
27
|
Emamian A, Emamian MH, Hashemi H, Fotouhi A. The association of ALT to HDL-C ratio with type 2 diabetes in 50-74 years old adults: a population-based study. Sci Rep 2024; 14:9390. [PMID: 38658745 PMCID: PMC11043380 DOI: 10.1038/s41598-024-60092-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/18/2024] [Indexed: 04/26/2024] Open
Abstract
There is limited information about the relationship between diabetes mellitus (DM) and ALT to HDL-C ratio. This study aims to investigate this relationship for the first time in Iran. The data of this study were taken from the third phase of the Shahroud Eye Cohort Study, which was conducted in 2019 with the participation of 4394 people aged 50-74. ALT and HDL-C levels were measured using a BT-1500 autoanalyzer. The mean ALT/HDL-C ratio was reported along with 95% confidence intervals (CI). The multiple logistic regression was used to examine the association between this ratio and DM, while controlling for the effects of other independent variables. The mean and standard deviation of the ALT/HDL-C ratio in all participants were 16.62 ± 11.22 (95% CI 16.28-16.96). The prevalence of DM was 34.7% and individuals with DM had a mean ALT/HDL-C ratio that was 1.80 units higher than those without diabetes (P < 0.001). Also, in individuals with DM, the HDL-C was found to be 0.035 (mmol/L) lower (P < 0.001), while ALT was 1.13 (IU/L) higher (P < 0.001) compared to those without diabetes. Additionally, after controlling for confounding factors, the odds of developing DM increased in a non-linear manner with an increase in the ALT/HDL-C ratio. Abdominal obesity, advanced age, female gender, and hypertension were also found to be associated with increased odds of DM. In conclusion, an increase in the ALT/ HDL-C ratiowas associated with higher odds of DM. This ratio can serve as an important predictor for diabetes mellitus.
Collapse
Affiliation(s)
- Abolfazl Emamian
- Student Research Committee, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Hassan Emamian
- Ophthalmic Epidemiology Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Hassan Hashemi
- Noor Research Center for Ophthalmic Epidemiology, Noor Eye Hospital, Tehran, Iran
| | - Akbar Fotouhi
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Wu O, Lu X, Leng J, Zhang X, Liu W, Yang F, Zhang H, Li J, Khederzadeh S, Liu X, Yuan C. Reevaluating Adiponectin's impact on obesity hypertension: a Chinese case-control study. BMC Cardiovasc Disord 2024; 24:208. [PMID: 38615012 PMCID: PMC11015577 DOI: 10.1186/s12872-024-03865-4] [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: 12/03/2023] [Accepted: 03/28/2024] [Indexed: 04/15/2024] Open
Abstract
BACKGROUND Obesity and hypertension are major risk factors for cardiovascular diseases that affect millions of people worldwide. Both conditions are associated with chronic low-grade inflammation, which is mediated by adipokines such as adiponectin. Adiponectin is the most abundant adipokine that has a beneficial impact on metabolic and vascular biology, while high serum concentrations are associated with some syndromes. This "adiponectin paradox" still needs to be clarified in obesity-associated hypertension. The aim of this study was to investigate how adiponectin affects blood pressure, inflammation, and metabolic function in obesity hypertension using a Chinese adult case-control study. METHODS A case-control study that had finished recruiting 153 subjects divided as four characteristic groups. Adiponectin serum levels were tested by ELISA in these subjects among these four characteristic Chinese adult physical examination groups. Waist circumference (WC), body mass index (BMI), systolic blood pressure (SB), diastolic blood pressure (DB), and other clinical laboratory data were collected. Analyzation of correlations between the research index and differences between groups was done by SPSS. RESULTS Serum adiponectin levels in the| normal healthy group (NH group) were significantly higher than those in the newly diagnosed untreated just-obesity group (JO group), and negatively correlated with the visceral adiposity index. With multiple linear egression analysis, it was found that, for serum adiponectin, gender, serum albumin (ALB), alanine aminotransferase (ALT) and high-density lipoprotein cholesterol (HDLC) were the significant independent correlates, and for SB, age and HDLC were the significant independent correlates, and for DB, alkaline phosphatase (ALP) was the significant independent correlate. The other variables did not reach significance in the model. CONCLUSIONS Our study reveals that adiponectin's role in obesity-hypertension is multifaceted and is influenced by the systemic metabolic homeostasis signaling axis. In obesity-related hypertension, compensatory effects, adiponectin resistance, and reduced adiponectin clearance from impaired kidneys and liver all contribute to the "adiponectin paradox".
Collapse
Affiliation(s)
- Ou Wu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People's Republic of China
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Xi Lu
- Hangzhou Vocational and Technical College, Hangzhou, Zhejiang, People's Republic of China
| | - Jianhang Leng
- Department of Central Laboratory/Medical Examination Center of Hangzhou, The Frist People's Hospital of Hangzhou, Hangzhou, Zhejiang, People's Republic of China
| | - Xingyu Zhang
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Wei Liu
- JFIntelligent Healthcare Technology Co., Ltd Building No.5-7, No.699 Tianxiang Avenue, Hi-Tech Zone, Nanchang, Jiangxi Province, People's Republic of China
| | - Fenfang Yang
- Department of Central Laboratory/Medical Examination Center of Hangzhou, The Frist People's Hospital of Hangzhou, Hangzhou, Zhejiang, People's Republic of China
| | - Hu Zhang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital Affiliated with Medical College of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jiajia Li
- Department of Central Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Saber Khederzadeh
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, People's Republic of China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaodong Liu
- Hangzhou Center for Disease Control and Prevention, Hangzhou, Zhejiang, People's Republic of China
| | - Chengda Yuan
- Department of Dermatology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
29
|
Liu S, Ezran C, Wang MFZ, Li Z, Awayan K, Long JZ, De Vlaminck I, Wang S, Epelbaum J, Kuo CS, Terrien J, Krasnow MA, Ferrell JE. An organism-wide atlas of hormonal signaling based on the mouse lemur single-cell transcriptome. Nat Commun 2024; 15:2188. [PMID: 38467625 PMCID: PMC10928088 DOI: 10.1038/s41467-024-46070-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 02/07/2024] [Indexed: 03/13/2024] Open
Abstract
Hormones mediate long-range cell communication and play vital roles in physiology, metabolism, and health. Traditionally, endocrinologists have focused on one hormone or organ system at a time. Yet, hormone signaling by its very nature connects cells of different organs and involves crosstalk of different hormones. Here, we leverage the organism-wide single cell transcriptional atlas of a non-human primate, the mouse lemur (Microcebus murinus), to systematically map source and target cells for 84 classes of hormones. This work uncovers previously-uncharacterized sites of hormone regulation, and shows that the hormonal signaling network is densely connected, decentralized, and rich in feedback loops. Evolutionary comparisons of hormonal genes and their expression patterns show that mouse lemur better models human hormonal signaling than mouse, at both the genomic and transcriptomic levels, and reveal primate-specific rewiring of hormone-producing/target cells. This work complements the scale and resolution of classical endocrine studies and sheds light on primate hormone regulation.
Collapse
Affiliation(s)
- Shixuan Liu
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Camille Ezran
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Michael F Z Wang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Zhengda Li
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kyle Awayan
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford, CA, USA
| | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Sheng Wang
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA
| | - Jacques Epelbaum
- Adaptive Mechanisms and Evolution (MECADEV), UMR 7179, National Center for Scientific Research, National Museum of Natural History, Brunoy, France
| | - Christin S Kuo
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jérémy Terrien
- Adaptive Mechanisms and Evolution (MECADEV), UMR 7179, National Center for Scientific Research, National Museum of Natural History, Brunoy, France
| | - Mark A Krasnow
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford, CA, USA.
| | - James E Ferrell
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
30
|
Sindzingre L, Bouaziz-Amar E, Mouton-Liger F, Cognat E, Dumurgier J, Vrillon A, Paquet C, Lilamand M. The role of adiponectin in Alzheimer's disease: A translational review. J Nutr Health Aging 2024; 28:100166. [PMID: 38280832 DOI: 10.1016/j.jnha.2024.100166] [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: 12/10/2023] [Accepted: 01/15/2024] [Indexed: 01/29/2024]
Abstract
Adiponectin is an adipokine playing a central role in the regulation of energy homeostasis, carbohydrate and lipid metabolism, as well as immunomodulation. The relationship between Alzheimer's disease (AD) and body composition has highlighted the bidirectional crosstalk between AD's pathophysiology and metabolic disorders. This review aimed to report the current state of knowledge about cellular and molecular mechanisms linking adiponectin and AD, in preclinical studies. Then, we reviewed human studies to assess the relationship between adiponectin levels and AD diagnosis. We also examined the risk of incident AD regarding the participants' baseline adiponectin level, as well as the relationship of adiponectin and cognitive decline in patients with AD. We conducted a systematic review, in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses reporting guideline, of studies published over the last decade on MEDLINE and Cochrane databases. Overall, we reviewed 34 original works about adiponectin in AD, including 11 preclinical studies, two both preclinical and human studies and 21 human studies. Preclinical studies brought convincing evidence for the neuroprotective role of adiponectin on several key mechanisms of AD. Human studies showed conflicting results regarding the relationship between AD and adiponectin levels, as well as regarding the cross-sectional association between cognitive function and adiponectin levels. Adiponectin did not appear as a predictor of incident AD, nor as a predictor of cognitive decline in patients with AD. Despite solid preclinical evidence suggesting the protective role of adiponectin in AD, inconsistent results in humans supports the need for further research.
Collapse
Affiliation(s)
- Louise Sindzingre
- Université Paris Cité, UMRS 1144, INSERM, Paris, France; Cognitive Neurology Center, AP-HP. Nord, Site Lariboisière Fernand-Widal, Paris, France.
| | - Elodie Bouaziz-Amar
- Université Paris Cité, UMRS 1144, INSERM, Paris, France; Biochemistry Department, AP-HP. Nord, Site Lariboisière Fernand-Widal, Paris, France
| | | | - Emmanuel Cognat
- Université Paris Cité, UMRS 1144, INSERM, Paris, France; Cognitive Neurology Center, AP-HP. Nord, Site Lariboisière Fernand-Widal, Paris, France
| | - Julien Dumurgier
- Université Paris Cité, UMRS 1144, INSERM, Paris, France; Cognitive Neurology Center, AP-HP. Nord, Site Lariboisière Fernand-Widal, Paris, France
| | - Agathe Vrillon
- Université Paris Cité, UMRS 1144, INSERM, Paris, France; Cognitive Neurology Center, AP-HP. Nord, Site Lariboisière Fernand-Widal, Paris, France
| | - Claire Paquet
- Université Paris Cité, UMRS 1144, INSERM, Paris, France; Cognitive Neurology Center, AP-HP. Nord, Site Lariboisière Fernand-Widal, Paris, France
| | - Matthieu Lilamand
- Université Paris Cité, UMRS 1144, INSERM, Paris, France; Geriatrics Department, AP-HP. Nord, Site Lariboisière Fernand-Widal, Paris, France
| |
Collapse
|
31
|
Hidalgo-Lozada GM, Villarruel-López A, Nuño K, García-García A, Sánchez-Nuño YA, Ramos-García CO. Clinically Effective Molecules of Natural Origin for Obesity Prevention or Treatment. Int J Mol Sci 2024; 25:2671. [PMID: 38473918 DOI: 10.3390/ijms25052671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
The prevalence and incidence of obesity and the comorbidities linked to it are increasing worldwide. Current therapies for obesity and associated pathologies have proven to cause a broad number of adverse effects, and often, they are overpriced or not affordable for all patients. Among the alternatives currently available, natural bioactive compounds stand out. These are frequently contained in pharmaceutical presentations, nutraceutical products, supplements, or functional foods. The clinical evidence for these molecules is increasingly solid, among which epigallocatechin-3-gallate, ellagic acid, resveratrol, berberine, anthocyanins, probiotics, carotenoids, curcumin, silymarin, hydroxy citric acid, and α-lipoic acid stand out. The molecular mechanisms and signaling pathways of these molecules have been shown to interact with the endocrine, nervous, and gastroenteric systems. They can regulate the expression of multiple genes and proteins involved in starvation-satiety processes, activate the brown adipose tissue, decrease lipogenesis and inflammation, increase lipolysis, and improve insulin sensitivity. This review provides a comprehensive view of nature-based therapeutic options to address the increasing prevalence of obesity. It offers a valuable perspective for future research and subsequent clinical practice, addressing everything from the molecular, genetic, and physiological bases to the clinical study of bioactive compounds.
Collapse
Affiliation(s)
| | - Angelica Villarruel-López
- Department of Pharmacobiology, University Center for Exact and Engineering Sciences, University of Guadalajara, Guadalajara 44430, Mexico
| | - Karla Nuño
- Department of Psychology, Education and Health, ITESO Jesuit University of Guadalajara, Guadalajara 45604, Mexico
| | - Abel García-García
- Institute of Science and Technology for Health Innovation, Guadalajara 44770, Mexico
- Department of Medical Clinic, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico
| | - Yaír Adonaí Sánchez-Nuño
- Department of Pharmacobiology, University Center for Exact and Engineering Sciences, University of Guadalajara, Guadalajara 44430, Mexico
| | | |
Collapse
|
32
|
Kim SY, Park SY, Kim JE. GULP1 deficiency reduces adipogenesis and glucose uptake via downregulation of PPAR signaling and disturbing of insulin/ERK signaling in 3T3-L1 cells. J Cell Physiol 2024; 239:e31173. [PMID: 38214103 DOI: 10.1002/jcp.31173] [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: 07/30/2023] [Revised: 11/10/2023] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
Obesity and metabolic disorders caused by alterations in lipid metabolism are major health issues in developed, affluent societies. Adipose tissue is the only organ that stores lipids and prevents lipotoxicity in other organs. Mature adipocytes can affect themselves and distant metabolism-related tissues by producing various adipokines, including adiponectin and leptin. The engulfment adaptor phosphotyrosine-binding domain-containing 1 (GULP1) regulates intracellular trafficking of glycosphingolipids and cholesterol, suggesting its close association with lipid metabolism. However, the role of GULP1 in adipocytes remains unknown. Therefore, this study aimed to investigate the function of GULP1 in adipogenesis, glucose uptake, and the insulin signaling pathway in adipocytes. A 3T3-L1 cell line with Gulp1 knockdown (shGulp1) and a 3T3-L1 control group (U6) were established. Changes in shGulp1 cells due to GULP1 deficiency were examined and compared to those in U6 cells using microarray analysis. Glucose uptake was monitored via insulin stimulation in shGulp1 and U6 cells using a 2-NBDG glucose uptake assay, and the insulin signaling pathway was investigated by western blot analysis. Adipogenesis was significantly delayed, lipid metabolism was altered, and several adipogenesis-related genes were downregulated in shGulp1 cells compared to those in U6 cells. Microarray analysis revealed significant inhibition of peroxisome proliferator-activated receptor signaling in shGulp1 cells compared with U6 cells. The production and secretion of adiponectin as well as the expression of adiponectin receptor were decreased in shGulp1 cells. In particular, compared with U6 cells, glucose uptake via insulin stimulation was significantly decreased in shGulp1 cells through the disturbance of ERK1/2 phosphorylation. This is the first study to identify the role of GULP1 in adipogenesis and insulin-stimulated glucose uptake by adipocytes, thereby providing new insights into the differentiation and functions of adipocytes and the metabolism of lipids and glucose, which can help better understand metabolic diseases.
Collapse
Affiliation(s)
- Soon-Young Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung-Yoon Park
- Department of Biochemistry, School of Medicine, Dongguk University, Gyeongju, Republic of Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Biomedical Science, BK21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
33
|
Choi HN, Kim JI. Daraesoon (shoot of hardy kiwi) mitigates hyperglycemia in db/db mice by alleviating insulin resistance and inflammation. Nutr Res Pract 2024; 18:88-97. [PMID: 38352218 PMCID: PMC10861346 DOI: 10.4162/nrp.2024.18.1.88] [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: 12/13/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Mitigating insulin resistance and hyperglycemia is associated with a decreased risk of diabetic complications. The effect of Daraesoon (shoot of hardy kiwi, Actinidia arguta) on hyperglycemia was investigated using a type 2 diabetes animal model. MATERIALS/METHODS Seven-week-old db/db mice were fed either an AIN-93G diet or a diet containing 0.4% of a 70% ethanol extract of Daraesoon, whereas db/+ mice were fed the AIN-93G diet for 7 weeks. RESULTS Consumption of Daraesoon significantly reduced serum glucose and blood glycated hemoglobin levels, along with homeostasis model assessment for insulin resistance in db/db mice. Conversely, Daraesoon elevated the serum adiponectin levels compared to the db/db control group. Furthermore, Daraesoon significantly decreased both serum and hepatic triglyceride levels, as well as serum total cholesterol levels. Additionally, consumption of Daraesoon resulted in decreased hepatic tumor necrosis factor-α and monocyte chemoattractant protein-1 expression. CONCLUSIONS These results suggest that hypoglycemic effect of Daraesoon is mediated through the improvement of insulin resistance and the downregulation of pro-inflammatory cytokine expression in db/db mice.
Collapse
Affiliation(s)
- Ha-Neul Choi
- Department of Food and Nutrition, Changwon National University, Changwon 51140, Korea
| | - Jung-In Kim
- Institute of Digital Anti-Aging Healthcare, Inje University, Gimhae 50834, Korea
| |
Collapse
|
34
|
Athar F, Karmani M, Templeman N. Metabolic hormones are integral regulators of female reproductive health and function. Biosci Rep 2024; 44:BSR20231916. [PMID: 38131197 PMCID: PMC10830447 DOI: 10.1042/bsr20231916] [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: 11/03/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023] Open
Abstract
The female reproductive system is strongly influenced by nutrition and energy balance. It is well known that food restriction or energy depletion can induce suppression of reproductive processes, while overnutrition is associated with reproductive dysfunction. However, the intricate mechanisms through which nutritional inputs and metabolic health are integrated into the coordination of reproduction are still being defined. In this review, we describe evidence for essential contributions by hormones that are responsive to food intake or fuel stores. Key metabolic hormones-including insulin, the incretins (glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1), growth hormone, ghrelin, leptin, and adiponectin-signal throughout the hypothalamic-pituitary-gonadal axis to support or suppress reproduction. We synthesize current knowledge on how these multifaceted hormones interact with the brain, pituitary, and ovaries to regulate functioning of the female reproductive system, incorporating in vitro and in vivo data from animal models and humans. Metabolic hormones are involved in orchestrating reproductive processes in healthy states, but some also play a significant role in the pathophysiology or treatment strategies of female reproductive disorders. Further understanding of the complex interrelationships between metabolic health and female reproductive function has important implications for improving women's health overall.
Collapse
Affiliation(s)
- Faria Athar
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Muskan Karmani
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Nicole M. Templeman
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| |
Collapse
|
35
|
Hafiane A. Adiponectin-mediated regulation of the adiponectin cascade in cardiovascular disease: Updates. Biochem Biophys Res Commun 2024; 694:149406. [PMID: 38134479 DOI: 10.1016/j.bbrc.2023.149406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/03/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023]
Abstract
The endocrine function of white adipose tissue is characterized by the synthesis of one its main hormones: adiponectin. Although the biological role of adiponectin has not been fully defined, clinical and experimental observations have shown that low plasma concentrations of adiponectin participate in the prevalence of insulin resistance and cardiovascular diseases, mainly in obese patients. Adiponectin also exerts its effects on the heart and blood vessels, thereby influencing their physiology. Studying the effects of adiponectin presents some complexities, primarily due to potential cross-interactions and interference with other pathways, such as the AdipoR1/R2 pathways. Under optimal conditions, the activation of the adiponectin cascade may involve signals such as AMPK and PPARα. Interestingly, these pathways may trigger similar responses, such as fatty acid oxidation. Understanding the downstream effectors of these pathways is crucial to comprehend the extent to which adiponectin signaling impacts metabolism. In this review, the aim is to explore the current mechanisms that regulate the adiponectin pathways. Additionally, updates on the major downstream factors involved in adiponectin signaling are provided, specifically in relation to metabolic syndrome and atherosclerosis.
Collapse
Affiliation(s)
- Anouar Hafiane
- Research Institute, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
36
|
Zhou M, Tamburini I, Van C, Molendijk J, Nguyen CM, Chang IYY, Johnson C, Velez LM, Cheon Y, Yeo R, Bae H, Le J, Larson N, Pulido R, Nascimento-Filho CHV, Jang C, Marazzi I, Justice J, Pannunzio N, Hevener AL, Sparks L, Kershaw EE, Nicholas D, Parker BL, Masri S, Seldin MM. Leveraging inter-individual transcriptional correlation structure to infer discrete signaling mechanisms across metabolic tissues. eLife 2024; 12:RP88863. [PMID: 38224289 PMCID: PMC10945578 DOI: 10.7554/elife.88863] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
Inter-organ communication is a vital process to maintain physiologic homeostasis, and its dysregulation contributes to many human diseases. Given that circulating bioactive factors are stable in serum, occur naturally, and are easily assayed from blood, they present obvious focal molecules for therapeutic intervention and biomarker development. Recently, studies have shown that secreted proteins mediating inter-tissue signaling could be identified by 'brute force' surveys of all genes within RNA-sequencing measures across tissues within a population. Expanding on this intuition, we reasoned that parallel strategies could be used to understand how individual genes mediate signaling across metabolic tissues through correlative analyses of gene variation between individuals. Thus, comparison of quantitative levels of gene expression relationships between organs in a population could aid in understanding cross-organ signaling. Here, we surveyed gene-gene correlation structure across 18 metabolic tissues in 310 human individuals and 7 tissues in 103 diverse strains of mice fed a normal chow or high-fat/high-sucrose (HFHS) diet. Variation of genes such as FGF21, ADIPOQ, GCG, and IL6 showed enrichments which recapitulate experimental observations. Further, similar analyses were applied to explore both within-tissue signaling mechanisms (liver PCSK9) and genes encoding enzymes producing metabolites (adipose PNPLA2), where inter-individual correlation structure aligned with known roles for these critical metabolic pathways. Examination of sex hormone receptor correlations in mice highlighted the difference of tissue-specific variation in relationships with metabolic traits. We refer to this resource as gene-derived correlations across tissues (GD-CAT) where all tools and data are built into a web portal enabling users to perform these analyses without a single line of code (gdcat.org). This resource enables querying of any gene in any tissue to find correlated patterns of genes, cell types, pathways, and network architectures across metabolic organs.
Collapse
Affiliation(s)
- Mingqi Zhou
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Ian Tamburini
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Cassandra Van
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Jeffrey Molendijk
- Department of Anatomy and Physiology, University of MelbourneMelbourneAustralia
| | - Christy M Nguyen
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | | | - Casey Johnson
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Leandro M Velez
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Youngseo Cheon
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Reichelle Yeo
- Translational Research Institute, AdventHealthOrlandoUnited States
| | - Hosung Bae
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Johnny Le
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Natalie Larson
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Ron Pulido
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Carlos HV Nascimento-Filho
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Cholsoon Jang
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Ivan Marazzi
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Jamie Justice
- Veterans Administration Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center (GRECC)Los AngelesUnited States
| | - Nicholas Pannunzio
- Divison of Hematology/Oncology, Department of Medicine, UC Irvine HealthIrvineUnited States
| | - Andrea L Hevener
- Department of Medicine, Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine at UCLALos AngelesUnited States
- Iris Cantor-UCLA Women’s Health Research Center, David Geffen School of Medicine at UCLALos AngelesUnited States
| | - Lauren Sparks
- Translational Research Institute, AdventHealthOrlandoUnited States
| | - Erin E Kershaw
- Division of Endocrinology, Department of Medicine, University of PittsburgPittsburghUnited States
| | - Dequina Nicholas
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California IrvineIrvineUnited States
| | - Benjamin L Parker
- Department of Anatomy and Physiology, University of MelbourneMelbourneAustralia
| | - Selma Masri
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| | - Marcus M Seldin
- Department of Biological Chemistry, UC IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, UC IrvineIrvineUnited States
| |
Collapse
|
37
|
Kim OY, Song J. Important roles of linoleic acid and α-linolenic acid in regulating cognitive impairment and neuropsychiatric issues in metabolic-related dementia. Life Sci 2024; 337:122356. [PMID: 38123015 DOI: 10.1016/j.lfs.2023.122356] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Metabolic syndrome (MetS), which is characterized by insulin resistance, high blood glucose, obesity, and dyslipidemia, is known to increase the risk of dementia accompanied by memory loss and depression. The direct pathways and specific mechanisms in the central nervous system (CNS) for addressing fatty acid imbalances in MetS have not yet been fully elucidated. Among polyunsaturated acids, linoleic acid (LA, n6-PUFA) and α-linolenic acid (ALA, n3-PUFA), which are two essential fatty acids that should be provided by food sources (e.g., vegetable oils and seeds), have been reported to regulate various cellular mechanisms including apoptosis, inflammatory responses, mitochondrial biogenesis, and insulin signaling. Furthermore, inadequate intake of LA and ALA is reported to be involved in neuropathology and neuropsychiatric diseases as well as imbalanced metabolic conditions. Herein, we review the roles of LA and ALA on metabolic-related dementia focusing on insulin resistance, dyslipidemia, synaptic plasticity, cognitive function, and neuropsychiatric issues. This review suggests that LA and ALA are important fatty acids for concurrent treatment of both MetS and neurological problems.
Collapse
Affiliation(s)
- Oh Yoen Kim
- Department of Food Science and Nutrition, Dong A University, Busan, Republic of Korea; Department of Health Sciences, Graduate School of Dong-A University, Busan, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Engin A. Adiponectin Resistance in Obesity: Adiponectin Leptin/Insulin Interaction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:431-462. [PMID: 39287861 DOI: 10.1007/978-3-031-63657-8_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adiponectin (APN) levels in obesity are negatively correlated with chronic subclinical inflammation markers. The hypertrophic adipocytes cause obesity-linked insulin resistance and metabolic syndrome. Furthermore, macrophage polarization is a key determinant regulating adiponectin receptor (AdipoR1/R2) expression and differential adiponectin-mediated macrophage inflammatory responses in obese individuals. In addition to decrease in adiponectin concentrations, the decline in AdipoR1/R2 messenger ribonucleic acid (mRNA) expression leads to a decrement in adiponectin binding to cell membrane, and this turns into attenuation in the adiponectin effects. This is defined as APN resistance, and it is linked with insulin resistance in high-fat diet-fed subjects. The insulin-resistant group has a significantly higher leptin-to-APN ratio. The leptin-to-APN ratio is more than twofold higher in obese individuals. An increase in expression of AdipoRs restores insulin sensitivity and β-oxidation of fatty acids via triggering intracellular signal cascades. The ratio of high molecular weight to total APN is defined as the APN sensitivity index (ASI). This index is correlated to insulin sensitivity. Homeostasis model of assessment (HOMA)-APN and HOMA-estimated insulin resistance (HOMA-IR) are the most suitable methods to estimate the metabolic risk in metabolic syndrome. While morbidly obese patients display a significantly higher plasma leptin and soluble (s)E-selectin concentrations, leptin-to-APN ratio, there is a significant negative correlation between leptin-to-APN ratio and sP-selectin in obese patients. When comparing the metabolic dysregulated obese group with the metabolically healthy obese group, postprandial triglyceride clearance, insulin resistance, and leptin resistance are significantly delayed following the oral fat tolerance test in the first group. A neuropeptide, Spexin (SPX), is positively correlated with the quantitative insulin sensitivity check index (QUICKI) and APN. APN resistance together with insulin resistance forms a vicious cycle. Despite normal or high APN levels, an impaired post-receptor signaling due to adaptor protein-containing pleckstrin homology domain, phosphotyrosine-binding domain, and leucine zipper motif 1 (APPL1)/APPL2 may alter APN efficiency and activity. However, APPL2 blocks adiponectin signaling through AdipoR1 and AdipoR2 because of the competitive inhibition of APPL1. APPL1, the intracellular binding partner of AdipoRs, is also an important mediator of adiponectin-dependent insulin sensitization. The elevated adiponectin levels with adiponectin resistance are compensatory responses in the condition of an unusual discordance between insulin resistance and APN unresponsiveness. Hypothalamic recombinant adeno-associated virus (rAAV)-leptin (Lep) gene therapy reduces serum APN levels, and it is a more efficient strategy for long-term weight maintenance.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
39
|
Avtanski D, Stojchevski R. Significance of Adipose Tissue as an Endocrine Organ. CONTEMPORARY ENDOCRINOLOGY 2024:1-46. [DOI: 10.1007/978-3-031-72570-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
40
|
Ahuja A, Zboinski E, das S, Zhu X, Ma Q, Xie Y, Tu Q, Chen J. Antidiabetic features of AdipoAI, a novel AdipoR agonist. Cell Biochem Funct 2024; 42:e3910. [PMID: 38269524 PMCID: PMC10811407 DOI: 10.1002/cbf.3910] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 01/26/2024]
Abstract
Adiponectin is an antidiabetic endogenous adipokine that plays a protective role against the unfavorable metabolic sequelae of obesity. Recent evidence suggests a sinister link between hypoadiponectinemia and development of insulin resistance/type 2 diabetes (T2D). Adiponectin's insulin-sensitizing property is mediated through the specific adiponectin receptors R1 and R2, which activate the AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor (PPAR) α pathways. AdipoAI is a novel synthetic analogue of endogenous adiponectin with possibly similar pharmacological effects. Thus, there is a need of orally active small molecules that activate Adipoq subunits, and their downstream signaling, which could ameliorate obesity related type 2 diabetes. In the study we aim to investigate the effects of AdipoAI on obesity and T2D. Through in-vitro and in-vivo analyses, we investigated the antidiabetic potentials of AdipoAI and compared it with AdipoRON, another orally active adiponectin receptors agonist. Our results showed that in-vitro treatment of AdipoAI (0-5 µM) increased adiponectin receptor subunits AdipoR1/R2 with increase in AMPK and APPL1 protein expression in C2C12 myotubes. Similarly, in-vivo, oral administration of AdipoAI (25 mg/kg) observed similar effects as that of AdipoRON (50 mg/kg) with improved control of blood glucose and insulin sensitivity in diet-induced obesity (DIO) mice models. Further, AdipoAI significantly reduced epididymal fat content with decrease in inflammatory markers and increase in PPAR-α and AMPK levels and exhibited hepatoprotective effects in liver. Further, AdipoAI and AdipoRON also observed similar results in adipose tissue. Thus, our results suggest that low doses of orally active small molecule agonist of adiponectin AdipoAI can be a promising therapeutic target for obesity and T2D.
Collapse
Affiliation(s)
- Akash Ahuja
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Elissa Zboinski
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Siddhartha das
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Xiaofang Zhu
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Qian Ma
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
- Department of General Dentistry, Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
| | - Ying Xie
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Qisheng Tu
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | - Jake Chen
- Division of Oral Biology, Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
- Dept. of Developmental, Molecular and Chemical Biology, Tufts School of Medicine; Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
41
|
Mester P, Räth U, Schmid S, Müller M, Buechler C, Pavel V. Exploring the Relationship between Plasma Adiponectin, Gender, and Underlying Diseases in Severe Illness. Biomedicines 2023; 11:3287. [PMID: 38137508 PMCID: PMC10741480 DOI: 10.3390/biomedicines11123287] [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: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Adiponectin is low in obesity, plays a crucial role in metabolic health, and, moreover, possesses immunoregulatory properties. However, studies examining its levels in patients with systemic inflammatory response syndrome (SIRS) or sepsis have yielded conflicting results. While females typically have higher systemic adiponectin levels than males, research on sex-specific associations in this context is limited. In this study of 156 SIRS/sepsis patients, including those with liver cirrhosis, we aimed to explore the relationship between plasma adiponectin, body mass index (BMI), gender, disease severity, and underlying etiological conditions. Our findings revealed that patients with liver cirrhosis, who are susceptible to infections, exhibited elevated circulating adiponectin levels, irrespective of sex. When excluding cirrhosis patients, plasma adiponectin levels were similar between male SIRS/sepsis patients and controls but lower in female patients compared to female controls. Plasma adiponectin was inversely related to BMI in female but not male patients. Further analysis within the non-cirrhosis subgroup demonstrated no significant differences in adiponectin levels between sexes among SIRS, sepsis, and septic shock patients. Ventilation, dialysis, and vasopressor therapy had no discernible impact on adiponectin levels in either sex. A negative correlation between adiponectin and C-reactive protein (CRP) existed in males only. Notably, patients with pancreatitis showed the lowest plasma adiponectin concentrations, although sex-specific differences were not significant. Infection with Gram-negative or Gram-positive bacteria had minimal effects on plasma adiponectin levels in both sexes. However, infection with the severe acute respiratory syndrome coronavirus type 2 led to decreased adiponectin levels in females exclusively. Multivariate analysis considering all factors affecting plasma adiponectin levels in males or females identified BMI in females and CRP levels in males to predict plasma adiponectin levels in SIRS/sepsis patients. Additionally, our study observed a trend where the 25 patients who did not survive had higher plasma adiponectin levels, particularly among males. In summary, our investigation highlights the influence of underlying diseases and sex on plasma adiponectin levels in SIRS/sepsis patients, shedding light on potential implications for disease management and prognosis.
Collapse
|
42
|
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
|
43
|
Valentini A, Cardillo C, Della Morte D, Tesauro M. The Role of Perivascular Adipose Tissue in the Pathogenesis of Endothelial Dysfunction in Cardiovascular Diseases and Type 2 Diabetes Mellitus. Biomedicines 2023; 11:3006. [PMID: 38002006 PMCID: PMC10669084 DOI: 10.3390/biomedicines11113006] [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: 09/25/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023] Open
Abstract
Cardiovascular diseases (CVDs) and type 2 diabetes mellitus (T2DM) are two of the four major chronic non-communicable diseases (NCDs) representing the leading cause of death worldwide. Several studies demonstrate that endothelial dysfunction (ED) plays a central role in the pathogenesis of these chronic diseases. Although it is well known that systemic chronic inflammation and oxidative stress are primarily involved in the development of ED, recent studies have shown that perivascular adipose tissue (PVAT) is implicated in its pathogenesis, also contributing to the progression of atherosclerosis and to insulin resistance (IR). In this review, we describe the relationship between PVAT and ED, and we also analyse the role of PVAT in the pathogenesis of CVDs and T2DM, further assessing its potential therapeutic target with the aim of restoring normal ED and reducing global cardiovascular risk.
Collapse
Affiliation(s)
- Alessia Valentini
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (A.V.); (D.D.M.)
| | - Carmine Cardillo
- Department of Aging, Policlinico A. Gemelli IRCCS, 00168 Roma, Italy;
- Department of Translational Medicine and Surgery, Catholic University, 00168 Rome, Italy
| | - David Della Morte
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (A.V.); (D.D.M.)
| | - Manfredi Tesauro
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (A.V.); (D.D.M.)
| |
Collapse
|
44
|
Zhou M, Tamburini IJ, Van C, Molendijk J, Nguyen CM, Chang IYY, Johnson C, Velez LM, Cheon Y, Yeo RX, Bae H, Le J, Larson N, Pulido R, Filho C, Jang C, Marazzi I, Justice JN, Pannunzio N, Hevener A, Sparks LM, Kershaw EE, Nicholas D, Parker B, Masri S, Seldin M. Leveraging inter-individual transcriptional correlation structure to infer discrete signaling mechanisms across metabolic tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540142. [PMID: 37214953 PMCID: PMC10197628 DOI: 10.1101/2023.05.10.540142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Abstract/IntroductionInter-organ communication is a vital process to maintain physiologic homeostasis, and its dysregulation contributes to many human diseases. Beginning with the discovery of insulin over a century ago, characterization of molecules responsible for signal between tissues has required careful and elegant experimentation where these observations have been integral to deciphering physiology and disease. Given that circulating bioactive factors are stable in serum, occur naturally, and are easily assayed from blood, they present obvious focal molecules for therapeutic intervention and biomarker development. For example, physiologic dissection of the actions of soluble proteins such as proprotein convertase subtilisin/kexin type 9 (PCSK9) and glucagon-like peptide 1 (GLP1) have yielded among the most promising therapeutics to treat cardiovascular disease and obesity, respectively1–4. A major obstacle in the characterization of such soluble factors is that defining their tissues and pathways of action requires extensive experimental testing in cells and animal models. Recently, studies have shown that secreted proteins mediating inter-tissue signaling could be identified by “brute-force” surveys of all genes within RNA-sequencing measures across tissues within a population5–9. Expanding on this intuition, we reasoned that parallel strategies could be used to understand how individual genes mediate signaling across metabolic tissues through correlative analyses of gene variation between individuals. Thus, comparison of quantitative levels of gene expression relationships between organs in a population could aid in understanding cross-organ signaling. Here, we surveyed gene-gene correlation structure across 18 metabolic tissues in 310 human individuals and 7 tissues in 103 diverse strains of mice fed a normal chow or HFHS diet. Variation of genes such asFGF21, ADIPOQ, GCGandIL6showed enrichments which recapitulate experimental observations. Further, similar analyses were applied to explore both within-tissue signaling mechanisms (liverPCSK9) as well as genes encoding enzymes producing metabolites (adiposePNPLA2), where inter-individual correlation structure aligned with known roles for these critical metabolic pathways. Examination of sex hormone receptor correlations in mice highlighted the difference of tissue-specific variation in relationships with metabolic traits. We refer to this resource asGene-DerivedCorrelationsAcrossTissues (GD-CAT) where all tools and data are built into a web portal enabling users to perform these analyses without a single line of code (gdcat.org). This resource enables querying of any gene in any tissue to find correlated patterns of genes, cell types, pathways and network architectures across metabolic organs.
Collapse
Affiliation(s)
- Mingqi Zhou
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Ian J. Tamburini
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Cassandra Van
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Jeffrey Molendijk
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Christy M Nguyen
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | | | - Casey Johnson
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Leandro M. Velez
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Youngseo Cheon
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Reichelle X. Yeo
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Hosung Bae
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Johnny Le
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Natalie Larson
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Ron Pulido
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Carlos Filho
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Ivan Marazzi
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Jamie N. Justice
- Veterans Administration Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center (GRECC), Los Angeles, CA, USA
| | - Nicholas Pannunzio
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Andrea Hevener
- Department of Medicine, Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Iris Cantor-UCLA Women’s Health Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Lauren M. Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Erin E. Kershaw
- Department of Internal Medicine, Section On Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Dequina Nicholas
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Benjamin Parker
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Selma Masri
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Marcus Seldin
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
| |
Collapse
|
45
|
Saliani N, Montasser Kouhsari S, Izad M. The Potential Hepatoprotective Effect of Vaccinium arctostaphylos L. Fruit Extract in Diabetic Rat. CELL JOURNAL 2023; 25:717-726. [PMID: 37865880 PMCID: PMC10591264 DOI: 10.22074/cellj.2023.2004742.1328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/08/2023] [Accepted: 08/28/2023] [Indexed: 10/23/2023]
Abstract
OBJECTIVE Vaccinium arctostaphylos has traditionally been employed in Iranian folk medicine to treat diabetes. However, the precise molecular mechanisms underlying its antidiabetic properties remain incompletely understood. The current experiment intended to explore the modulatory effects of V. arctostaphylos fruit ethanolic extract (VAE) on biochemical and molecular events in the livers of diabetic rats. MATERIALS AND METHODS In this experimental study, male Wistar rats were randomly assigned to four groups: normal control, normal rats with VAE treatment, diabetic control, and diabetic rats with VAE treatment. Following 42 days of treatment, the impact of VAE on diabetes-induced rats was assessed by measuring various serum biochemical parameters, including insulin, free fatty acids (FFA), tumor necrosis factor-α (TNF-α), reactive oxygen species (ROS), and adiponectin levels. The activities of hepatic carbohydrate metabolic enzymes and glycogen content were determined. Additionally, expression levels of selected genes implicated in carbohydrate/lipid metabolism and miR-27b expression were evaluated. H and E-stained liver sections were prepared for light microscopy examination. RESULTS Treatment with VAE elevated levels of insulin and adiponectin that reduced levels of FFA, ROS, and TNF-α in the serum of diabetic rats. VAE-treated rats exhibited increased activities of hepatic glucokinase (GK), glucose-6-phosphate dehydrogenase (G6PD), and glycogen concentrations, in conjunction with decreased activities of glucose-6-phosphatase (G6Pase) and fructose-1,6-bisphosphatase (FBPase). Furthermore, VAE significantly upregulated the transcription levels of hepatic insulin receptor substrate 1 (Irs1) and glucose transporter 2 (Glut2), while considerably downregulated the expression of peroxisome proliferator-activated receptor gamma (Pparg) and sterol regulatory element-binding protein 1c (Srebp1c). VAE remarkably enhanced the expression of miR27-b in the hepatic tissues of diabetic rats. Abnormal histological signs were dramatically normalized in diabetic rats receiving VAE compared to those in the diabetic control group. CONCLUSION Our findings underscore the hypoglycemic and hypolipidemic activities of V. arctostaphylos and assist in better comprehension of its antidiabetic properties.
Collapse
Affiliation(s)
- Negar Saliani
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Shideh Montasser Kouhsari
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Maryam Izad
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Gupta A, Gupta P, Singh AK, Gupta V. Association of adipokines with insulin resistance and metabolic syndrome including obesity and diabetes. GHM OPEN 2023; 3:7-19. [PMID: 40143837 PMCID: PMC11933950 DOI: 10.35772/ghmo.2023.01004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 03/28/2025]
Abstract
Adipose tissue (AT) acts as a highly active endocrine organ, which secretes a wide range of adipokine hormones. In the past few years, several adipokines (leptin, adiponectin, resistin etc.) have been discovered showing metabolic consequences in relation to insulin resistance (IR), obesity and diabetes. These adipokines are considered to be an important component playing an important role in the regulation of energy metabolism. They have been shown to be involved in the pathogenesis of metabolic syndrome (MetS) and cardiac diseases. The current article provides a holistic summary of recent knowledge on adipokines and emphasizes their importance in association with IR, obesity, diabetes and MetS. Adipokines such as leptin, adiponectin, resistin and tumor necrosis factor-alpha (TNF-α) have been involved in the regulation of an array of metabolic functions and disease associated with it, e.g. appetite and energy balance of the body, suppression of atherosclerosis and liver fibrosis, obesity with type 2 diabetes (T2D) and IR. An important adipokine, Interleukin-6 (IL-6), also correlates positively with human obesity and IR and also the elevated level of IL-6 predicts development of T2D. All of these hormones have important correlation with energy homeostasis, glucose and lipid metabolism, cardiovascular function and immunity. All the possible connections have extended the biological emphasis of AT secreted adipokines as an investigator in the development of MetS, and are now no longer considered as only an energy storage site.
Collapse
Affiliation(s)
- Abhishek Gupta
- Department of Physiology, King George's Medical University, Lucknow, India
| | - Priyanka Gupta
- Department of Medicine, King George's Medical University, Lucknow, India
| | - Arun Kumar Singh
- Department of Physiology, King George's Medical University, Lucknow, India
| | - Vani Gupta
- Department of Physiology, King George's Medical University, Lucknow, India
| |
Collapse
|
47
|
Blandin A, Amosse J, Froger J, Hilairet G, Durcin M, Fizanne L, Ghesquière V, Prieur X, Chaigneau J, Vergori L, Dray C, Pradère JP, Blandin S, Dupont J, Ducluzeau PH, Dubois S, Boursier J, Cariou B, Le Lay S. Extracellular vesicles are carriers of adiponectin with insulin-sensitizing and anti-inflammatory properties. Cell Rep 2023; 42:112866. [PMID: 37605533 DOI: 10.1016/j.celrep.2023.112866] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/06/2023] [Accepted: 07/11/2023] [Indexed: 08/23/2023] Open
Abstract
Recent evidence supporting that adipose tissue (AT)-derived extracellular vesicles (EVs) carry an important part of the AT secretome led us to characterize the EV-adipokine profile. In addition to evidencing a high AT-derived EV secretion ability that is further increased by obesity, we identify enrichment of oligomeric forms of adiponectin in small EVs (sEVs). This adipokine is mainly distributed at the EV external surface as a result of nonspecific adsorption of soluble adiponectin. EVs also constitute stable conveyors of adiponectin in the blood circulation. Adiponectin-enriched sEVs display in vitro insulin-sensitizing effects by binding to regular adiponectin receptors. Adoptive transfer of adiponectin-enriched sEVs in high-fat-diet-fed mice prevents animals from gaining weight and ameliorated insulin resistance and tissue inflammation, with major effects observed in the AT and liver. Our results therefore provide information regarding adiponectin-related metabolic responses by highlighting EVs as delivery platforms of metabolically active forms of adiponectin molecules.
Collapse
Affiliation(s)
- Alexia Blandin
- L'Institut du Thorax, CNRS, INSERM, Nantes Université, 44000 Nantes, France; Université Angers, SFR ICAT, 49000 Angers, France; L'institut du Thorax, CNRS, INSERM, CHU Nantes, Nantes Université, 44000 Nantes, France
| | - Jérémy Amosse
- Université Angers, SFR ICAT, 49000 Angers, France; IRSET Laboratory, Inserm, UMR 1085, Rennes, France
| | - Josy Froger
- L'Institut du Thorax, CNRS, INSERM, Nantes Université, 44000 Nantes, France; Université Angers, SFR ICAT, 49000 Angers, France
| | | | - Maëva Durcin
- Université Angers, SFR ICAT, 49000 Angers, France
| | - Lionel Fizanne
- HIFIH, CHU Angers, Université Angers, SFR ICAT, 49000 Angers, France
| | - Valentine Ghesquière
- L'Institut du Thorax, CNRS, INSERM, Nantes Université, 44000 Nantes, France; Université Angers, SFR ICAT, 49000 Angers, France
| | - Xavier Prieur
- L'Institut du Thorax, CNRS, INSERM, Nantes Université, 44000 Nantes, France
| | - Julien Chaigneau
- HIFIH, CHU Angers, Université Angers, SFR ICAT, 49000 Angers, France
| | | | - Cédric Dray
- RESTORE, UMR 1301 Inserm, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | | | - Stéphanie Blandin
- CHU Nantes, CNRS, Inserm BioCore US16, SFR Bonamy, Nantes Université, 44000 Nantes, France
| | - Joëlle Dupont
- CNRS, IFCE, INRAE, PRC, Université de Tours, 37380 Nouzilly, France
| | - Pierre-Henri Ducluzeau
- CNRS, IFCE, INRAE, PRC, Université de Tours, 37380 Nouzilly, France; Service de Médecine Interne, Unité d'Endocrinologie Diabétologie et Nutrition, Centre Hospitalier Universitaire et Faculté de Médecine, Université de Tours, Tours, France
| | | | - Jérôme Boursier
- HIFIH, CHU Angers, Université Angers, SFR ICAT, 49000 Angers, France; CHU Angers, Angers, France
| | - Bertrand Cariou
- L'Institut du Thorax, CNRS, INSERM, Nantes Université, 44000 Nantes, France
| | - Soazig Le Lay
- L'Institut du Thorax, CNRS, INSERM, Nantes Université, 44000 Nantes, France; Université Angers, SFR ICAT, 49000 Angers, France.
| |
Collapse
|
48
|
Garella R, Bernacchioni C, Chellini F, Tani A, Palmieri F, Parigi M, Guasti D, Cassioli E, Castellini G, Ricca V, Bani D, Sassoli C, Donati C, Squecco R. Adiponectin Modulates Smooth Muscle Cell Morpho-Functional Properties in Murine Gastric Fundus via Sphingosine Kinase 2 Activation. Life (Basel) 2023; 13:1812. [PMID: 37763216 PMCID: PMC10532860 DOI: 10.3390/life13091812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/31/2023] [Accepted: 08/15/2023] [Indexed: 09/29/2023] Open
Abstract
Adipokines are peptide hormones produced by the adipose tissue involved in several biological functions. Among adipokines, adiponectin (ADPN) has antidiabetic and anti-inflammatory properties. It can also modulate food intake at central and peripheral levels, acting on hypothalamus and facilitating gastric relaxation. ADPN exerts its action interacting with two distinct membrane receptors and triggering some well-defined signaling cascades. The ceramidase activity of ADPN receptor has been reported in many tissues: it converts ceramide into sphingosine. In turn, sphingosine kinase (SK) phosphorylates it into sphingosine-1 phosphate (S1P), a crucial mediator of many cellular processes including contractility. Using a multidisciplinary approach that combined biochemical, electrophysiological and morphological investigations, we explored for the first time the possible role of S1P metabolism in mediating ADPN effects on the murine gastric fundus muscle layer. By using a specific pharmacological inhibitor of SK2, we showed that ADPN affects smooth muscle cell membrane properties and contractile machinery via SK2 activation in gastric fundus, adding a piece of knowledge to the action mechanisms of this hormone. These findings help to identify ADPN and its receptors as new therapeutic targets or as possible prognostic markers for diseases with altered energy balance and for pathologies with fat mass content alterations.
Collapse
Affiliation(s)
- Rachele Garella
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (F.P.); (R.S.)
| | - Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Flaminia Chellini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Francesco Palmieri
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (F.P.); (R.S.)
| | - Martina Parigi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Daniele Guasti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Emanuele Cassioli
- Psychiatry Unit, Department of Health Sciences, University of Florence, 50134 Florence, Italy; (E.C.); (G.C.); (V.R.)
| | - Giovanni Castellini
- Psychiatry Unit, Department of Health Sciences, University of Florence, 50134 Florence, Italy; (E.C.); (G.C.); (V.R.)
| | - Valdo Ricca
- Psychiatry Unit, Department of Health Sciences, University of Florence, 50134 Florence, Italy; (E.C.); (G.C.); (V.R.)
| | - Daniele Bani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Chiara Sassoli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (F.P.); (R.S.)
| |
Collapse
|
49
|
Dubuisson N, Versele R, Davis-López de Carrizosa MA, Selvais CM, Noel L, Planchon C, Van den Bergh PYK, Brichard SM, Abou-Samra M. The Adiponectin Receptor Agonist, ALY688: A Promising Therapeutic for Fibrosis in the Dystrophic Muscle. Cells 2023; 12:2101. [PMID: 37626911 PMCID: PMC10453606 DOI: 10.3390/cells12162101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is one of the most devastating myopathies, where severe inflammation exacerbates disease progression. Previously, we demonstrated that adiponectin (ApN), a hormone with powerful pleiotropic effects, can efficiently improve the dystrophic phenotype. However, its practical therapeutic application is limited. In this study, we investigated ALY688, a small peptide ApN receptor agonist, as a potential novel treatment for DMD. Four-week-old mdx mice were subcutaneously treated for two months with ALY688 and then compared to untreated mdx and wild-type mice. In vivo and ex vivo tests were performed to assess muscle function and pathophysiology. Additionally, in vitro tests were conducted on human DMD myotubes. Our results showed that ALY688 significantly improved the physical performance of mice and exerted potent anti-inflammatory, anti-oxidative and anti-fibrotic actions on the dystrophic muscle. Additionally, ALY688 hampered myonecrosis, partly mediated by necroptosis, and enhanced the myogenic program. Some of these effects were also recapitulated in human DMD myotubes. ALY688's protective and beneficial properties were mainly mediated by the AMPK-PGC-1α axis, which led to suppression of NF-κβ and TGF-β. Our results demonstrate that an ApN mimic may be a promising and effective therapeutic prospect for a better management of DMD.
Collapse
Affiliation(s)
- Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
- Neuromuscular Reference Center, Department of Neurology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium;
| | - Romain Versele
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| | - Maria A. Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| | - Camille M. Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| | - Chloé Planchon
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| | - Peter Y. K. Van den Bergh
- Neuromuscular Reference Center, Department of Neurology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium;
| | - Sonia M. Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| | - Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research (IREC), Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium; (N.D.); (R.V.); (M.A.D.-L.d.C.); (C.M.S.); (L.N.); (C.P.); (S.M.B.)
| |
Collapse
|
50
|
Ayala-García JC, Díaz-Benítez CE, Lagunas-Martínez A, Orbe-Orihuela YC, Castañeda-Márquez AC, Ortiz-Panozo E, Bermúdez-Morales VH, Cruz M, Burguete-García AI. Mediation Analysis of Waist Circumference in the Association of Gut Microbiota with Insulin Resistance in Children. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1382. [PMID: 37628382 PMCID: PMC10453241 DOI: 10.3390/children10081382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023]
Abstract
BACKGROUND Persistent gut microbiota (GM) imbalance has been associated with metabolic disease development. This study evaluated the mediating role of waist circumference in the association between GM and insulin resistance (IR) in children. METHODS This cross-sectional study included 533 children aged between 6 and 12. The anthropometry, metabolic markers, and relative abundance (RA) of five intestinal bacterial species were measured. Path coefficients were estimated using path analysis to assess direct, indirect (mediated by waist circumference), and total effects on the association between GM and IR. RESULTS The results indicated a positive association mediated by waist circumference between the medium and high RA of S. aureus with homeostatic model assessments for insulin resistance (HOMA-IR) and for insulin resistance adiponectin-corrected (HOMA-AD). We found a negative association mediated by waist circumference between the low and medium RA of A. muciniphila and HOMA-IR and HOMA-AD. Finally, when we evaluated the joint effect of S. aureus, L. casei, and A. muciniphila, we found a waist circumference-mediated negative association with HOMA-IR and HOMA-AD. CONCLUSIONS Waist circumference is a crucial mediator in the association between S. aureus and A. muciniphila RA and changes in HOMA-IR and HOMA-AD scores in children.
Collapse
Affiliation(s)
- Juan Carlos Ayala-García
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (C.E.D.-B.); (A.L.-M.); (Y.C.O.-O.); (V.H.B.-M.)
| | - Cinthya Estefhany Díaz-Benítez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (C.E.D.-B.); (A.L.-M.); (Y.C.O.-O.); (V.H.B.-M.)
| | - Alfredo Lagunas-Martínez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (C.E.D.-B.); (A.L.-M.); (Y.C.O.-O.); (V.H.B.-M.)
| | - Yaneth Citlalli Orbe-Orihuela
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (C.E.D.-B.); (A.L.-M.); (Y.C.O.-O.); (V.H.B.-M.)
| | | | - Eduardo Ortiz-Panozo
- Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico;
| | - Víctor Hugo Bermúdez-Morales
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (C.E.D.-B.); (A.L.-M.); (Y.C.O.-O.); (V.H.B.-M.)
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquímica, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico;
| | - Ana Isabel Burguete-García
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (C.E.D.-B.); (A.L.-M.); (Y.C.O.-O.); (V.H.B.-M.)
| |
Collapse
|