1
|
Zhang Y, Wang A, Zhao W, Qin J, Zhang Y, Liu B, Yao C, Long J, Yuan M, Yan D. Microbial succinate promotes the response to metformin by upregulating secretory immunoglobulin a in intestinal immunity. Gut Microbes 2025; 17:2450871. [PMID: 39812329 DOI: 10.1080/19490976.2025.2450871] [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: 01/07/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Metformin is the first-line pharmacotherapy for type 2 diabetes mellitus; however, many patients respond poorly to this drug in clinical practice. The potential involvement of microbiota-mediated intestinal immunity and related signals in metformin responsiveness has not been previously investigated. In this study, we successfully constructed a humanized mouse model by fecal transplantation of the gut microbiota from clinical metformin-treated - responders and non-responders, and reproduced the difference in clinical phenotypes of responsiveness to metformin. The abundance of Bacteroides thetaiotaomicron, considered a representative differential bacterium of metformin responsiveness, and the level of secretory immunoglobulin A (SIgA) in intestinal immunity increased significantly in responder recipient mice following metformin treatment. In contrast, no significant alterations in B. thetaiotaomicron and SIgA were observed in non-responder recipient mice. The study of IgA-/- mice confirmed that downregulated expression or deficiency of SIgA resulted in non-response to metformin, meaning that metformin was unable to improve dysfunctional glucose metabolism and reduce intestinal and adipose tissue inflammation, ultimately leading to systemic insulin resistance. Furthermore, supplementation with succinate, a microbial product of B. thetaiotaomicron, potentially reversed the non-response to metformin by inducing the production of SIgA. In conclusion, we demonstrated that upregulated SIgA, which could be regulated by succinate, was functionally involved in metformin response through its influence on immune cell-mediated inflammation and insulin resistance. Conversely, an inability to regulate SIgA may result in a lack of response to metformin.
Collapse
Affiliation(s)
- Ying Zhang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Aiting Wang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Zhao
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jia'an Qin
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yu Zhang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bing Liu
- Department of Endocrinology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chengcheng Yao
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jianglan Long
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mingxia Yuan
- Department of Endocrinology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dan Yan
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Aiassa V, Ferreira MDR, Ingaramo P, D'Alessandro ME. Salvia hispanica L. (chia) seed have beneficial effects upon visceral adipose tissues extracellular matrix disorders and inflammation developed in a sucrose-rich diet-induced adiposity rodent model. Mol Cell Endocrinol 2025; 597:112438. [PMID: 39638143 DOI: 10.1016/j.mce.2024.112438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
We have previously demonstrated that dietary Salvia hispanica L. (chia) seed, rich in α-linolenic acid (ALA), was able to reduce visceral adiposity and improves insulin sensitivity in a rodent experimental model of adiposity induced by the administration of a sucrose-rich diet (SRD). The evidence suggests that the pathological expansion of visceral adipose tissue (VAT) is accompanied by changes in the extracellular matrix (ECM) components, which can lead to fibrosis, and/or a greater expression of pro-inflammatory adipokines. The aim of the present work was to evaluate the effect of chia seed administration upon key components and modulators of ECM remodeling and inflammation in different white adipose tissues (WAT) (epididymal-eWAT- and retroperitoneal-rWAT-) in a SRD-induced adiposity rodent model. The results showed that chia seed reduced the increased hydroxyproline levels observed in SRD-fed group and this was accompanied by changes in the activity/expression of matrix metalloproteinases MMP-2 and MMP-9. No changes were observed in transforming growth factor β (TGF-β) expression levels. In addition, this nutritional intervention was able to reduce the levels of PAI-1 and MCP-1, and to increase the levels of adiponectin in both VAT. An increase in the ratio of n-3/n-6 polyunsaturated fatty acids in the membrane phospholipids of both VAT was also observed. The present study demonstrated that chia seed have anti-fibrotic and anti-inflammatory actions in the VAT which could play a key role in the amelioration of visceral adiposity and whole-body insulin insensitivity developed in SRD-fed rats.
Collapse
Affiliation(s)
- Victoria Aiassa
- Laboratorio de Estudio de Enfermedades Metabólicas Relacionadas con la Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - María Del Rosario Ferreira
- Laboratorio de Estudio de Enfermedades Metabólicas Relacionadas con la Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Paola Ingaramo
- Instituto de Salud y Ambiente del Litoral (ISAL- CONICET), Facultad de Bioquímica y Cs. Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - María Eugenia D'Alessandro
- Laboratorio de Estudio de Enfermedades Metabólicas Relacionadas con la Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina.
| |
Collapse
|
3
|
Sakamoto K, Butera MA, Zhou C, Maurizi G, Chen B, Ling L, Shawkat A, Patlolla L, Thakker K, Calle V, Morgan DA, Rahmouni K, Schwartz GJ, Tahiri A, Buettner C. Overnutrition causes insulin resistance and metabolic disorder through increased sympathetic nervous system activity. Cell Metab 2025; 37:121-137.e6. [PMID: 39437790 PMCID: PMC11711004 DOI: 10.1016/j.cmet.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/19/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
The mechanisms underlying obesity-induced insulin resistance remain incompletely understood, as impaired cellular insulin signaling, traditionally considered the primary driver of insulin resistance, does not always accompany impaired insulin action. Overnutrition rapidly increases plasma norepinephrine (NE), suggesting overactivation of the sympathetic nervous system (SNS). However, the role of the SNS in obesity is controversial, as both increased and decreased SNS activity (SNA) have been reported. Here, we show that reducing catecholamine (CA) release from the SNS protects against overnutrition-induced insulin resistance as well as hyperglucagonemia, adipose tissue dysfunction, and fatty liver disease, as we demonstrate utilizing a mouse model of inducible and peripherally restricted deletion of tyrosine hydroxylase (th; THΔper). A key mechanism through which heightened SNA induces insulin resistance is by triggering adipose tissue lipolysis. Increased SNA emerges as a critical driver in the pathogenesis of overnutrition-induced insulin resistance and metabolic disease independent of cellular insulin signaling.
Collapse
Affiliation(s)
- Kenichi Sakamoto
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary A Butera
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chunxue Zhou
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulia Maurizi
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bandy Chen
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Ling
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adham Shawkat
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Likhitha Patlolla
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Kavira Thakker
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Victor Calle
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Gary J Schwartz
- Department of Medicine & Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| | - Azeddine Tahiri
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Christoph Buettner
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Schleh MW, Ryan BJ, Ahn C, Ludzki AC, Van Pelt DW, Pitchford LM, Chugh OK, Luker AT, Luker KE, Samovski D, Abumrad NA, Burant CF, Horowitz JF. Impaired suppression of fatty acid release by insulin is a strong predictor of reduced whole-body insulin-mediated glucose uptake and skeletal muscle insulin receptor activation. Acta Physiol (Oxf) 2025; 241:e14249. [PMID: 39487600 DOI: 10.1111/apha.14249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 11/04/2024]
Abstract
AIM To examine factors underlying why most, but not all, adults with obesity exhibit impaired insulin-mediated glucose uptake, we compared: (1) adipose tissue fatty acid (FA) release, (2) skeletal muscle lipid droplet (LD) characteristics, and (3) insulin signalling events, in skeletal muscle of adults with obesity with relatively high versus low insulin-mediated glucose uptake. METHODS Seventeen adults with obesity (BMI: 36 ± 3 kg/m2) completed a 2 h hyperinsulinemic-euglycemic clamp with stable isotope tracer infusions to measure glucose rate of disappearance (glucose Rd) and FA rate of appearance (FA Ra). Skeletal muscle biopsies were collected at baseline and 30 min into the insulin infusion. Participants were stratified into HIGH (n = 7) and LOW (n = 10) insulin sensitivity cohorts by their glucose Rd during the hyperinsulinemic clamp (LOW< 400; HIGH >550 nmol/kgFFM/min/[μU/mL]). RESULTS Insulin-mediated suppression of FA Ra was lower in LOW compared with HIGH (p < 0.01). In skeletal muscle, total intramyocellular lipid content did not differ between cohorts. However, the size of LDs in the subsarcolemmal region (SS) of type II muscle fibres was larger in LOW compared with HIGH (p = 0.01). Additionally, insulin receptor-β (IRβ) interactions with regulatory proteins CD36 and Fyn were lower in LOW versus HIGH (p < 0.01), which aligned with attenuated insulin-mediated Tyr phosphorylation of IRβ and downstream insulin-signalling proteins in LOW. CONCLUSION Collectively, reduced ability for insulin to suppress FA mobilization, with accompanying modifications in intramyocellular LD size and distribution, and diminished IRβ interaction with key regulatory proteins may be key contributors to impaired insulin-mediated glucose uptake commonly found in adults with obesity.
Collapse
Affiliation(s)
- Michael W Schleh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Benjamin J Ryan
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Cheehoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alison C Ludzki
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Douglas W Van Pelt
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lisa M Pitchford
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Olivia K Chugh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Austin T Luker
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Dmitri Samovski
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nada A Abumrad
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Charles F Burant
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
5
|
Teodoro L, Carreira ACO, Sogayar MC. Exploring the Complexity of Pan-Cancer: Gene Convergences and in silico Analyses. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:913-934. [PMID: 39691553 PMCID: PMC11651076 DOI: 10.2147/bctt.s489246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024]
Abstract
Cancer is a complex and multifaceted group of diseases characterized by highly intricate mechanisms of tumorigenesis and tumor progression, which complicates diagnosis, prognosis, and treatment. In recent years, targeted therapies have gained prominence by focusing on specific mutations and molecular features unique to each tumor type, offering more effective and personalized treatment options. However, it is equally critical to explore the genetic commonalities across different types of cancer, which has led to the rise of pan-cancer studies. These approaches help identify shared therapeutic targets across various tumor types, enabling the development of broader and potentially more widely applicable treatment strategies. This review aims to provide a comprehensive overview of key concepts related to tumors, including tumorigenesis processes, the tumor microenvironment, and the role of extracellular vesicles in tumor biology. Additionally, we explore the molecular interactions and mechanisms driving tumor progression, with a particular focus on the pan-cancer perspective. To achieve this, we conducted an in silico analysis using publicly available datasets, which facilitated the identification of both common and divergent genetic and molecular patterns across different tumor types. By integrating these diverse areas, this review offers a clearer and deeper understanding of the factors influencing tumorigenesis and highlights potential therapeutic targets.
Collapse
Affiliation(s)
- Leandro Teodoro
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, São Paulo, 05508-900, Brazil
| | - Ana Claudia O Carreira
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Center of Human and Natural Sciences, Federal University of ABC, Santo André, São Paulo, 09280-560, Brazil
| | - Mari C Sogayar
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, São Paulo, 05508-900, Brazil
| |
Collapse
|
6
|
Alotaiq N, Khalifa AS, Youssef A, El-Nagar EG, Elwali NE, Habib HM, AlZaim I, Eid AH, Bakkar NMZ, El-Yazbi AF. Targeting GSK-3β for adipose dysfunction and cardiovascular complications of metabolic disease: An entangled WNT/β-catenin question. FASEB J 2024; 38:e70273. [PMID: 39726401 DOI: 10.1096/fj.202402470r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Individuals with metabolic syndrome have a high risk of developing cardiovascular disorders that is closely tied to visceral adipose tissue dysfunction, as well as an altered interaction between adipose tissue and the cardiovascular system. In metabolic syndrome, adipose tissue dysfunction is associated with increased hypertrophy, reduced vascularization, and hypoxia of adipocytes, leading to a pro-oxidative and pro-inflammatory environment. Among the pathways regulating adipose tissue homeostasis is the wingless-type mammary tumor virus integration site family (Wnt) signaling pathway, with both its canonical and non-canonical arms. Various modulators of the Wnt signaling have been identified to contribute to the development of metabolic diseases and their cardiovascular complications, with a particularly significant role played by Glycogen Synthase Kinase-3β (GSK-3β). GSK-3β levels and activities have various and often contrasting roles in obesity and related metabolic disorders, as well as their cardiovascular sequelae. Here, we explore the possibility that altered Wnt signaling and GSK-3β activities could serve as a connection between adipose tissue dysfunction and the development of cardiovascular disease in individuals with metabolic syndrome. We attempt to define a context-specific approach for intervention, which could possibly serve as a novel disease modifying therapy for the mitigation of such complications.
Collapse
Affiliation(s)
- Nasser Alotaiq
- Health Sciences Research Center, Imam Muhammad Ibn Saud Islamic University (IMISIU), Riyadh, Kingdom of Saudi Arabia
| | - Ahmed S Khalifa
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Amr Youssef
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Esraa G El-Nagar
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Nasr Eldin Elwali
- Deanship of Scientific Research, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Kingdom of Saudi Arabia
| | - Hosam M Habib
- Research & Innovation Hub, Alamein International University, Alamein, Egypt
| | - Ibrahim AlZaim
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Ahmed F El-Yazbi
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
- Research & Innovation Hub, Alamein International University, Alamein, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
7
|
Guggeri L, Sosa-Redaelli I, Cárdenas-Rodríguez M, Alonso M, González G, Naya H, Prieto-Echagüe V, Lepanto P, Badano JL. Follistatin like-1 ( Fstl1) regulates adipose tissue development in zebrafish. Adipocyte 2024; 13:2435862. [PMID: 39644214 PMCID: PMC11633180 DOI: 10.1080/21623945.2024.2435862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024] Open
Abstract
Obesity is a highly prevalent disorder with complex aetiology. Therefore, studying its associated cellular and molecular pathways may be aided by analysing genetic tractable diseases. In this context, the study of ciliopathies such as Bardet-Biedl syndrome has highlighted the relevance of primary cilia in obesity, both in the central nervous system and peripheral tissues. Based on our previous in vitro results supporting the role of a novel Bbs4-cilia-Fstl1 axis in adipocyte differentiation, we evaluated the in vivo relevance of the zebrafish orthologous genes fstl1a and fstl1b in primary cilia and adipose tissue development. Using a combination of knockdowns and a new fstl1a mutant line, we show that fstl1a promotes primary cilia formation in early embryos and participates in adipose tissue formation in larvae. We also show that fstl1b partially compensates for the loss of fstl1a. Moreover, in high fat diet, fstl1a depletion affects the expression of differentiation and mature adipocyte markers. These results agree with our previous in vitro data and provide further support for the role of FSTL1 as a regulator of adipose tissue formation. Dissecting the exact biological role of proteins such as FSTL1 will likely contribute to understand obesity onset and presentation.
Collapse
Affiliation(s)
- Lucía Guggeri
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Ileana Sosa-Redaelli
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | | | - Martina Alonso
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Gisell González
- Zebrafish Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Hugo Naya
- Bioinformatics Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Facultad de Agronomía, Universidad de la República, Montevideo, Uruguay
| | | | - Paola Lepanto
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Jose L. Badano
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| |
Collapse
|
8
|
Lysaght J, Conroy MJ. The multifactorial effect of obesity on the effectiveness and outcomes of cancer therapies. Nat Rev Endocrinol 2024; 20:701-714. [PMID: 39313571 DOI: 10.1038/s41574-024-01032-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/19/2024] [Indexed: 09/25/2024]
Abstract
Epidemiology studies have demonstrated a clear association between obesity and the development of several distinct malignancies, with excessive visceral adiposity being an increasingly prevalent feature in patients with cancer presenting for therapeutic intervention. Clinical trials and meta-analyses have helped to inform effective and safe dosing of traditional systemically administered anticancer agents in adult patients with cancer and obesity, but there remains much debate not only regarding the effect of obesity on the more novel targeted molecular and immune-based therapies, but also about how obesity is best defined and measured clinically. Low muscle mass is associated with poor outcomes in cancer, and body composition studies using biochemical and imaging modalities are helping to fully delineate the importance of both obesity and sarcopenia in clinical outcomes; such studies might also go some way to explaining how obesity can paradoxically be associated with favourable clinical outcomes in certain cancers. As the cancer survivorship period increases and the duration of anticancer treatment lengthens, this Review highlights the challenges facing appropriate treatment selection and emphasizes how a multidisciplinary approach is warranted to manage weight and skeletal muscle loss during and after cancer treatment.
Collapse
Affiliation(s)
- Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, School of Medicine, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, Trinity College Dublin, St James's Hospital, Dublin, Ireland.
| | - Melissa J Conroy
- Cancer Immunology Research Group, Department of Anatomy, School of Medicine, Trinity Biomedical Sciences Institute and Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
9
|
Berezina TA, Berezin OO, Lichtenauer M, Berezin AE. Predictors for Irreversibility of Contrast-Induced Acute Kidney Injury in Patients with Obesity After Contrast-Enhanced Computed Tomography Coronary Angiography. Adv Ther 2024:10.1007/s12325-024-03036-z. [PMID: 39527336 DOI: 10.1007/s12325-024-03036-z] [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: 09/07/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Although contrast-induced (CI) acute kidney injury (AKI) is a common complication in high-risk individuals requiring evaluation with contrast-enhanced angiography, the possible predictors of CI-AKI in patients with obesity are not fully understood. The aim of this study was to elucidate plausible factors associated with the irreversibility of CI-AKI in individuals with obesity undergoing contrast-enhanced computed tomography coronary angiography. METHODS A total of 96 adult patients with obesity and the KDIGO criteria of CI-AKI (increase of serum levels of creatinine ≥ 25% or ≥ 500 µmol/L at 48 h after procedure) were retrospectively screened from the cohort of 1833 patients who underwent iodine contrast medium (ICM)-enhanced computed tomography coronary angiography, and were included in the study. The patients were divided into two cohorts: 96 adult patients with obesity and recovery of CI-AKI in 7 days after initiating of the event, and 57 individuals with irreversibility of CI-AKI. Serum concentrations of conventional biochemistry and urine biomarkers [i.e., hemoglobin, creatinine, high-sensitivity C-reactive protein, urinary albumin/creatinine ratio (UACR)] as well as natriuretic peptide, adropin, apelin, irisin, tumor necrosis factor-alpha (TNF-alpha), were determined at baseline. The levels of creatinine were measured at baseline, at the event, and in 7 days after the event. RESULTS We identified 12 variables, which were associated with irreversibility of CI-AKI: age > 75 years [odds ratio (OR) = 1.22. P = 0.001], male gender (OR = 1.03, P = 0.042), stable coronary artery disease (OR = 1.06, P = 0.048), chronic kidney disease (CKD) 1-3 grade (OR = 1.60, P = 0.001), heart failure with preserved ejection fraction (HFpEF) (OR = 1.07, P = 0.046), baseline estimated GFR < 80 mL/min/1.73 m2 (OR = 1.10, P = 0.040), UACR > 17.5 mg/g Cr (OR = 1.05, P = 0.048), TNF-alpha > 3.11 pg/mL (OR = 1.12, P = 0.001), and adropin < 2.43 ng/mL (OR = 1.18, P = 0.001). After adjustment for CKD and UACR > 17.5 mg/g Cr, only HFpEF (OR = 1.06, P = 0.042) and adropin < 2.43 ng/mL (OR = 1.11, P = 0.001) remained independent predictors of CI-AKI irreversibility. Yet, adropin < 2.43 ng/mL at baseline exerted sufficiently better predictive ability than both HFpEF and preexisting CKD 1-3 grade. CONCLUSION In a multivariate prediction model adjusted for CKD and urinary albumin/creatinine ratio > 17.5 mg/g Cr, low levels of adropin (< 2.43 ng/mL) in individuals with non-morbid obesity together with the presence of HFpEF were independent predictors of CI-AKI irreversibility after ICM-enhanced computed tomography coronary angiography.
Collapse
Affiliation(s)
- Tetiana A Berezina
- Department of Internal Medicine and Nephrology, VitaCenter, Zaporozhye, 69000, Ukraine
| | - Oleksandr O Berezin
- Department of Alter Psychiatry, Luzerne Psychiatry AG, 4915 St., Urban, Switzerland
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020, Salzburg, Austria
| | - Alexander E Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020, Salzburg, Austria.
| |
Collapse
|
10
|
Xiang Y, Shen L, Xue Y, Wang Z, Zhou R, Cao Y, Zhu Z, Xu P, Yu X, Fang P, Shang W. Efficacy and safety of diacerein monotherapy in adults with obesity: A randomized, double-blind, placebo-controlled trial. Diabetes Obes Metab 2024; 26:5293-5303. [PMID: 39192530 DOI: 10.1111/dom.15881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024]
Abstract
AIM To assess the efficacy and safety of diacerein monotherapy in adults with obesity. METHODS Forty-two adults with obesity participated in the study and were randomly assigned to receive diacerein or placebo in addition to lifestyle modification for 14 weeks, in a double-blinded fashion. Differences in changes in body weight, body composition, metabolic variables, fatty liver-related indicators, cardiovascular system variables, lifestyle score and metabolic factors were compared. RESULTS Post-treatment weight loss percentage from baseline was -6.56% (-8.71%, -4.41%) in the diacerein group and -0.59% (-2.74%, 1.56%) in the placebo group. Compared with the placebo group, the diacerein group showed significant improvements in body composition, metabolic variables and indicators related to fatty liver. In addition, after 14 weeks of treatment, diacerein led to a significant reduction in serum visfatin concentration versus the placebo group. The reductions in total body fat mass and visceral fat area mediated the weight loss induced by diacerein. No significant differences were found between the groups in the number of adverse events and safety variables. CONCLUSIONS For adults with obesity, diacerein led to a clinically meaningful weight loss and provided multiple metabolic benefits with acceptable safety. These results support that diacerein is a promising candidate medicine to be developed for obesity management.
Collapse
Affiliation(s)
- Yingying Xiang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lixuan Shen
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yingying Xue
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziwei Wang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ruonan Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Cao
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziwei Zhu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Pingyuan Xu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xizhong Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenbin Shang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
11
|
Nogajski Ł, Mazuruk M, Kacperska M, Kurpias M, Mączewski M, Nowakowski M, Mączewski M, Michałowska I, Leszek P, Paterek A. Epicardial fat density obtained with computed tomography imaging - more important than volume? Cardiovasc Diabetol 2024; 23:389. [PMID: 39472958 PMCID: PMC11523889 DOI: 10.1186/s12933-024-02474-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
Epicardial adipose tissue (EAT) is a unique fat depot located between the myocardium and the visceral layer of pericardium. It can be further subdivided into pericoronary (PCAT), periatrial (PAAT) and periventricular adipose tissue (PVentAT), each of them exhibiting specific characteristics and association with the underlying tissue. Since no physical barrier separates EAT from the myocardium, this fat tissue can easily interact with the underlying cardiac structure. EAT can be visualized using various imaging modalities. Computed tomography provides not only information on EAT volume, but also on its density. Indeed, EAT density reflected by the recently developed fat attenuation index (FAI) is emerging as a useful index of PCAT inflammation, PAAT inflammation and fibrosis, while the relevance of density of PVentAT is much less known. The emerging data indicates that FAI can be an important diagnostic and prognostic tool in both coronary artery disease and atrial fibrillation. Future studies will demonstrate if it also could be used as a marker of efficacy of therapies and whether FAI PVentAT could indicate ventricular pathologies, such as heart failure. The aim of the review is to present computed tomography derived FAI as an important tool both to study and better understand the epicardial fat and as a potential predictive marker in cardiovascular disorders.
Collapse
Affiliation(s)
- Łukasz Nogajski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Maciej Mazuruk
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Marta Kacperska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Mikołaj Kurpias
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Maciej Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Maksymilian Nowakowski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Ilona Michałowska
- Department of Radiology, National Institute of Cardiology, Warsaw, Poland
| | - Przemysław Leszek
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland.
| |
Collapse
|
12
|
Hellberg A, Salamon D, Ujvari D, Rydén M, Hirschberg AL. Weight Changes Are Linked to Adipose Tissue Genes in Overweight Women with Polycystic Ovary Syndrome. Int J Mol Sci 2024; 25:11566. [PMID: 39519120 PMCID: PMC11547111 DOI: 10.3390/ijms252111566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Women with polycystic ovary syndrome (PCOS) have varying difficulties in achieving weight loss by lifestyle intervention, which may depend on adipose tissue metabolism. The objective was to study baseline subcutaneous adipose tissue gene expression as a prediction of weight loss by lifestyle intervention in obese/overweight women with PCOS. This is a secondary analysis of a randomized controlled trial where women with PCOS, aged 18-40 and body mass index (BMI) ≥ 27 were initially randomized to either a 4-month behavioral modification program or minimal intervention according to standard care. Baseline subcutaneous adipose tissue gene expression was related to weight change after the lifestyle intervention. A total of 55 obese/overweight women provided subcutaneous adipose samples at study entry. Weight loss was significant after behavioral modification (-2.2%, p = 0.0014), while there was no significant weight loss in the control group (-1.1%, p = 0.12). In microarray analysis of adipose samples, expression of 40 genes differed significantly between subgroups of those with the greatest weight loss or weight gain. 10 genes were involved in metabolic pathways including glutathione metabolism, gluconeogenesis, and pyruvate metabolism. Results were confirmed by real-time polymerase chain reaction (RT-PCR) in all 55 subjects. Expressions of GSTM5, ANLN, and H3C2 correlated with weight change (R = -0.41, p = 0.002; R = -0.31, p = 0.023 and R = -0.32, p = 0.016, respectively). GSTM5, involved in glutathione metabolism, was the strongest predictor of weight loss, and together with baseline waist-hip ratio (WHR) explained 31% of the variation in body weight change. This study shows that baseline subcutaneous adipose tissue genes play a role for body weight outcome in response to lifestyle intervention in overweight/obese women with PCOS.
Collapse
Affiliation(s)
- Anton Hellberg
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
| | - Daniel Salamon
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
| | - Dorina Ujvari
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
- Department of Microbiology, Tumor and Cell Biology, National Pandemic Centre, Centre for Translational Microbiome Research, Karolinska Institutet, 17165 Solna, Sweden
| | - Mikael Rydén
- Department of Medicine Huddinge (H7), Karolinska Institutet, C2-94 Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden;
| | - Angelica Lindén Hirschberg
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital Solna, 17176 Stockholm, Sweden
| |
Collapse
|
13
|
Huang CC, Wang CH, Yeh HY, Tsai HC, Yang CW, Li TH, Su CW, Yang YY, Lin HC, Hou MC. Chronic Peroxisome Proliferator-Activated Receptor α/γ and Cannabinoid Receptor 2 Agonist Treatments Attenuated Visceral Adipose Tissue (VAT)-Derived Extracellular Vesicle-Related VAT and Intestinal Abnormalities in Nonalcoholic Steatohepatitis Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00398-5. [PMID: 39490440 DOI: 10.1016/j.ajpath.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024]
Abstract
This study explores the mechanisms and combined effects of chronic peroxisome proliferator-activated receptor (PPAR)α/γ and cannabinoid receptor 2 (CB2R) agonists on visceral adipose tissue (VAT)-derived extracellular vesicle (EV) release and associated systemic/VAT inflammation, decreased VAT capillary density/fibrosis, and intestinal inflammation/hyperpermeability in nonalcoholic steatohepatitis (NASH) mice. NASH mice received 1 month of PPARα/γ agonist aleglitazar (10 mg/kg per day) or CB2R agonist JWH015 (3 mg/kg per day) alone or combined. High EV release from VAT of NASH mice was associated with severe systemic/VAT/intestinal inflammation, reduced capillary network of VAT, and intestinal hyperpermeability. Combined JWH015 with aleglitazar treatment significantly suppressed high-fat diet-induced obesity/adiposity, inhibited VAT expansion, reduced VAT inflammation/fibrosis, normalized VAT capillary network, and attenuated intestinal mucosal injury, inflammation, and hyperpermeability in NASH + aleglitazar + JWH015 mice. The inhibition of AT-derived EV release and hypoxia-inducible factor (HIF)1α levels in AT-derived EV, normalization of CB2R, PPARα, PPARγ, PPARγ1, PPARγ2, tight junction proteins, vascular endothelial growth factor/CD31 expression, and down-regulation of HIF1α, monocyte chemoattractant protein-1, and transforming growth factor-β1 were observed in the VAT and intestine of the NASH + aleglitazar + jwh015 group. In vitro experiments revealed that PPARα/γ and CB2R activation attenuated NASH AT-derived EV-induced pathogenic changes in the J774/SVEC4-10/Caco2/3T3-L1 cell system. This study suggested that VAT-derived EVs contribute to the pathogenesis of nonalcoholic fatty liver disease and that combined PPARα/γ and CB2R agonist treatment reduces VAT-released EV release and HIF1/monocyte chemoattractant protein-1 signals to ameliorate hepatic steatosis and VAT/intestine abnormalities of NASH mice.
Collapse
Affiliation(s)
- Chia-Chang Huang
- Department of Medical Education, Taipei Veterans General Hospital; Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University
| | | | - Hsiao-Yun Yeh
- Department of Medical Education, Taipei Veterans General Hospital; Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University
| | - Hung-Cheng Tsai
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University; Department of Internal Medicine, Division of Allergy, Immunology, and Rheumatology, Taipei Veterans General Hospital
| | - Ching-Wen Yang
- Department of Medical Education, Taipei Veterans General Hospital
| | - Tzu-Hao Li
- Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Foundation Hosiptal, Taipei City; School of Medicine, College of Medicine, Fu Jen Catholic University
| | - Chien-Wei Su
- Department of Internal Medicine, Division of Allergy, Immunology, and Rheumatology, Taipei Veterans General Hospital
| | - Ying-Ying Yang
- Department of Medical Education, Taipei Veterans General Hospital; Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University.
| | - Han-Chieh Lin
- Department of Internal Medicine, Division of Allergy, Immunology, and Rheumatology, Taipei Veterans General Hospital
| | - Ming-Chih Hou
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University; Department of Internal Medicine, Division of Allergy, Immunology, and Rheumatology, Taipei Veterans General Hospital
| |
Collapse
|
14
|
Nah J, Yun N, Yoo H, Park S, Pae M. Time-Restricted Feeding Attenuates Adipose Tissue Inflammation and Fibrosis in Mice Under Chronic Light Exposure. Int J Mol Sci 2024; 25:11524. [PMID: 39519077 PMCID: PMC11546375 DOI: 10.3390/ijms252111524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Time-restricted feeding (TRF) has emerged as a promising dietary approach for improving metabolic parameters associated with obesity. However, it remains largely unclear whether TRF offers benefits for obesity related to exposure to light at night. This study examined whether lean and obese mice under chronic light exposure could benefit from TRF intervention. Six-week-old C57BL/6 male mice were fed either a low-fat diet or a high-fat diet under a 12 h light/12 h dark cycle for 6 weeks. They were then divided into three subgroups: control light, chronic 24 h light, and chronic light with a daily 10 h TRF. Chronic light exposure led to increased weight gain and higher expression of inflammatory and fibrotic markers in the adipose tissue of both lean and obese mice. It also increased hepatic triglyceride content in mice, regardless of their weight status. TRF protected both lean and obese mice from weight gain, normalized inflammatory and fibrotic gene expression, and reduced adipose tissue collagen and liver triglyceride accumulation caused by light exposure alone or in combination with obesity. These results suggest that TRF could have clinical implications for preventing obesity associated with night shift work, regardless of current weight status.
Collapse
Affiliation(s)
| | | | | | | | - Munkyong Pae
- Department of Food and Nutrition, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju 28644, Republic of Korea; (J.N.); (N.Y.); (H.Y.); (S.P.)
| |
Collapse
|
15
|
Adolf R, Krinke I, Datz J, Cassese S, Kastrati A, Joner M, Schunkert H, Wall W, Hadamitzky M, Engel LC. Specific calcium deposition on pre-procedural CCTA at the time of percutaneous coronary intervention predicts in-stent restenosis in symptomatic patients. J Cardiovasc Comput Tomogr 2024:S1934-5925(24)00444-1. [PMID: 39433461 DOI: 10.1016/j.jcct.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 07/28/2024] [Accepted: 09/26/2024] [Indexed: 10/23/2024]
Abstract
PURPOSE To characterize preprocedural coronary atherosclerotic lesions derived from CCTA and assess their association with in-stent restenosis (ISR) after percutaneous coronary intervention (PCI). MATERIALS AND METHODS This retrospective cohort-study included patients who underwent CCTA for suspected coronary artery disease, subsequent index angiography including PCI and surveillance angiography within 6-8 months after the index procedure. We performed a plaque analysis of culprit lesions on CCTA using a dedicated plaque analysis software including assessment of the surrounding pericoronary fat attenuation index (FAI) and compared findings between lesions with and without ISR at surveillance angiography after stenting. RESULTS Overall 278 coronary lesions in 209 patients were included. Of these lesions, 43 (15.5 %) had ISR at surveillance angiography after stenting while 235 (84.5 %) did not. Likewise, plaque composition such as volume of calcification [129.8 mm3 (83.3-212.6) vs. 94.4 mm3 (60.4-160.5) p = 0.06] and lipid-rich and fibrous plaque volume [38.4 mm3 (19.4-71.2) vs. 38.0 mm3 (14.0-59.1), p = 0.11 and 50.4 mm3 (26.1-77.6) vs. 42.1 mm3 (31.1-60.3), p = 0.16] between lesion with and without ISR were not statistically significant. However lesions associated with ISR were more eccentric (n = 37, 86.0 % versus n = 159, 67,7 %; p = 0.03) and more frequently demonstrated calcified portions on opposite sides on the vessel wall on cross-sectional datasets (n = 24, 55.8 % versus n = 55, 23.4 %, p = 0.001). FAIlesion was significantly different in lesions with ISR as compared to those without ISR [-76.5 (-80.1 to -73.6) vs. -80.9 (-88.9 to -74.0), p = 0.02]. There was no difference with respect to FAIRCA between the two groups [-77.4 (-81.9 to -75.6) vs. -78.5 (-86.0 to -71.0), p = 0.41]. CONCLUSION Coronary lesions associated with ISR at surveillance angiography demonstrated differences in the arrangement of calcified portions as well as an increased lesion-specific pericoronary fat attenuation index at baseline CCTA. This latter finding suggests that perivascular inflammation at baseline may play a major role in the development of in-stent restenosis.
Collapse
Affiliation(s)
- Rafael Adolf
- Institut für Radiologie und Nuklearmedizin, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Insa Krinke
- Institut für Radiologie und Nuklearmedizin, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Janina Datz
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Lazarettstrasse 36, 80636 Munich, Germany; Institute for Computational Mechanics, Technical University of Munich, 85748 Garching b., München, Germany
| | - Salvatore Cassese
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Adnan Kastrati
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Michael Joner
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Heribert Schunkert
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Lazarettstrasse 36, 80636 Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Wolfgang Wall
- Institute for Computational Mechanics, Technical University of Munich, 85748 Garching b., München, Germany
| | - Martin Hadamitzky
- Institut für Radiologie und Nuklearmedizin, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Lazarettstrasse 36, 80636 Munich, Germany
| | - Leif-Christopher Engel
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Klinik an der Technischen Universität München, Lazarettstrasse 36, 80636 Munich, Germany.
| |
Collapse
|
16
|
Oliveira ES, Kattah FM, Lima GC, Horst MA, Figueiredo N, Lima GB, Whitton RGM, de Souza GIDMH, Oyama LM, Silveira EA, Corgosinho FC. Association between Serum Fatty Acids Profile and MetScore in Women with Severe Obesity. Nutrients 2024; 16:3508. [PMID: 39458502 PMCID: PMC11510271 DOI: 10.3390/nu16203508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) is a set of conditions associated with an increased cardiovascular risk. Several serum fatty acids (FAs) seem to play an essential role in the development of cardiometabolic diseases and mortality. Thus, it is imperative to explore the impact of FAs on MetS parameters, using an early MetS screening tool such as MetScore, which is readily available in clinical practice. AIM The aim of this study was to assess the potential correlation between serum FAs and cardiovascular risk using a MetScore. METHODS This cross-sectional study involved 41 women with severe obesity. The MetScore was calculated, and participants were categorized into high- and low-cardiovascular-risk groups based on the median MetScore value. Gas chromatography was used to quantify serum FAs. Generalized Linear Models were used to compare group means. The association was assessed through simple logistic regression, and an adjusted logistic regression was conducted to validate the association between Metscore and serum FAs. RESULTS The high-cardiovascular-risk group exhibited elevated values of HOMA-IR, palmitic, oleic, cis-vaccenic, and monounsaturated fatty acids, as well as the SCD-18C, indicating a heightened cardiovascular risk. Conversely, HDL-c, QUICK, gamma-linolenic, and eicosatetraenoic fatty acids showed lower values compared to the low-risk group. CONCLUSIONS Women with severe obesity and high cardiovascular risk have lower values of some omega-3 and omega-6 FAs, considered cardioprotective and anti-inflammatory, and have higher lipogenic activity and FAs, correlated with high cardiovascular risk. These findings emphasize the need to address lipid metabolism in this population as a therapeutic target to reduce cardiovascular risk. Future research should explore clinical interventions that modulate fatty acid metabolism to mitigate cardiometabolic complications.
Collapse
Affiliation(s)
- Emilly Santos Oliveira
- Postgraduate Program in Nutrition and Health, Federal University of Goiás, Rua 227, Viela Q. 68, Goiânia 74605-080, Brazil; (F.M.K.); (G.C.L.); (M.A.H.); (G.B.L.); (F.C.C.)
| | - Fabiana Martins Kattah
- Postgraduate Program in Nutrition and Health, Federal University of Goiás, Rua 227, Viela Q. 68, Goiânia 74605-080, Brazil; (F.M.K.); (G.C.L.); (M.A.H.); (G.B.L.); (F.C.C.)
| | - Glaucia Carielo Lima
- Postgraduate Program in Nutrition and Health, Federal University of Goiás, Rua 227, Viela Q. 68, Goiânia 74605-080, Brazil; (F.M.K.); (G.C.L.); (M.A.H.); (G.B.L.); (F.C.C.)
| | - Maria Aderuza Horst
- Postgraduate Program in Nutrition and Health, Federal University of Goiás, Rua 227, Viela Q. 68, Goiânia 74605-080, Brazil; (F.M.K.); (G.C.L.); (M.A.H.); (G.B.L.); (F.C.C.)
| | - Nayra Figueiredo
- Postgraduate Program in Health Sciences, Federal University of Goiás, Goiânia 74605-080, Brazil; (N.F.); (E.A.S.)
| | - Gislene Batista Lima
- Postgraduate Program in Nutrition and Health, Federal University of Goiás, Rua 227, Viela Q. 68, Goiânia 74605-080, Brazil; (F.M.K.); (G.C.L.); (M.A.H.); (G.B.L.); (F.C.C.)
| | | | | | - Lila Missae Oyama
- Department of Physiology, UNIFESP—Federal University of São Paulo, São Paulo 18290-000, Brazil; (G.I.d.M.H.d.S.); (L.M.O.)
| | - Erika Aparecida Silveira
- Postgraduate Program in Health Sciences, Federal University of Goiás, Goiânia 74605-080, Brazil; (N.F.); (E.A.S.)
| | - Flávia Campos Corgosinho
- Postgraduate Program in Nutrition and Health, Federal University of Goiás, Rua 227, Viela Q. 68, Goiânia 74605-080, Brazil; (F.M.K.); (G.C.L.); (M.A.H.); (G.B.L.); (F.C.C.)
- Postgraduate Program in Health Sciences, Federal University of Goiás, Goiânia 74605-080, Brazil; (N.F.); (E.A.S.)
| |
Collapse
|
17
|
LaPoint A, Singer JM, Ferguson D, Shew TM, Renkemeyer MK, Palacios HH, Field RL, Yerrathota S, Kumari R, Shankaran M, Smith GI, Yoshino J, He M, Patti GJ, Hellerstein MK, Klein S, Brestoff JR, Morris EM, Finck BN, Lutkewitte AJ. Adipocyte lipin 1 expression associates with human metabolic health and regulates systemic metabolism in mice. J Clin Invest 2024; 134:e169722. [PMID: 39405118 PMCID: PMC11601902 DOI: 10.1172/jci169722] [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: 02/14/2023] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
Dysfunctional adipose tissue is believed to promote the development of hepatic steatosis and systemic insulin resistance, but many of the mechanisms involved are still unclear. Lipin 1 catalyzes the conversion of phosphatidic acid to diacylglycerol (DAG), the penultimate step of triglyceride synthesis, which is essential for lipid storage. Herein we found that adipose tissue LPIN1 expression is decreased in people with obesity compared to lean subjects, and low LPIN1 expression correlated with multi-tissue insulin resistance and increased rates of hepatic de novo lipogenesis. Comprehensive metabolic and multi-omic phenotyping demonstrated that adipocyte-specific Lpin1-/- mice had a metabolically-unhealthy phenotype, including liver and skeletal muscle insulin resistance, hepatic steatosis, increased hepatic de novo lipogenesis, and transcriptomic signatures of metabolically associated steatohepatitis that was exacerbated by high-fat diets. We conclude that adipocyte lipin 1-mediated lipid storage is vital for preserving adipose tissue and systemic metabolic health, and its loss predisposes mice to metabolically associated steatohepatitis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rachael L. Field
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sireesha Yerrathota
- Department of Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Roshan Kumari
- Department of Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mahalakshmi Shankaran
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, USA
| | | | - Jun Yoshino
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Shimane, Japan
- Center for Integrated Kidney Research and Advance (IKRA), Shimane University, Izumo, Shimane, Japan
| | - Mai He
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Gary J. Patti
- Department of Medicine and
- Department of Chemistry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marc K. Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, USA
| | | | - Jonathan R. Brestoff
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - E. Matthew Morris
- Department of Cell Biology and Physiology and
- KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, Kansas, USA
- Center for Children’s Healthy Lifestyles and Nutrition, Kansas City, Missouri, USA
| | | | - Andrew J. Lutkewitte
- Department of Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, Kansas, USA
- KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
18
|
Johnson T, Su J, Andres J, Henning A, Ren J. Sex Differences in Fat Distribution and Muscle Fat Infiltration in the Lower Extremity: A Retrospective Diverse-Ethnicity 7T MRI Study in a Research Institute Setting in the USA. Diagnostics (Basel) 2024; 14:2260. [PMID: 39451583 PMCID: PMC11506611 DOI: 10.3390/diagnostics14202260] [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: 07/15/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
Background: Fat infiltration in skeletal muscle is related to declining muscle strength, whereas excess subcutaneous fat is implicated in the development of metabolic diseases. Methods: Using multi-slice axial T2-weighted (T2w) MR images, this retrospective study characterized muscle fat infiltration (MFI) and fat distribution in the lower extremity of 107 subjects (64M/43F, age 11-79 years) with diverse ethnicities (including White, Black, Latino, and Asian subjects). Results: MRI data analysis shows that MFI, evaluated by the relative intensities of the pixel histogram profile in the calf muscle, tends to increase with both age and BMI. However, statistical significance was found only for the age correlation in women (p < 0.002), and the BMI correlation in men (p = 0.04). Sex disparities were also seen in the fat distribution, which was assessed according to subcutaneous fat thickness (SFT) and the fibula bone marrow cross-sectional area (BMA). SFT tends to decrease with age in men (p < 0.01), whereas SFT tends to increase with BMI only in women (p < 0.01). In contrast, BMA tends to increase with age in women (p < 0.01) and with BMI in men (p = 0.04). Additionally, MFI is positively correlated with BMA but not with SFT, suggesting that compromised bone structure may contribute to fat infiltration in the surrounding skeletal muscle. Conclusions: The findings of this study highlight a sex factor affecting MFI and fat distribution, which may offer valuable insights into effective strategies to prevent and treat MFI in women versus men.
Collapse
Affiliation(s)
- Talon Johnson
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Department of Mathematics, University of Arlington, Arlington, TX 76019, USA;
| | - Jianzhong Su
- Department of Mathematics, University of Arlington, Arlington, TX 76019, USA;
| | - Johnathan Andres
- Department of Mathematics, University of Houston, Houston, TX 77004, USA;
| | - Anke Henning
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jimin Ren
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
19
|
Rahimi Kahmini A, Valera IC, Crawford RQ, Samarah L, Reis G, Elsheikh S, Kanashiro-Takeuchi RM, Mohammadipoor N, Olateju BS, Matthews AR, Parvatiyar MS. Aging reveals a sex-dependent susceptibility of sarcospan-deficient mice to cardiometabolic disease. Am J Physiol Heart Circ Physiol 2024; 327:H1067-H1085. [PMID: 39120469 PMCID: PMC11482229 DOI: 10.1152/ajpheart.00702.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Numerous genes including sarcospan (SSPN) have been designated as obesity-susceptibility genes by human genome-wide association studies. Variants in the SSPN locus have been linked with sex-dependent obesity-associated traits; however, this association has not been investigated in vivo. To delineate the role SSPN plays in regulating metabolism with potential to impact cardiac function, we subjected young and aged global SSPN-deficient (SSPN-/-) male and female mice to obesogenic conditions (60% fat diet). We hypothesized that loss of SSPN combined with metabolic stress would increase susceptibility of mice to cardiometabolic disease. Baseline and end-point assessments of several anthropometric parameters were performed including weight, glucose tolerance, and fat distribution of mice fed control (CD) and high-fat (HFD) diet. Doppler echocardiography was used to monitor cardiac function. White adipose and cardiac tissues were assessed for inflammation by histological, gene expression, and cytokine analysis. Overall, SSPN deficiency protected both sexes and ages from diet-induced obesity, with a greater effect in females. SSPN-/- HFD mice gained less weight than wild-type (WT) cohorts, while SSPN-/- CD groups increased weight. Furthermore, aged SSPN-/- mice developed glucose intolerance regardless of diet. Echocardiography showed preserved systolic function for all groups; however, aged SSPN-/- males exhibited significant increases in left ventricular mass (CD) and signs of diastolic dysfunction (HFD). Cytokine analysis revealed significantly increased IL-1α and IL-17Α in white adipose tissue from young SSPN-/- male mice, which may be protective from diet-induced obesity. Overall, these studies suggest that several sex-dependent mechanisms influence the role SSPN plays in metabolic responses that become evident with age.NEW & NOTEWORTHY Young and aged sarcospan (SSPN)-deficient mice were examined to assess the role of SSPN in obesity and cardiometabolic disease. Both sexes displayed a "leaner" phenotype in response to high-fat diet (HFD). Notably, several sex differences were identified in aged SSPN-deficient mice: 1) females developed glucose intolerance (control and HFD) and 2) males exhibited increased left ventricular mass (control) and diastolic dysfunction (HFD). Therefore, we conclude that SSPN exerts a sex-dependent influence on obesity-associated diseases.
Collapse
Affiliation(s)
- Aida Rahimi Kahmini
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, United States
| | - Isela C Valera
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, United States
| | - Rhiannon Q Crawford
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, United States
| | - Luaye Samarah
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, United States
| | - Gisienne Reis
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, United States
| | - Salma Elsheikh
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, United States
| | - Rosemeire M Kanashiro-Takeuchi
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Nazanin Mohammadipoor
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, United States
| | - Bolade S Olateju
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, United States
| | - Aaron R Matthews
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, United States
| | - Michelle S Parvatiyar
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
20
|
Mo YY, Han YX, Xu SN, Jiang HL, Wu HX, Cai JM, Li L, Bu YH, Xiao F, Liang HD, Wen Y, Liu YZ, Yin YL, Zhou HD. Adipose Tissue Plasticity: A Comprehensive Definition and Multidimensional Insight. Biomolecules 2024; 14:1223. [PMID: 39456156 PMCID: PMC11505740 DOI: 10.3390/biom14101223] [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: 08/18/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Adipose tissue is composed of adipocytes, stromal vascular fraction, nerves, surrounding immune cells, and the extracellular matrix. Under various physiological or pathological conditions, adipose tissue shifts cellular composition, lipid storage, and organelle dynamics to respond to the stress; this remodeling is called "adipose tissue plasticity". Adipose tissue plasticity includes changes in the size, species, number, lipid storage capacity, and differentiation function of adipocytes, as well as alterations in the distribution and cellular composition of adipose tissue. This plasticity has a major role in growth, obesity, organismal protection, and internal environmental homeostasis. Moreover, certain thresholds exist for this plasticity with significant individualized differences. Here, we comprehensively elaborate on the specific connotation of adipose tissue plasticity and the relationship between this plasticity and the development of many diseases. Meanwhile, we summarize possible strategies for treating obesity in response to adipose tissue plasticity, intending to provide new insights into the dynamic changes in adipose tissue and contribute new ideas to relevant clinical problems.
Collapse
Affiliation(s)
- Yu-Yao Mo
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Xin Han
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Shi-Na Xu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hong-Li Jiang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hui-Xuan Wu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Jun-Min Cai
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Long Li
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yan-Hong Bu
- Department of Blood Transfusion, The Second Xiangya Hospital, Central South University, Changsha 410012, China;
| | - Fen Xiao
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Han-Dan Liang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Ying Wen
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Ze Liu
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China;
| | - Yu-Long Yin
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| |
Collapse
|
21
|
Ahn C, Divoux A, Zhou M, Seldin MM, Sparks LM, Whytock KL. An optimized pipeline for high-throughput bulk RNA-Seq deconvolution illustrates the impact of obesity and weight loss on cell composition of human adipose tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614489. [PMID: 39386599 PMCID: PMC11463495 DOI: 10.1101/2024.09.23.614489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Cellular heterogeneity of human adipose tissue, is linked to the pathophysiology of obesity and may impact the response to energy restriction and changes in fat mass. Here, we provide an optimized pipeline to estimate cellular composition in human abdominal subcutaneous adipose tissue (ASAT) from publicly available bulk RNA-Seq using signature profiles from our previously published full-length single nuclei (sn)RNA-Seq of the same depot. Individuals with obesity had greater proportions of macrophages and lower proportions of adipocyte sub-populations and vascular cells compared with lean individuals. Two months of diet-induced weight loss (DIWL) increased the estimated proportions of macrophages; however, two years of DIWL reduced the estimated proportions of macrophages, thereby suggesting a bi-phasic nature of cellular remodeling of ASAT during weight loss. Our optimized high-throughput pipeline facilitates the assessment of composition changes of highly characterized cell types in large numbers of ASAT samples using low-cost bulk RNA-Seq. Our data reveal novel changes in cellular heterogeneity and its association with cardiometabolic health in humans with obesity and following weight loss.
Collapse
Affiliation(s)
- Cheehoon Ahn
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Adeline Divoux
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Mingqi Zhou
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA
| | - Marcus M Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| |
Collapse
|
22
|
Odimegwu CL, Uwaezuoke SN, Chikani UN, Mbanefo NR, Adiele KD, Nwolisa CE, Eneh CI, Ndiokwelu CO, Okpala SC, Ogbuka FN, Odo KE, Ohuche IO, Obiora-Izuka CE. Targeting the Epigenetic Marks in Type 2 Diabetes Mellitus: Will Epigenetic Therapy Be a Valuable Adjunct to Pharmacotherapy? Diabetes Metab Syndr Obes 2024; 17:3557-3576. [PMID: 39323929 PMCID: PMC11423826 DOI: 10.2147/dmso.s479077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/03/2024] [Indexed: 09/27/2024] Open
Abstract
Although genetic, environmental, and lifestyle factors largely contribute to type 2 diabetes mellitus (T2DM) risk, the role of epigenetics in its pathogenesis is now well established. The epigenetic mechanisms in T2DM mainly consist of DNA methylation, histone modifications and regulation by noncoding RNAs (ncRNAs). For instance, DNA methylation at CpG islands in the promoter regions of specific genes encoding insulin signaling and glucose metabolism suppresses these genes. Modulating the enzyme mediators of these epigenetic marks aims to restore standard gene expression patterns and improve glycemic control. In targeting these epigenetic marks, using epigenetic drugs such as DNA methyltransferase (DNAMT), histone deacetylase (HDAC) and histone acetyltransferase (HAT) inhibitors has led to variable success in humans and experimental murine models. Specifically, the United States' Food and Drug Administration (US FDA) has approved DNAMT inhibitors like 5-azacytidine and 5-aza-2'-deoxycytidine for use in diabetic retinopathy: a T2DM microvascular complication. These DNAMT inhibitors block the genes for methylation of mitochondrial superoxide dismutase 2 (SOD2) and matrix metallopeptidase 9 (MMP-9): the epigenetic marks in diabetic retinopathy. Traditional pharmacotherapy with metformin also have epigenetic effects in T2DM and positively alter disease outcomes when combined with epigenetic drugs like DNAMT and HDAC inhibitors, raising the prospect of using epigenetic therapy as a valuable adjunct to pharmacotherapy. However, introducing small interfering RNAs (siRNAs) in cells to silence specific target genes remains in the exploratory phase. Future research should focus on regulating gene expression in T2DM using long noncoding RNA (lncRNA) molecules, another type of ncRNA. This review discusses the epigenetics of T2DM and that of its macro- and microvascular complications, and the potential benefits of combining epigenetic therapy with pharmacotherapy for optimal results.
Collapse
Affiliation(s)
- Chioma Laura Odimegwu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Samuel Nkachukwu Uwaezuoke
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ugo N Chikani
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ngozi Rita Mbanefo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ken Daberechi Adiele
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | - Chizoma Ihuarula Eneh
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Chibuzo Obiora Ndiokwelu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Somkenechi C Okpala
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Francis N Ogbuka
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Kenneth E Odo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | | |
Collapse
|
23
|
Masood A, Benabdelkamel H, Joy SS, Alhossan A, Alsuwayni B, Abdeen G, Aldhwayan M, Alfadda NA, Miras AD, Alfadda AA. Label-free quantitative proteomic profiling reveals differential plasma protein expression in patients with obesity after treatment with liraglutide. Front Mol Biosci 2024; 11:1458675. [PMID: 39324112 PMCID: PMC11422103 DOI: 10.3389/fmolb.2024.1458675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/12/2024] [Indexed: 09/27/2024] Open
Abstract
Introduction Treatment and management of obesity is clinically challenging. The inclusion of GLP-1 receptor agonists (GLP1RA) in the medical management of obesity has proven to be efficacious. However, mechanisms underlying the molecular changes arising from GLP1RA treatment in patients with obesity remain to be elucidated. Methods A single-center, prospective study was undertaken to evaluate the changes in the plasma proteins after liraglutide 3 mg therapy in twenty patients (M/F: 7/13) with obesity (mean BMI 40.65 ± 3.7 kg/m2). Anthropometric and laboratory parameters were measured, and blood samples were collected at two time points: baseline, before initiating treatment (pretreatment group, PT), and after three months of receiving the full dose liraglutide 3 mg (posttreatment group, PoT). An untargeted label-free LC MSMS mass spectrometric approach combined with bioinformatics and network pathway analysis was used to determine changes in the proteomic profiles. Results The mean age of the study participants was 36.0 ± 11.1 years. A statistically significant change was observed in weight, BMI and HbA1c levels between the PT and PoT groups (paired t-test, P < 0.001). A significant dysregulation was noted in the abundances of 151 proteins (31 up and 120 downregulated) between the two groups. The potential biomarkers were evaluated using receiver operating characteristic (ROC) curves. The top ten proteins (area under the curve (AUC) of 0.999 (95% CI)) were identified as potential biomarkers between PT and PoT groups and included Cystatin-B, major vault protein, and plastin-3, which were upregulated, whereas multimerin-2, large ribosomal P2, and proline-rich acidic protein 1 were downregulated in the PoT group compared with the PT group. The top network pathway identified using ingenuity pathway analysis (IPA), centered around dysregulation of MAPK, AKT, and PKc signaling pathways and related to cell-to-cell signaling and interaction, cellular assembly and organization, cellular compromise and a score of 46 with 25 focus proteins. Discussion Through label-free quantitative proteomic analysis, our study revealed significant dysregulation of plasma proteins after liraglutide 3 mg treatment in patients with obesity. The alterations in the proteomic profile between the PT and PoT groups demonstrated a decrease in levels of proteins involved in inflammation and oxidative stress pathways. On the other hand proteins involved in the glycolytic and lipolytic metabolic pathways as well as those participating in cytoskeletal and endothelial reorganization were observed to be increased. Understanding actions of liraglutide at a molecular and proteomic levels provides a holistic look into how liraglutide impacts metabolism, induces weight loss and improves overall metabolic health.
Collapse
Affiliation(s)
- Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Salini Scaria Joy
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdulaziz Alhossan
- Corporate of Pharmacy Services, King Saud University Medical City, Riyadh, Saudi Arabia
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Bashayr Alsuwayni
- Corporate of Pharmacy Services, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Ghalia Abdeen
- Department of Community Health Sciences, Clinical Nutrition, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Madhawi Aldhwayan
- Department of Community Health Sciences, Clinical Nutrition, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Nora A. Alfadda
- Department of Community Health Sciences, Clinical Nutrition, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Alexander Dimitri Miras
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolic Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
- School of Medicine, Ulster University, Derry, United Kingdom
| | - Assim A. Alfadda
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
24
|
Choisez A, Ishihara S, Ishii T, Xu Y, Firouzjah SD, Haga H, Nagatomi R, Kusuyama J. Matrix stiffness regulates the triad communication of adipocytes/macrophages/endothelial cells through CXCL13. J Lipid Res 2024; 65:100620. [PMID: 39151591 PMCID: PMC11406362 DOI: 10.1016/j.jlr.2024.100620] [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: 12/06/2023] [Revised: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 08/19/2024] Open
Abstract
Adipose tissue remodeling and plasticity are dynamically regulated by the coordinated functions of adipocytes, macrophages, and endothelial cells and extracellular matrix (ECM) that provides stiffness networks in adipose tissue component cells. Inflammation and fibrosis are crucial exogenous factors that dysregulate adipose tissue functions and drastically change the mechanical properties of the ECM. Therefore, communication among the ECM and adipose tissue component cells is necessary to understand the multifaceted functions of adipose tissues. To obtain in vivo stiffness, we used genipin as a crosslinker for collagen gels. Meanwhile, we isolated primary adipocytes, macrophages, and endothelial cells from C57BL/6J mice and incubated these cells in the differentiation media on temperature-responsive culture dishes. After the differentiation, these cell sheets were transferred onto genipin-crosslinked collagen gels with varying matrix stiffness. We found that inflammatory gene expressions were induced by hard matrix, whereas antiinflammatory gene expressions were promoted by soft matrix in all three types of cells. Interestingly, the coculture experiments of adipocytes, macrophages, and endothelial cells showed that the effects of soft or hard matrix stiffness stimulation on adipocytes were transmitted to the distant adipose tissue component cells, altering their gene expression profiles under normal matrix conditions. Finally, we identified that a hard matrix induces the secretion of CXCL13 from adipocytes, and CXCL13 is one of the important transmitters for stiffness communication with macrophages and endothelial cells. These findings provide insight into the mechanotransmission into distant cells and the application of stiffness control for chronic inflammation in adipose tissues with metabolic dysregulation.
Collapse
Affiliation(s)
- Arthur Choisez
- Department of Biosignals and Inheritance, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Division of Biomedical Engineering for Health and Welfare, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Seiichiro Ishihara
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Takuro Ishii
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | - Yidan Xu
- Department of Medicine and Science in Sports and Exercise, Tohoku University School of Medicine, Sendai, Japan
| | - Sepideh D Firouzjah
- Department of Medicine and Science in Sports and Exercise, Tohoku University School of Medicine, Sendai, Japan
| | - Hisashi Haga
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Ryoichi Nagatomi
- Department of Biosignals and Inheritance, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | - Joji Kusuyama
- Department of Biosignals and Inheritance, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Division of Biomedical Engineering for Health and Welfare, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan; Department of Medicine and Science in Sports and Exercise, Tohoku University School of Medicine, Sendai, Japan.
| |
Collapse
|
25
|
Fitch AK, Malhotra S, Conroy R. Differentiating monogenic and syndromic obesities from polygenic obesity: Assessment, diagnosis, and management. OBESITY PILLARS 2024; 11:100110. [PMID: 38766314 PMCID: PMC11101890 DOI: 10.1016/j.obpill.2024.100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024]
Abstract
Background Obesity is a multifactorial neurohormonal disease that results from dysfunction within energy regulation pathways and is associated with increased morbidity, mortality, and reduced quality of life. The most common form is polygenic obesity, which results from interactions between multiple gene variants and environmental factors. Highly penetrant monogenic and syndromic obesities result from rare genetic variants with minimal environmental influence and can be differentiated from polygenic obesity depending on key symptoms, including hyperphagia; early-onset, severe obesity; and suboptimal responses to nontargeted therapies. Timely diagnosis of monogenic or syndromic obesity is critical to inform management strategies and reduce disease burden. We outline the physiology of weight regulation, role of genetics in obesity, and differentiating characteristics between polygenic and rare genetic obesity to facilitate diagnosis and transition toward targeted therapies. Methods In this narrative review, we focused on case reports, case studies, and natural history studies of patients with monogenic and syndromic obesities and clinical trials examining the efficacy, safety, and quality of life impact of nontargeted and targeted therapies in these populations. We also provide comprehensive algorithms for diagnosis of patients with suspected rare genetic causes of obesity. Results Patients with monogenic and syndromic obesities commonly present with hyperphagia (ie, pathologic, insatiable hunger) and early-onset, severe obesity, and the presence of hallmark characteristics can inform genetic testing and diagnostic approach. Following diagnosis, specialized care teams can address complex symptoms, and hyperphagia is managed behaviorally. Various pharmacotherapies show promise in these patient populations, including setmelanotide and glucagon-like peptide-1 receptor agonists. Conclusion Understanding the pathophysiology and differentiating characteristics of monogenic and syndromic obesities can facilitate diagnosis and management and has led to development of targeted pharmacotherapies with demonstrated efficacy for reducing body weight and hunger in the affected populations.
Collapse
Affiliation(s)
| | - Sonali Malhotra
- Harvard Medical School, Boston, MA, USA
- Rhythm Pharmaceuticals, Inc., Boston, MA, USA
- Massachussetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
26
|
Sancar G, Birkenfeld AL. The role of adipose tissue dysfunction in hepatic insulin resistance and T2D. J Endocrinol 2024; 262:e240115. [PMID: 38967989 PMCID: PMC11378142 DOI: 10.1530/joe-24-0115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 07/05/2024] [Indexed: 07/07/2024]
Abstract
The root cause of type 2 diabetes (T2D) is insulin resistance (IR), defined by the failure of cells to respond to circulating insulin to maintain lipid and glucose homeostasis. While the causes of whole-body insulin resistance are multifactorial, a major contributing factor is dysregulation of liver and adipose tissue function. Adipose dysfunction, particularly adipose tissue-IR (adipo-IR), plays a crucial role in the development of hepatic insulin resistance and the progression of metabolic dysfunction-associated steatotic liver disease (MASLD) in the context of T2D. In this review, we will focus on molecular mechanisms of hepatic insulin resistance and its association with adipose tissue function. A deeper understanding of the pathophysiological mechanisms of the transition from a healthy state to insulin resistance, impaired glucose tolerance, and T2D may enable us to prevent and intervene in the progression to T2D.
Collapse
Affiliation(s)
- Gencer Sancar
- German Center for Diabetes Research, Neuherberg, Germany
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- German Center for Diabetes Research, Neuherberg, Germany
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
27
|
Long-term exercise training has positive effects on adipose tissue in overweight or obesity. Nat Metab 2024; 6:1657-1658. [PMID: 39256591 DOI: 10.1038/s42255-024-01102-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
|
28
|
Moqaddam MA, Nemati M, Dara MM, Hoteit M, Sadek Z, Ramezani A, Rand MK, Abbassi-Daloii A, Pashaei Z, Almaqhawi A, Razi O, Escobar KA, Supriya R, Saeidi A, Zouhal H. Exploring the Impact of Astaxanthin Supplementation in Conjunction with a 12-Week CrossFit Training Regimen on Selected Adipo-Myokines Levels in Obese Males. Nutrients 2024; 16:2857. [PMID: 39275173 PMCID: PMC11397083 DOI: 10.3390/nu16172857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/22/2024] [Accepted: 08/14/2024] [Indexed: 09/16/2024] Open
Abstract
OBJECTIVE Obesity is associated with an exacerbated metabolic condition that is mediated through impairing balance in the secretion of some adipo-myokines. Therefore, the objective of the present study was to explore the impact of astaxanthin supplementation in conjunction with a 12-week CrossFit training regimen on some selected adipo-myokines, insulin insensitivity, and serum lipid levels in obese males. MATERIAL AND METHODS This study is a randomized control trial design; 60 obese males were randomly divided into four groups of 15, including the control group (CG), supplement group (SG), training group (TG), and combined training and supplement group (TSG). The participants were subjected to 12 weeks of astaxanthin (AST) supplementation [20 mg/d capsule, once/d] or CrossFit training or a combination of both interventions. The training regimen comprised 36 sessions of CrossFit, each lasting 60 min, conducted three times per week. The metabolic indices, body composition, anthropometrical, cardio-respiratory, and also some plasma adipo-myokine factors, including decorin (DCN), activin A, myostatin (MST), transforming growth factor (TGF)-β1, and follistatin (FST), were examined 12 and 72 h before the initiation of the main interventional protocols, and then 72 h after the final session of the training protocol. RESULTS There was no significant difference in the baseline data between the groups (p > 0.05). There were significant interactions between group x time for DCN (η2 = 0.82), activin A (η2 = 0.50), FST (η2 = 0.92), MST (η2 = 0.75), and TGFB-1 (η2 = 0.67) (p < 0.001 for all the variables). Significantly changes showed for DCN in TSG compared to TG and SG and also TG compared to SG (p = 0.0001); for activin A in SG compared to TG (p = 0.01) and TSG (p = 0.002); for FST in SG compared to TG and TSG (p = 0.0001), also in TSG compared to TG (p = 0.0001); for MST in SG, TG, and TSG compared to CG (p = 0.0001) and also in TSG compared to SG (p = 0.0001) and TG (p = 0.001); for TGFB-1 in SG, TG, and TSG compared to CG (p = 0.0001) and also TSG compared to SG (p = 0.0001) and TG (p = 0.001). CONCLUSIONS The 12-week CrossFit training concurrent with AST supplementation reduced anthropometric and metabolic factors and also serum lipid levels while producing positive changes in body composition and cardiovascular factors. Increased FST and DCN and reduced activin A, MST, and TGF-β1 were other affirmative responses to both interventions.
Collapse
Affiliation(s)
- Mohammad Ahmadi Moqaddam
- Department of Physical Education and Sport Science, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Morteza Nemati
- Department of Biomechanics and Sports Injuries, Faculty of Physical Education and Sports Sciences, Kharazmi University, Tehran 1571914911, Iran
| | - Marjan Mansouri Dara
- Department of Physical Education and Sport Science, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Maha Hoteit
- Food Science Unit, National Council for Scientific Research of Lebanon (CNRS-L), Beirut 11-8281, Lebanon
- Section 1, Faculty of Public Health, Lebanese University, Beirut 6573, Lebanon
| | - Zahra Sadek
- Section 1, Faculty of Public Health, Lebanese University, Beirut 6573, Lebanon
- Laboratory of Motor System, Handicap and Rehabilitation (MOHAR), Faculty of Public Health, Lebanese University, Beirut 6573, Lebanon
| | - Akbar Ramezani
- Ayatollah Amoli Branch, Department of Exercise Physiology, Islamic Azad University, Amol 6134937333, Iran
| | - Mahboubeh Khak Rand
- Ayatollah Amoli Branch, Department of Exercise Physiology, Islamic Azad University, Amol 6134937333, Iran
| | - Asieh Abbassi-Daloii
- Ayatollah Amoli Branch, Department of Exercise Physiology, Islamic Azad University, Amol 6134937333, Iran
| | - Zhaleh Pashaei
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, University of Tabriz, Tabriz 5166616471, Iran
| | - Abdullah Almaqhawi
- Department of Family Medicine and Community, College of Medicine, King Faisal University, Al Ahsa 31982, Saudi Arabia
| | - Omid Razi
- Department of Exercise Physiology, Faculty of Physical Education and Sports Science, Razi University, Kermanshah 6714414971, Iran
| | - Kurt A Escobar
- Department of Kinesiology, California State University, Long Beach, CA 90840, USA
| | - Rashmi Supriya
- Centre for Health and Exercise Science Research, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
- Academy of Wellness and Human Development, Faculty of Arts and Social Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
| | - Ayoub Saeidi
- Department of Physical Education and Sport Sciences, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj 1517566177, Iran
| | - Hassane Zouhal
- M2S (Laboratoire Mouvement, Sport, Santé)-EA 1274, Université Rennes, 35044 Rennes, France
- Institut International des Sciences du Sport (2I2S), 35850 Irodouer, France
| |
Collapse
|
29
|
Zhan W, Luo H, Feng J, Li R, Yang Y. Diagnosis of perimenopausal coronary heart disease patients using radiomics signature of pericoronary adipose tissue based on coronary computed tomography angiography. Sci Rep 2024; 14:19643. [PMID: 39179762 PMCID: PMC11344045 DOI: 10.1038/s41598-024-70218-8] [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: 05/22/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024] Open
Abstract
To assess whether the radiomics signature of pericoronary adipose tissue (PCAT) from coronary computed tomography angiography (CCTA) can distinguish between perimenopausal women with coronary heart disease (CHD) and those without coronary artery disease (CAD). This single-center retrospective case-control study comprised 140 perimenopausal women with CHD presenting with chest pain who underwent CCTA within 48 h of admission. They were matched with 140 control patients presenting with chest pain but without CAD, based on age, risk factors, radiation dose and CT tube voltage. For all participants, PCAT around the proximal right coronary artery was segmented, from which radiomics features and the fat attenuation index (FAI) were extracted and analyzed. Subsequently, corresponding models were developed and internally validated using Bootstrap methods. Model performance was assessed through measures of identification, calibration, and clinical utility. Using logistic regression analysis, an integrated model that combines clinical features, fat attenuation index and radiomics parameters demonstrated enhanced discrimination ability for perimenopausal CHD (area under the curve [AUC]: 0.80, 95% confidence interval [CI]:0.740-0.845). This model outperformed both the combination of clinical features and PCAT attenuation (AUC 0.67, 95% CI 0.602-0.727) and the use of clinical features alone (AUC 0.66, 95% CI 0.603-0.732). Calibration curves for the three predictive models indicated satisfactory fit (all p > 0.05). Moreover, decision curve analysis demonstrated that the integrated model offered greater clinical benefit compared to the other two models. The CCTA-based radiomics signature derived from the PCAT model outperforms the FAI model in differentiating perimenopausal CHD patients from non-CAD individuals. Integrating PCAT radiomics with the FAI could enhance the diagnostic accuracy for perimenopausal CHD.
Collapse
Affiliation(s)
- Weisheng Zhan
- Department of Cardiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hui Luo
- Department of Thoracic Surgery, Nanchong Central Hospital, Nanchong, China
| | - Jie Feng
- Department of Cardiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Rui Li
- Department of Cardiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China.
| | - Ying Yang
- Department of Cardiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China.
| |
Collapse
|
30
|
Wang C, Wang Y, Zhu W, Tang Q, Wang X, Zhang L. The involvement of epidural fat in ossification of the ligamentum flavum: From the perspective of exosomal proteome. Heliyon 2024; 10:e34755. [PMID: 39144971 PMCID: PMC11320449 DOI: 10.1016/j.heliyon.2024.e34755] [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: 02/05/2024] [Revised: 06/18/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024] Open
Abstract
Ossification of the ligamentum flavum (OLF) is the primary etiology of thoracic spinal stenosis. The functional properties of epidural fat (EF), an adipose tissue located in close proximity to ligamentum flavum (LF), have been scarcely investigated. The metabolic state of adipocytes significantly influences their functionality, and exosomes play a pivotal role in intercellular communication. This study aimed to investigate the role of EF-derived exosomes in OLF and characterize their protein profile by proteomics analysis. Our findings demonstrate that exosomes obtained from EF adjacent to OLF possess the ability to enhance osteogenesis of fibroblasts in vitro. Furthermore, proteomics analysis revealed metabolic dysfunction in EF adipocytes and identified lactate dehydrogenase A (LDHA) as a potential mediator involved in the development of OLF. This study provides new insights into the pathogenic mechanism underlying OLF and offers a theoretical basis for preventing and treating ligament ossification.
Collapse
Affiliation(s)
- Chao Wang
- Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yida Wang
- Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Weihang Zhu
- Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Tang
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuekang Wang
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
31
|
Wang J, Apizi A, Qiu H, Tao N, An H. Association between metabolic obesity phenotypes and the risk of developing prostate cancer: a propensity score matching study based on Xinjiang. Front Endocrinol (Lausanne) 2024; 15:1442740. [PMID: 39165513 PMCID: PMC11333236 DOI: 10.3389/fendo.2024.1442740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024] Open
Abstract
Background Obesity-induced metabolic dysfunction increases the risk of developing tumors, however, the relationship between metabolic obesity phenotypes and prostate cancer (PCa) remains unclear. Methods The term metabolic obesity phenotypes was introduced based on metabolic status and BMI categories. Participants were categorized into four groups: metabolically healthy nonobesity (MHNO), metabolically healthy obesity (MHO), metabolically unhealthy nonobesity (MUNO), and metabolically unhealthy obesity (MUO). Propensity score matching was conducted based on age, ethnicity, marriage, etc. Univariate and multivariate conditional logistic regression analyses were used to assess the relationship between metabolic obesity phenotypes, metabolic risk factors, and PCa. Sensitivity analysis was performed to verify the robustness of the results. Results After propensity score matching among 564 PCa patients and 1418 healthy individuals, 209 were selected for each of the case and control groups. There were no statistically significant differences in the basic characteristics between the two groups. Univariate and multivariate conditional logistic regression suggested that the risk of developing PCa in both MHO and MUO individuals was higher than in MHNO individuals. Specifically, the risk of developing PCa in MHO individuals was 2.166 times higher than in MHNO individuals (OR=2.166, 95%CI: 1.133-4.139), and the risk in MUO individuals was is 2.398 times higher than in MHNO individuals(OR=2.398, 95%CI:1.271-4.523). Individuals with hyperglycemia and elevated triglycerides also had a higher risk of developing PCa (hyperglycemia:OR=1.488, 95%CI: 1.001-2.210; elevated triglycerides: OR=2.292, 95%CI: 1.419-3.702). Those with more than or equal to three metabolic risk factors had an increased risk of PCa (OR=1.990, 95%CI: 1.166-3.396). Sensitivity analysis indicated an increased risk of PCa in MUO individuals compared to MHNO individuals. Conclusion In this retrospective study, individuals with MHO and MUO had a higher risk of developing PCa.
Collapse
Affiliation(s)
- Jinru Wang
- School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Aireti Apizi
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hao Qiu
- School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Ning Tao
- School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Hengqing An
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
32
|
Elsaid S, Wu X, Tee SS. Fructose vs. glucose: modulating stem cell growth and function through sugar supplementation. FEBS Open Bio 2024; 14:1277-1290. [PMID: 38923793 PMCID: PMC11301265 DOI: 10.1002/2211-5463.13846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/17/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
In multicellular organisms, stem cells are impacted by microenvironmental resources such as nutrient availability and oxygen tension for their survival, growth, and differentiation. However, the accessibility of these resources in the pericellular environment greatly varies from organ to organ. This divergence in resource availability leads to variations in the potency and differentiation potential of stem cells. This study aimed to explore the distinct effects of glucose and fructose, as well as different oxygen tensions, on the growth dynamics, cytokine production, and differentiation of stem cells. We showed that replacing glucose with fructose subjected stem cells to stress, resulting in increased Hif1α expression and stability, which in turn led to a reduction in cell proliferation, and alterations in cytokine production. However, fructose failed to induce differentiation of human mesenchymal stem cells (hMSCs) as well as mouse fibroblasts into mature adipocytes compared to glucose, despite the upregulation of key markers of adipogenesis, including C/EBPβ, and PPARγ. Conversely, we showed that fructose induced undifferentiated mouse fibroblasts to release cytokines associated with senescence, including IL1α1, IL6, IL8, MCP1, and TNF1α, suggesting that these cells were undergoing lipolysis. Taken together, our results suggest that altering the culture conditions through changes in hexose levels and oxygen tension places considerable stress on stem cells. Additional research is required to further characterize the mechanisms governing stem cell response to their microenvironments.
Collapse
Affiliation(s)
- Salaheldeen Elsaid
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Xiangdong Wu
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Sui Seng Tee
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| |
Collapse
|
33
|
Braga GDC, Simões JLB, Teixeira Dos Santos YJ, Filho JCM, Bagatini MD. The impacts of obesity in rheumatoid arthritis and insights into therapeutic purinergic modulation. Int Immunopharmacol 2024; 136:112357. [PMID: 38810303 DOI: 10.1016/j.intimp.2024.112357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024]
Abstract
Rheumatoid Arthritis (RA) is an autoimmune condition responsible for the impairment of synovia and joints, endangering the functionality of individuals and contributing to mortality. Currently, obesity is increasing worldwide, and recent studies have suggested an association between such condition and RA. In this sense, obese individuals present a lower capacity for achieving remission and present more intense symptoms of the disease, demonstrating a link between both disorders. Different studies aim to understand the possible connection between the conditions; however, few is known in this sense. Therefore, knowing that obesity can alter the activity of multiple body systems, this work's objective is to evaluate the main modifications caused by obesity, which can be linked to the pathophysiology of RA, highlighting as relevant topics obesity's negative impact triggering systemic inflammation, intestinal dysbiosis, endocrine disbalances. Furthermore, the relationship between oxidative stress and obesity also deserves to be highlighted, considering the influence of reactive oxygen species (ROS) accumulation in RA exacerbation. Additionally, many of those characteristics influenced by obesity, along with the classic peculiarities of RA pathophysiology, can also be associated with purinergic signaling. Hence, this work suggests possible connections between the purinergic system and RA, proposing potential therapeutic targets against RA to be studied.
Collapse
|
34
|
DeBari MK, Johnston EK, Scott JV, Ilzuka E, Sun W, Webster-Wood VA, Abbott RD. A Preliminary Study on Factors That Drive Patient Variability in Human Subcutaneous Adipose Tissues. Cells 2024; 13:1240. [PMID: 39120271 PMCID: PMC11311805 DOI: 10.3390/cells13151240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Adipose tissue is a dynamic regulatory organ that has profound effects on the overall health of patients. Unfortunately, inconsistencies in human adipose tissues are extensive and multifactorial, including large variability in cellular sizes, lipid content, inflammation, extracellular matrix components, mechanics, and cytokines secreted. Given the high human variability, and since much of what is known about adipose tissue is from animal models, we sought to establish correlations and patterns between biological, mechanical, and epidemiological properties of human adipose tissues. To do this, twenty-six independent variables were cataloged for twenty patients, which included patient demographics and factors that drive health, obesity, and fibrosis. A factorial analysis for mixed data (FAMD) was used to analyze patterns in the dataset (with BMI > 25), and a correlation matrix was used to identify interactions between quantitative variables. Vascular endothelial growth factor A (VEGFA) and actin alpha 2, smooth muscle (ACTA2) gene expression were the highest loadings in the first two dimensions of the FAMD. The number of adipocytes was also a key driver of patient-related differences, where a decrease in the density of adipocytes was associated with aging. Aging was also correlated with a decrease in overall lipid percentage of subcutaneous tissue, with lipid deposition being favored extracellularly, an increase in transforming growth factor-β1 (TGFβ1), and an increase in M1 macrophage polarization. An important finding was that self-identified race contributed to variance between patients in this study, where Black patients had significantly lower gene expression levels of TGFβ1 and ACTA2. This finding supports the urgent need to account for patient ancestry in biomedical research to develop better therapeutic strategies for all patients. Another important finding was that TGFβ induced factor homeobox 1 (TGIF1), an understudied signaling molecule, which is highly correlated with leptin signaling, was correlated with metabolic inflammation. Furthermore, this study draws attention to what we define as "extracellular lipid droplets", which were consistently found in collagen-rich regions of the obese adipose tissues evaluated here. Reduced levels of TGIF1 were correlated with higher numbers of extracellular lipid droplets and an inability to suppress fibrotic changes in adipose tissue. Finally, this study indicated that M1 and M2 macrophage markers were correlated with each other and leptin in patients with a BMI > 25. This finding supports growing evidence that macrophage polarization in obesity involves a complex, interconnecting network system rather than a full switch in activation patterns from M2 to M1 with increasing body mass. Overall, this study reinforces key findings in animal studies and identifies important areas for future research, where human and animal studies are divergent. Understanding key drivers of human patient variability is required to unravel the complex metabolic health of unique patients.
Collapse
Affiliation(s)
- Megan K. DeBari
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Elizabeth K. Johnston
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Jacqueline V. Scott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| | - Erica Ilzuka
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Wenhuan Sun
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Victoria A. Webster-Wood
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Rosalyn D. Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (M.K.D.); (E.K.J.); (J.V.S.); (E.I.); (V.A.W.-W.)
| |
Collapse
|
35
|
Halabitska I, Babinets L, Oksenych V, Kamyshnyi O. Diabetes and Osteoarthritis: Exploring the Interactions and Therapeutic Implications of Insulin, Metformin, and GLP-1-Based Interventions. Biomedicines 2024; 12:1630. [PMID: 39200096 PMCID: PMC11351146 DOI: 10.3390/biomedicines12081630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/18/2024] [Accepted: 07/21/2024] [Indexed: 09/01/2024] Open
Abstract
Diabetes mellitus (DM) and osteoarthritis (OA) are prevalent chronic conditions with shared pathophysiological links, including inflammation and metabolic dysregulation. This study investigates the potential impact of insulin, metformin, and GLP-1-based therapies on OA progression. Methods involved a literature review of clinical trials and mechanistic studies exploring the effects of these medications on OA outcomes. Results indicate that insulin, beyond its role in glycemic control, may modulate inflammatory pathways relevant to OA, potentially influencing joint health. Metformin, recognized for its anti-inflammatory properties via AMPK activation, shows promise in mitigating OA progression by preserving cartilage integrity and reducing inflammatory markers. GLP-1-based therapies, known for enhancing insulin secretion and improving metabolic profiles in DM, also exhibit anti-inflammatory effects that may benefit OA by suppressing cytokine-mediated joint inflammation and supporting cartilage repair mechanisms. Conclusions suggest that these medications, while primarily indicated for diabetes management, hold therapeutic potential in OA by targeting common underlying mechanisms. Further clinical trials are warranted to validate these findings and explore optimal therapeutic strategies for managing both DM and OA comorbidities effectively.
Collapse
Affiliation(s)
- Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Voli Square, 1, 46001 Ternopil, Ukraine
| | - Liliia Babinets
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Voli Square, 1, 46001 Ternopil, Ukraine
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine;
| |
Collapse
|
36
|
Dahdah N, Tercero-Alcázar C, Malagón MM, Garcia-Roves PM, Guzmán-Ruiz R. Interrelation of adipose tissue macrophages and fibrosis in obesity. Biochem Pharmacol 2024; 225:116324. [PMID: 38815633 DOI: 10.1016/j.bcp.2024.116324] [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: 01/09/2024] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Obesity is characterized by adipose tissue expansion, extracellular matrix remodelling and unresolved inflammation that contribute to insulin resistance and fibrosis. Adipose tissue macrophages represent the most abundant class of immune cells in adipose tissue inflammation and could be key mediators of adipocyte dysfunction and fibrosis in obesity. Although macrophage activation states are classically defined by the M1/M2 polarization nomenclature, novel studies have revealed a more complex range of macrophage phenotypes in response to external condition or the surrounding microenvironment. Here, we discuss the plasticity of adipose tissue macrophages (ATMs) in response to their microenvironment in obesity, with special focus on macrophage infiltration and polarization, and their contribution to adipose tissue fibrosis. A better understanding of the role of ATMs as regulators of adipose tissue remodelling may provide novel therapeutic strategies against obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Norma Dahdah
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Carmen Tercero-Alcázar
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María M Malagón
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Cell Biology, Physiology and Immunology, IMIBIC, Reina Sofía University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Pablo Miguel Garcia-Roves
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Rocío Guzmán-Ruiz
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Cell Biology, Physiology and Immunology, IMIBIC, Reina Sofía University Hospital, University of Córdoba, 14004 Córdoba, Spain.
| |
Collapse
|
37
|
Sandoval-Bórquez A, Carrión P, Hernández MP, Pérez JA, Tapia-Castillo A, Vecchiola A, Fardella CE, Carvajal CA. Adipose Tissue Dysfunction and the Role of Adipocyte-Derived Extracellular Vesicles in Obesity and Metabolic Syndrome. J Endocr Soc 2024; 8:bvae126. [PMID: 38988671 PMCID: PMC11234198 DOI: 10.1210/jendso/bvae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Indexed: 07/12/2024] Open
Abstract
Obesity is a major public health issue that is associated with metabolic diseases including diabetes mellitus type 2 and metabolic syndrome. This pathology leads to detrimental cardiovascular health and secondary effects, such as lipotoxicity, inflammation, and oxidative stress. Recently, extracellular vesicles (EVs) have been highlighted as novel players participating in human physiology and pathophysiology. In obesity, adipose tissue is related to the active shedding of adipocyte-derived extracellular vesicles (AdEVs). The current review explores and highlights the role of AdEVs and their cargo in obesity and metabolic syndrome. AdEVs are proposed to play an important role in obesity and its comorbidities. AdEVs are biological nanoparticles mainly shed by visceral and subcutaneous adipose tissue, acting in physiological and pathophysiological conditions, and also carrying different cargo biomolecules, such as RNA, microRNA (miRNA), proteins, and lipids, among others. RNA and miRNA have local and systemic effects affecting gene expression in target cell types via paracrine and endocrine actions. State of the art analyses identified some miRNAs, such as miR-222, miR-23b, miR-4429, miR-148b, and miR-4269, that could potentially affect cell pathways involved in obesity-related comorbidities, such as chronic inflammation and fibrosis. Similarly, AdEVs-proteins (RBP4, perilipin-A, FABP, mimecan, TGFBI) and AdEVs-lipids (sphingolipids) have been linked to the obesity pathophysiology. The current knowledge about AdEVs along with further research would support and reveal novel pathways, potential biomarkers, and therapeutic options in obesity.
Collapse
Affiliation(s)
- Alejandra Sandoval-Bórquez
- School of Medical Technology, Faculty of Science, Pontificia Universidad Católica de Valparaiso, Valparaiso 2373223, Chile
| | - Pablo Carrión
- Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago 8330074, Chile
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile
| | - María Paz Hernández
- Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago 8330074, Chile
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile
| | - Jorge A Pérez
- Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago 8330074, Chile
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile
| | - Alejandra Tapia-Castillo
- Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago 8330074, Chile
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile
| | - Andrea Vecchiola
- Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago 8330074, Chile
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile
| | - Carlos E Fardella
- Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago 8330074, Chile
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile
| | - Cristian A Carvajal
- Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago 8330074, Chile
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile
| |
Collapse
|
38
|
GAO X, QIANG P, CHANG J, FAN L, YANG F, XU Q. Huoxue Jiedu Huayu recipe inhibits macrophage-secreted vascular endothelial growth factor-a on angiogenesis and alleviates renal fibrosis in the contralateral kidneys of unilateral ureteral obstruction rats. J TRADIT CHIN MED 2024; 44:458-467. [PMID: 38767629 PMCID: PMC11077158 DOI: 10.19852/j.cnki.jtcm.20240423.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/07/2023] [Indexed: 05/22/2024]
Abstract
OBJECTIVE:To elucidate the mechanism by which Huoxue Jiedu Huayu recipe (, HJHR) regulates angiogenesis in the contralateral kidney of unilateral ureteral obstruction (UUO) rats and the mechanism by which it reduces of renal fibrosis. METHODS Male Wistar rats were randomly divided into 4 groups: the sham group, UUO group (180 d of left ureter ligation), UUO plus eplerenone (EPL) group, and UUO plus HJHR group. After 180 d of oral drug administration, blood and contralateral kidneys were collected for analysis. Angiogenesis- and fibrosis-related indexes were detected. RESULTS HJHR and EPL improved structural damage and renal interstitial fibrosis in the contralateral kidney and reduced the protein expression levels of α-smooth muscle actin (α-SMA), vimentin and collagen I. Moreover, these treatments could reduce the expression of vascular endothelial growth factor-A (VEGFA) by inhibiting the infiltration of macrophages. Furthermore, HJHR and EPL significantly reduced the expression of CD34 and CD105 by downregulating VEGFA production, which inhibited angiogenesis. Finally, the coexpressions of CD34, CD105 and α-SMA were decreased in the HJHR and EPL groups, indicating that endothelial-to-mesenchymal transition was inhibited. CONCLUSIONS These findings confirm that HJHR alleviates contralateral renal fibrosis by inhibiting VEGFA-induced angiogenesis, encourage the use of HJHR against renal interstitial fibrosis and provide a theoretical basis for the clinical management of patients with CKD.
Collapse
Affiliation(s)
- Xiaomeng GAO
- 1 Graduate School, Hebei University of Chinese Medicine; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Panpan QIANG
- 1 Graduate School, Hebei University of Chinese Medicine; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Jingyue CHANG
- 1 Graduate School, Hebei University of Chinese Medicine; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Lili FAN
- 1 Graduate School, Hebei University of Chinese Medicine; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Fan YANG
- 2 Graduate School, Hebei University of Chinese Medicine; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine; Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Qingyou XU
- 2 Graduate School, Hebei University of Chinese Medicine; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine; Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| |
Collapse
|
39
|
Wang F, Baverel V, Chaumonnot K, Bourragat A, Bellenger J, Bellenger S, Zhou W, Narce M, Garrido C, Kohli E. The endoplasmic reticulum stress protein GRP94 modulates cathepsin L activity in M2 macrophages in conditions of obesity-associated inflammation and contributes to their pro-inflammatory profile. Int J Obes (Lond) 2024; 48:830-840. [PMID: 38351251 DOI: 10.1038/s41366-024-01478-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 01/11/2024] [Accepted: 01/18/2024] [Indexed: 02/19/2024]
Abstract
BACKGROUND/OBJECTIVES Adipose tissue macrophages (ATM) are key actors in the pathophysiology of obesity-related diseases. They have a unique intermediate M2-M1 phenotype which has been linked to endoplasmic reticulum (ER) stress. We previously reported that human M2 macrophages treated with the ER stress inducer thapsigargin switched to a pro-inflammatory phenotype that depended on the stress protein GRP94. In these conditions, GRP94 promoted cathepsin L secretion and was co-secreted with complement C3. As cathepsin L and complement C3 have been reported to play a role in the pathophysiology of obesity, in this work we studied the involvement of GRP94 in the pro-inflammatory phenotype of ATM. METHODS GRP94, cathepsin L and C3 expression were analyzed in CD206 + ATM from mice, WT or obesity-resistant transgenic fat-1, fed a high-fat diet (HFD) or a standard diet. GRP94 colocalization with cathepsin L and C3 and its effects were analyzed in human primary macrophages using thapsigargin as a control to induce ER stress and palmitic acid (PA) as a driver of metabolic activation. RESULTS In WT, but not in fat-1 mice, fed a HFD, we observed an increase in crown-like structures consisting of CD206 + pSTAT1+ macrophages showing high expression of GRP94 that colocalized with cathepsin L and C3. In vitro experiments showed that PA favored a M2-M1 switch depending on GRP94. This switch was prevented by omega-3 fatty acids. PA-induced GRP94-cathepsin L colocalization and a decrease in cathepsin L enzymatic activity within the cells (while the enzymatic activity in the extracellular medium was increased). These effects were prevented by the GRP94 inhibitor PU-WS13. CONCLUSIONS GRP94 is overexpressed in macrophages both in in vivo and in vitro conditions of obesity-associated inflammation and is involved in changing their profile towards a more pro-inflammatory profile. It colocalizes with complement C3 and cathepsin L and modulates cathepsin L activity.
Collapse
Affiliation(s)
- Fangmin Wang
- UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- UMR INSERM/uB/AGROSUP 1231, Team HSP-Pathies, labellisée Ligue Nationale contre le Cancer and Laboratoire d'Excellence LipSTIC, UBFC, Dijon, France
- Zhejiang Provincial Key Lab of Addiction, The Affiliated Kangning Hospital of Ningbo University, Ningbo Kangning Hospital, Ningbo University, Ningbo, China
| | - Valentin Baverel
- UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- UMR INSERM/uB/AGROSUP 1231, Team HSP-Pathies, labellisée Ligue Nationale contre le Cancer and Laboratoire d'Excellence LipSTIC, UBFC, Dijon, France
| | - Killian Chaumonnot
- UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- UMR INSERM/uB/AGROSUP 1231, Team HSP-Pathies, labellisée Ligue Nationale contre le Cancer and Laboratoire d'Excellence LipSTIC, UBFC, Dijon, France
| | - Amina Bourragat
- UMR INSERM/uB/AGROSUP 1231, Team Lipness, Labellisée Laboratoire d'Excellence LipSTIC, Dijon, France
- UFR Sciences Vie Terre Environnement, Université de Bourgogne, Dijon, France
| | - Jerome Bellenger
- UMR INSERM/uB/AGROSUP 1231, Team Lipness, Labellisée Laboratoire d'Excellence LipSTIC, Dijon, France
- UFR Sciences Vie Terre Environnement, Université de Bourgogne, Dijon, France
| | - Sandrine Bellenger
- UMR INSERM/uB/AGROSUP 1231, Team Lipness, Labellisée Laboratoire d'Excellence LipSTIC, Dijon, France
- UFR Sciences Vie Terre Environnement, Université de Bourgogne, Dijon, France
| | - Wenhua Zhou
- Zhejiang Provincial Key Lab of Addiction, The Affiliated Kangning Hospital of Ningbo University, Ningbo Kangning Hospital, Ningbo University, Ningbo, China
| | - Michel Narce
- UMR INSERM/uB/AGROSUP 1231, Team Lipness, Labellisée Laboratoire d'Excellence LipSTIC, Dijon, France
- UFR Sciences Vie Terre Environnement, Université de Bourgogne, Dijon, France
| | - Carmen Garrido
- UFR des Sciences de Santé, Université de Bourgogne, Dijon, France
- UMR INSERM/uB/AGROSUP 1231, Team HSP-Pathies, labellisée Ligue Nationale contre le Cancer and Laboratoire d'Excellence LipSTIC, UBFC, Dijon, France
- Centre Anti-Cancéreux Georges François Leclerc, Dijon, France
| | - Evelyne Kohli
- UFR des Sciences de Santé, Université de Bourgogne, Dijon, France.
- UMR INSERM/uB/AGROSUP 1231, Team HSP-Pathies, labellisée Ligue Nationale contre le Cancer and Laboratoire d'Excellence LipSTIC, UBFC, Dijon, France.
- CHU, Dijon, France.
| |
Collapse
|
40
|
Fei S, Liu M, Shanshan H, Xie R, Danni W, Ningying Z. Association between weight-adjusted-waist index and depression: a cross-sectional study. Endocr Connect 2024; 13:e230450. [PMID: 38657652 PMCID: PMC11227061 DOI: 10.1530/ec-23-0450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/23/2024] [Indexed: 04/26/2024]
Abstract
Background Depression has become a multifaceted global health issue, with complex connections to obesity. Weight-adjusted-waist index (WWI) can effectively evaluate central obesity, but the relationship between WWI and depression has not been well studied. The study aims to investigate the potential correlation between these two health parameters. Methods According to the data from National Health and Nutrition Examination Survey, this cross-sectional study used multiple regression analysis, subgroup analysis, and smooth curve fitting to explore the relationship between WWI and depression. The assessment ability of WWI was evaluated and compared to other obesity indicators using the receiver operating characteristic (ROC) curve. Results This study analyzed 38,154 participants. Higher WWI is associated with higher depression scores (β = 0.41; 95% CI, 0.36-0.47). After adjusting for various confounding factors, the positive correlation between WWI and depression remained significant (P for trend < 0.0001). Nonlinear positive correlation was detected with a breakpoint of 11.14. ROC analysis shows that compared to other obesity indicators (ROCWWI = 0.593; ROCBMI = 0.584; and ROCWC = 0.581), the correlation between WWI and depression has better discrimination and accuracy. DII mediated 4.93%, SII mediated 5.08%, and sedentary mediated 0.35% of the total association between WWI and depression. Conclusion WWI levels were related to an increased likelihood of depression and showed a stronger relationship than BMI and waist circumference. Our findings indicated that WWI may serve as a simple anthropometric index to evaluate depression.
Collapse
Affiliation(s)
- Sun Fei
- Wuxi Medical College of Jiangnan University, Wuxi, China
| | - Min Liu
- Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Hu Shanshan
- Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Ruijie Xie
- Department of Microsurgery, University of South China, Hengyang, China
| | - Wu Danni
- Wuxi Medical College of Jiangnan University, Wuxi, China
| | - Zhou Ningying
- Wuxi Medical College of Jiangnan University, Wuxi, China
| |
Collapse
|
41
|
Chandrasekaran P, Weiskirchen R. The signaling pathways in obesity-related complications. J Cell Commun Signal 2024; 18:e12039. [PMID: 38946722 PMCID: PMC11208128 DOI: 10.1002/ccs3.12039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 07/02/2024] Open
Abstract
Obesity, a rapidly expanding epidemic worldwide, is known to exacerbate many medical conditions, making it a significant factor in multiple diseases and their associated complications. This threatening epidemic is linked to various harmful conditions such as type 2 diabetes mellitus, hypertension, metabolic dysfunction-associated steatotic liver disease, polycystic ovary syndrome, cardiovascular diseases (CVDs), dyslipidemia, and cancer. The rise in urbanization and sedentary lifestyles creates an environment that fosters obesity, leading to both psychosocial and medical complications. To identify individuals at risk and ensure timely treatment, it is crucial to have a better understanding of the pathophysiology of obesity and its comorbidities. This comprehensive review highlights the relationship between obesity and obesity-associated complications, including type 2 diabetes, hypertension, (CVDs), dyslipidemia, polycystic ovary syndrome, metabolic dysfunction-associated steatotic liver disease, gastrointestinal complications, and obstructive sleep apnea. It also explores the potential mechanisms underlying these associations. A thorough analysis of the interplay between obesity and its associated complications is vital in developing effective therapeutic strategies to combat the exponential increase in global obesity rates and mitigate the deadly consequences of this polygenic condition.
Collapse
Affiliation(s)
| | - Ralf Weiskirchen
- Institute of Molecular PathobiochemistryExperimental Gene Therapy and Clinical Chemistry (IFMPEGKC)RWTH University Hospital AachenAachenGermany
| |
Collapse
|
42
|
Iraji Asiabadi A, Esmaeil N, Zargar Kharazi A, Dabiri A, Varshosaz J. Harnessing IL-10 induced anti-inflammatory response in maturing macrophages in presence of electrospun dexamethasone-loaded PLLA scaffold. J Biomed Mater Res B Appl Biomater 2024; 112:e35411. [PMID: 38773758 DOI: 10.1002/jbm.b.35411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/19/2024] [Accepted: 04/13/2024] [Indexed: 05/24/2024]
Abstract
The ultimate goal of tissue engineering is to repair and regenerate damaged tissue or organ. Achieving this goal requires blood vessel networks to supply oxygen and nutrients to new forming tissues. Macrophages are part of the immune system whose behavior plays a significant role in angiogenesis and blood vessel formation. On the other hand, macrophages are versatile cells that change their behavior in response to environmental stimuli. Given that implantation of a biomaterial is followed by inflammation; therefore, we reasoned that this inflammatory condition in tissue spaces modulates the final phenotype of macrophages. Also, we hypothesized that anti-inflammatory glucocorticoid dexamethasone improves modulating macrophages behavior. To check these concepts, we investigated the macrophages that had matured in an inflammatory media. Furthermore, we examined macrophages' behavior after maturation on a dexamethasone-containing scaffold and analyzed how the behavioral change of maturing macrophages stimulates other macrophages in the same environment. In this study, the expression of pro-inflammatory markers TNFa and NFκB1 along with pro-healing markers IL-10 and CD163 were investigated to study the behavior of macrophages. Our results showed that macrophages that were matured in the inflammatory media in vitro increase expression of IL-10, which in turn decreased the expression of pro-inflammatory markers TNFa and NFκB in maturing macrophages. Also, macrophages that were matured on dexamethasone-containing scaffolds decreased the expression of IL-10, TNFa, and NFκB and increase the expression of CD163 compared to the control group. Moreover, the modulation of anti-inflammatory response in maturing macrophages on dexamethasone-containing scaffold resulted in increased expression of TNFa and CD163 by other macrophages in the same media. The results obtained in this study, proposing strategies to improve healing through controlling the behavior of maturing macrophages and present a promising perspective for inflammation control using tissue engineering scaffolds.
Collapse
Affiliation(s)
- Arash Iraji Asiabadi
- Tissue Engineering and Nanotechnology, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anousheh Zargar Kharazi
- Tissue Engineering and Nanotechnology, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arezou Dabiri
- Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
43
|
Lékó AH, Gregory-Flores A, Marchette RCN, Gomez JL, Vendruscolo JCM, Repunte-Canonigo V, Choung V, Deschaine SL, Whiting KE, Jackson SN, Cornejo MP, Perello M, You ZB, Eckhaus M, Rasineni K, Janda KD, Zorman B, Sumazin P, Koob GF, Michaelides M, Sanna PP, Vendruscolo LF, Leggio L. Genetic or pharmacological GHSR blockade has sexually dimorphic effects in rodents on a high-fat diet. Commun Biol 2024; 7:632. [PMID: 38796563 PMCID: PMC11127961 DOI: 10.1038/s42003-024-06303-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 05/08/2024] [Indexed: 05/28/2024] Open
Abstract
The stomach-derived hormone ghrelin regulates essential physiological functions. The ghrelin receptor (GHSR) has ligand-independent actions; therefore, GHSR gene deletion may be a reasonable approach to investigate the role of this system in feeding behaviors and diet-induced obesity (DIO). Here, we investigate the effects of a long-term (12-month) high-fat (HFD) versus regular diet on obesity-related measures in global GHSR-KO and wild-type (WT) Wistar male and female rats. Our main findings are that the GHSR gene deletion protects against DIO and decreases food intake during HFD in male but not in female rats. GHSR gene deletion increases thermogenesis and brain glucose uptake in male rats and modifies the effects of HFD on brain glucose metabolism in a sex-specific manner, as assessed with small animal positron emission tomography. We use RNA-sequencing to show that GHSR-KO rats have upregulated expression of genes responsible for fat oxidation in brown adipose tissue. Central administration of a novel GHSR inverse agonist, PF-5190457, attenuates ghrelin-induced food intake, but only in male, not in female mice. HFD-induced binge-like eating is reduced by inverse agonism in both sexes. Our results support GHSR as a promising target for new pharmacotherapies for obesity.
Collapse
Affiliation(s)
- András H Lékó
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Adriana Gregory-Flores
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Renata C N Marchette
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Juan L Gomez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Janaina C M Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Vez Repunte-Canonigo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Vicky Choung
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Sara L Deschaine
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
| | - Kimberly E Whiting
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Shelley N Jackson
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Maria Paula Cornejo
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Argentina
| | - Mario Perello
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Argentina
| | - Zhi-Bing You
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Eckhaus
- Pathology Service, Division of Veterinary Resources, Office of Research Services, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Karuna Rasineni
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kim D Janda
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Barry Zorman
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Pavel Sumazin
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - George F Koob
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Pietro P Sanna
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI, USA.
- Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Department of Neuroscience, Georgetown University Medical Center, Washington DC, USA.
| |
Collapse
|
44
|
Touceda V, Fontana Estevez F, Cacciagiú L, Finocchietto P, Bustos R, Vidal A, Berg G, Morales C, González G, Miksztowicz V. Liraglutide improves adipose tissue remodeling and mitochondrial dynamics in a visceral obesity model induced by a high-fat diet. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 6:100185. [PMID: 38846009 PMCID: PMC11153889 DOI: 10.1016/j.crphar.2024.100185] [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: 02/02/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
Central obesity is characterized by visceral adipose tissue (VAT) expansion, considered one of the main risk factors for metabolic complications. In recent years, new drugs have been studied for obesity treatment. Liraglutide (LGT), a GLP-1 agonist, decreases body weight, however, several mechanisms of action on VAT are still unknown. Aim to study the effect of LGT on factors associated with VAT remodeling and mitochondrial dynamics in mice fed a high-fat diet (HFD). Methods C57BL/6 mice were divided into Control (C) and HFD. After 15 weeks of feeding, each group was subdivided according to LGT administration for 5 weeks: C, C + LGT, HFD, and HFD + LGT. In epididymal AT (EAT) we evaluated histological and mitochondrial characteristics, vascularity, gelatinase activity (MMPs), and galectin-3 expression. Results HFD presented larger adipocytes (p < 0.05), and lower vascular density and MMP-9 activity (p < 0.01) than C, while a major number of smaller adipocytes (p < 0.05) and an increase in vascularity (p < 0.001) and MMP-9 activity (p < 0.01) was observed in HFD + LGT. Collagen content was higher (p < 0.05) in EAT from HFD and decreased in HFD + LGT. In C, C + LGT, and HFD + LGT, mitochondria were predominantly tubular-shaped while in HFD mitochondria were mostly spherical (p < 0.001). Conclusion LGT positively influences VAT behavior by modulating gelatinase activity, enhancing vascularization, and improving adipocyte histological characteristics. Additionally, LGT improves mitochondrial dynamics, a process that would favor VAT functionality.
Collapse
Affiliation(s)
- Vanessa Touceda
- Pontificia Universidad Católica Argentina. Facultad de Medicina, Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Bioquímica General y Bucal, Buenos Aires, Argentina
| | - Florencia Fontana Estevez
- Pontificia Universidad Católica Argentina. Facultad de Medicina, Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Buenos Aires, Argentina
| | - Leonardo Cacciagiú
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Bioquímica General y Bucal, Buenos Aires, Argentina
- Hospital General de Agudos Teodoro Álvarez, Laboratorio Central, Sección Bioquímica, Buenos Aires, Argentina
| | - Paola Finocchietto
- Universidad de Buenos Aires, Facultad de Medicina. Instituto de Inmunología, Genética y Metabolismo (INIGEM. UBA-CONICET), Laboratorio de Metabolismo del Oxígeno, Buenos Aires, Argentina
| | - Romina Bustos
- Pontificia Universidad Católica Argentina. Facultad de Medicina, Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Buenos Aires, Argentina
| | - Agustina Vidal
- Pontificia Universidad Católica Argentina. Facultad de Medicina, Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Buenos Aires, Argentina
| | - Gabriela Berg
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica, Laboratorio de Lípidos y Aterosclerosis, Buenos Aires, Argentina
| | - Celina Morales
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, Argentina
| | - Germán González
- Pontificia Universidad Católica Argentina. Facultad de Medicina, Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Buenos Aires, Argentina
| | - Veronica Miksztowicz
- Pontificia Universidad Católica Argentina. Facultad de Medicina, Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Bioquímica General y Bucal, Buenos Aires, Argentina
| |
Collapse
|
45
|
Zhang RR, You HR, Geng YY, Li XG, Sun Y, Hou J, Ji LC, Shi JL, Zhang LB, Yang BQ. Predicting major adverse cardiovascular events within 3 years by optimization of radiomics model derived from pericoronary adipose tissue on coronary computed tomography angiography: a case-control study. BMC Med Imaging 2024; 24:117. [PMID: 38773416 PMCID: PMC11110286 DOI: 10.1186/s12880-024-01295-4] [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: 02/13/2023] [Accepted: 05/07/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Coronary inflammation induces changes in pericoronary adipose tissue (PCAT) can be detected by coronary computed tomography angiography (CCTA). Our aim was to investigate whether different PCAT radiomics model based on CCTA could improve the prediction of major adverse cardiovascular events (MACE) within 3 years. METHODS This retrospective study included 141 consecutive patients with MACE and matched to patients with non-MACE (n = 141). Patients were randomly assigned into training and test datasets at a ratio of 8:2. After the robust radiomics features were selected by using the Spearman correlation analysis and the least absolute shrinkage and selection operator, radiomics models were built based on different machine learning algorithms. The clinical model was then calculated according to independent clinical risk factors. Finally, an overall model was established using the radiomics features and the clinical factors. Performance of the models was evaluated for discrimination degree, calibration degree, and clinical usefulness. RESULTS The diagnostic performance of the PCAT model was superior to that of the RCA-model, LAD-model, and LCX-model alone, with AUCs of 0.723, 0.675, 0.664, and 0.623, respectively. The overall model showed superior diagnostic performance than that of the PCAT-model and Cli-model, with AUCs of 0.797, 0.723, and 0.706, respectively. Calibration curve showed good fitness of the overall model, and decision curve analyze demonstrated that the model provides greater clinical benefit. CONCLUSION The CCTA-based PCAT radiomics features of three major coronary arteries have the potential to be used as a predictor for MACE. The overall model incorporating the radiomics features and clinical factors offered significantly higher discrimination ability for MACE than using radiomics or clinical factors alone.
Collapse
Affiliation(s)
- Rong-Rong Zhang
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning Province, 110016, P.R. China
- Jinzhou Medical University, Jinzhou, China
| | - Hong-Rui You
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning Province, 110016, P.R. China
- Jinzhou Medical University, Jinzhou, China
| | - Ya-Yuan Geng
- Shukun Technology Co., Ltd, West Beichen Road, Beijing, China
| | - Xiao-Gang Li
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning Province, 110016, P.R. China
- Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, China
| | - Yu Sun
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning Province, 110016, P.R. China
- Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, China
| | - Jie Hou
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning Province, 110016, P.R. China
- Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, China
| | - Lian-Chang Ji
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning Province, 110016, P.R. China
- Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, China
| | | | - Li-Bo Zhang
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning Province, 110016, P.R. China
- Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, China
| | - Ben-Qiang Yang
- Department of Radiology, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning Province, 110016, P.R. China.
- Key Laboratory of Cardiovascular Imaging and Research of Liaoning Province, Shenyang, China.
| |
Collapse
|
46
|
Klotsman M, Anderson WH, Gilor C. Drug release profile of a novel exenatide long-term drug delivery system (OKV-119) administered to cats. BMC Vet Res 2024; 20:211. [PMID: 38762728 PMCID: PMC11102179 DOI: 10.1186/s12917-024-04051-6] [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: 05/26/2023] [Accepted: 05/02/2024] [Indexed: 05/20/2024] Open
Abstract
Beneficial weight-loss properties of glucagon-like peptide-1 receptor agonists (GLP-1RA) in obese people, with corresponding improvements in cardiometabolic risk factors, are well established. OKV-119 is an investigational drug delivery system that is being developed for the long-term delivery of the GLP-1RA exenatide to feline patients. The purpose of this study was to evaluate the drug release characteristics of subcutaneous OKV-119 implants configured to release exenatide for 84 days. Following a 7-day acclimation period, five purpose-bred cats were implanted with OKV-119 protypes and observed for a 112-day study period. Food intake, weekly plasma exenatide concentrations and body weight were measured. Exenatide plasma concentrations were detected at the first measured timepoint (Day 7) and maintained above baseline for over 84 Days. Over the first 28 days, reduced caloric intake and a reduction in body weight were observed in four of five cats. In these cats, a body weight reduction of at least 5% was maintained throughout the 112-day study period. This study demonstrates that a single OKV-119 implant can deliver the GLP-1RA exenatide for a months long duration. Results suggest that exposure to exenatide plasma concentrations ranging from 1.5 ng/ml to 4 ng/ml are sufficient for inducing weight loss in cats.
Collapse
Affiliation(s)
| | - Wayne H Anderson
- Okava Pharmaceuticals, San Francisco, CA, USA
- Pulmonary and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chen Gilor
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
47
|
Liang Y, Shuai Q, Zhang X, Jin S, Guo Y, Yu Z, Xu X, Ao R, Peng Z, Lv H, He S, Wang C, Song G, Liu Z, Zhao H, Feng Q, Du R, Zheng B, Chen Z, Xie J. Incorporation of Decidual Stromal Cells Derived Exosomes in Sodium Alginate Hydrogel as an Innovative Therapeutic Strategy for Advancing Endometrial Regeneration and Reinstating Fertility. Adv Healthc Mater 2024; 13:e2303674. [PMID: 38315148 DOI: 10.1002/adhm.202303674] [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: 11/20/2023] [Indexed: 02/07/2024]
Abstract
Intrauterine adhesion (IUA) stands as a prevalent medical condition characterized by endometrial fibrosis and scar tissue formation within the uterine cavity, resulting in infertility and, in severe cases, recurrent miscarriages. Cell therapy, especially with stem cells, offers an alternative to surgery, but concerns about uncontrolled differentiation and tumorigenicity limit its use. Exosomes, more stable and immunogenicity-reduced than parent cells, have emerged as a promising avenue for IUA treatment. In this study, a novel approach has been proposed wherein exosomes originating from decidual stromal cells (DSCs) are encapsulated within sodium alginate hydrogel (SAH) scaffolds to repair endometrial damage and restore fertility in a mouse IUA model. Current results demonstrate that in situ injection of DSC-derived exosomes (DSC-exos)/SAH into the uterine cavity has the capability to induce uterine angiogenesis, initiate mesenchymal-to-epithelial transformation (MET), facilitate collagen fiber remodeling and dissolution, promote endometrial regeneration, enhance endometrial receptivity, and contribute to the recovery of fertility. RNA sequencing and advanced bioinformatics analysis reveal miRNA enrichment in exosomes, potentially supporting endometrial repair. This finding elucidates how DSC-exos/SAH mechanistically fosters collagen ablation, endometrium regeneration, and fertility recovery, holding the potential to introduce a novel IUA treatment and offering invaluable insights into the realm of regenerative medicine.
Collapse
Affiliation(s)
- Yuxiang Liang
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
- Shanxi Key Laboratory of Human Disease and Animal Models, Experimental Animal Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Qizhi Shuai
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| | - Xiao Zhang
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| | - Shanshan Jin
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| | - Yuqian Guo
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| | - Zhaowei Yu
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| | - Xinrui Xu
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| | - Ruifang Ao
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| | - Zhiwei Peng
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| | - Huimin Lv
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
- Department of Obstetrics and Gynecology, Third Hospital of Shanxi Medical University (Shanxi Bethune Hospital), Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
| | - Sheng He
- Department of Radiology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Chunfang Wang
- Shanxi Key Laboratory of Human Disease and Animal Models, Experimental Animal Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Guohua Song
- Shanxi Key Laboratory of Human Disease and Animal Models, Experimental Animal Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Zhizhen Liu
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| | - Hong Zhao
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| | - Qilong Feng
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| | - Ruochen Du
- Shanxi Key Laboratory of Human Disease and Animal Models, Experimental Animal Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Bin Zheng
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Zhaoyang Chen
- Shanxi Key Laboratory of Human Disease and Animal Models, Experimental Animal Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jun Xie
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Taiyuan, Shanxi, 030001, China
| |
Collapse
|
48
|
da Cruz SP, da Cruz SP, Pereira S, Saboya C, Ramalho A. Vitamin D and the Metabolic Phenotype in Weight Loss After Bariatric Surgery: A Longitudinal Study. Obes Surg 2024; 34:1561-1568. [PMID: 38459277 DOI: 10.1007/s11695-024-07148-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] [Received: 12/25/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024]
Abstract
PURPOSE To evaluate the influence of vitamin D (VD) concentrations coupled with metabolic phenotypes preoperatively and 6 months after Roux-en-Y gastric bypass (RYGB) on body variables and weight loss. MATERIALS AND METHODS A longitudinal, retrospective, analytical study comprising 30 adult individuals assessed preoperatively (T0) and 6 months (T1) after undergoing Roux-en-Y gastric bypass. The participants were distributed preoperatively into metabolically healthy obese (MHO) and metabolically unhealthy obese individuals (MUHO) according to the HOMA-IR classification, as well as the adequacy and inadequacy of vitamin D concentrations in the form of 25(OH)D. All participants were assessed for weight, height, body mass index (BMI), waist circumference (WC), waist-to-height ratio (WHtR), visceral adiposity index (VAI), body circularity index (BCI), body adiposity index (BAI), weight loss, and assessment of 25(OH)D and 1,25(OH)2D concentrations using high-performance liquid chromatography with an ultraviolet detector (HPLC-UV). The statistical program used was SPSS version 21. RESULTS VD adequacy and a healthy phenotype in the preoperative period may play an important role concerning body fat distribution, as the body averages for WHtR (0.020*) and BCI (0.020*) were lower in MHO participants. In comparison, those with VD inadequacy and MUHOs had higher BAI averages (0.000*) in the postoperative period. Furthermore, it is possible that VD inadequacy before and after RYGB, even in the presence of an unhealthy phenotype, may contribute to the increase in VAI values (0.029*) after this surgery. Only those with inadequate VD and MUHOs had higher 25(OH)D concentrations. Besides, this unhealthy phenotype had a greater reduction in BMI in the early postoperative period (p < 0.001). CONCLUSION This study suggests that VD adequacy and the presence of a healthy phenotype appear to have a positive impact on the reduction of visceral fat in the context of pre- and postoperative obesity. In addition, there was a greater weight reduction in those with VD inadequacy and in MUHO, which suggests that the volumetric dilution effect of VD and catabolism after bariatric surgery is more pronounced in this specific metabolic phenotype.
Collapse
Affiliation(s)
- Suelem Pereira da Cruz
- Center for Research On Micronutrients, Federal University of Rio de Janeiro (NPqM/UFRJ), Rio de Janeiro, 21941-902, Brazil.
| | - Sabrina Pereira da Cruz
- Center for Research On Micronutrients, Federal University of Rio de Janeiro (NPqM/UFRJ), Rio de Janeiro, 21941-902, Brazil
| | - Silvia Pereira
- Multidisciplinary Center for Bariatric and Metabolic Surgery, Federal University of Rio de Janeiro (NPqM/UFRJ), Rio de Janeiro, 21941-902, Brazil
| | - Carlos Saboya
- Brazilian Society of Bariatric and Metabolic Surgery, Rio de Janeiro, 22280-020, Brazil
| | - Andréa Ramalho
- Center for Research On Micronutrients, Federal University of Rio de Janeiro (NPqM/UFRJ), Rio de Janeiro, 21941-902, Brazil
| |
Collapse
|
49
|
Hateley C, Olona A, Halliday L, Edin ML, Ko JH, Forlano R, Terra X, Lih FB, Beltrán-Debón R, Manousou P, Purkayastha S, Moorthy K, Thursz MR, Zhang G, Goldin RD, Zeldin DC, Petretto E, Behmoaras J. Multi-tissue profiling of oxylipins reveal a conserved up-regulation of epoxide:diol ratio that associates with white adipose tissue inflammation and liver steatosis in obesity. EBioMedicine 2024; 103:105127. [PMID: 38677183 PMCID: PMC11061246 DOI: 10.1016/j.ebiom.2024.105127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Obesity drives maladaptive changes in the white adipose tissue (WAT) which can progressively cause insulin resistance, type 2 diabetes mellitus (T2DM) and metabolic dysfunction-associated liver disease (MASLD). Obesity-mediated loss of WAT homeostasis can trigger liver steatosis through dysregulated lipid pathways such as those related to polyunsaturated fatty acid (PUFA)-derived oxylipins. However, the exact relationship between oxylipins and metabolic syndrome remains elusive and cross-tissue dynamics of oxylipins are ill-defined. METHODS We quantified PUFA-related oxylipin species in the omental WAT, liver biopsies and plasma of 88 patients undergoing bariatric surgery (female N = 79) and 9 patients (female N = 4) undergoing upper gastrointestinal surgery, using UPLC-MS/MS. We integrated oxylipin abundance with WAT phenotypes (adipogenesis, adipocyte hypertrophy, macrophage infiltration, type I and VI collagen remodelling) and the severity of MASLD (steatosis, inflammation, fibrosis) quantified in each biopsy. The integrative analysis was subjected to (i) adjustment for known risk factors and, (ii) control for potential drug-effects through UPLC-MS/MS analysis of metformin-treated fat explants ex vivo. FINDINGS We reveal a generalized down-regulation of cytochrome P450 (CYP)-derived diols during obesity conserved between the WAT and plasma. Notably, epoxide:diol ratio, indicative of soluble epoxide hydrolyse (sEH) activity, increases with WAT inflammation/fibrosis, hepatic steatosis and T2DM. Increased 12,13-EpOME:DiHOME in WAT and liver is a marker of worsening metabolic syndrome in patients with obesity. INTERPRETATION These findings suggest a dampened sEH activity and a possible role of fatty acid diols during metabolic syndrome in major metabolic organs such as WAT and liver. They also have implications in view of the clinical trials based on sEH inhibition for metabolic syndrome. FUNDING Wellcome Trust (PS3431_WMIH); Duke-NUS (Intramural Goh Cardiovascular Research Award (Duke-NUS-GCR/2022/0020); National Medical Research Council (OFLCG22may-0011); National Institute of Environmental Health Sciences (Z01 ES025034); NIHR Imperial Biomedical Research Centre.
Collapse
Affiliation(s)
- Charlotte Hateley
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Antoni Olona
- Centre for Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Laura Halliday
- Department of Surgery and Cancer, Imperial College London, UK
| | - Matthew L Edin
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Jeong-Hun Ko
- Division of Brain Sciences, Imperial College Faculty of Medicine, London, UK
| | - Roberta Forlano
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Ximena Terra
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, MoBioFood Research Group, Tarragona, Spain
| | - Fred B Lih
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Raúl Beltrán-Debón
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, MoBioFood Research Group, Tarragona, Spain
| | - Penelopi Manousou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Sanjay Purkayastha
- Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK; University of Brunel, Kingston Lane, Uxbridge, London, UB8 3PH, UK
| | - Krishna Moorthy
- Department of Surgery and Cancer, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Mark R Thursz
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Guodong Zhang
- Department of Nutrition, College of Agriculture and Environmental Sciences, 3135 Meyer Hall, One Shields Avenue, UC Davis, Davis, CA, 95616, USA
| | - Robert D Goldin
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Darryl C Zeldin
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Enrico Petretto
- Centre for Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore; Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University (CPU), Nanjing, China
| | - Jacques Behmoaras
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK; Centre for Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
50
|
Jalil JE, Gabrielli L, Ocaranza MP, MacNab P, Fernández R, Grassi B, Jofré P, Verdejo H, Acevedo M, Cordova S, Sanhueza L, Greig D. New Mechanisms to Prevent Heart Failure with Preserved Ejection Fraction Using Glucagon-like Peptide-1 Receptor Agonism (GLP-1 RA) in Metabolic Syndrome and in Type 2 Diabetes: A Review. Int J Mol Sci 2024; 25:4407. [PMID: 38673991 PMCID: PMC11049921 DOI: 10.3390/ijms25084407] [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: 02/07/2024] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
This review examines the impact of obesity on the pathophysiology of heart failure with preserved ejection fraction (HFpEF) and focuses on novel mechanisms for HFpEF prevention using a glucagon-like peptide-1 receptor agonism (GLP-1 RA). Obesity can lead to HFpEF through various mechanisms, including low-grade systemic inflammation, adipocyte dysfunction, accumulation of visceral adipose tissue, and increased pericardial/epicardial adipose tissue (contributing to an increase in myocardial fat content and interstitial fibrosis). Glucagon-like peptide 1 (GLP-1) is an incretin hormone that is released from the enteroendocrine L-cells in the gut. GLP-1 reduces blood glucose levels by stimulating insulin synthesis, suppressing islet α-cell function, and promoting the proliferation and differentiation of β-cells. GLP-1 regulates gastric emptying and appetite, and GLP-1 RA is currently indicated for treating type 2 diabetes (T2D), obesity, and metabolic syndrome (MS). Recent evidence indicates that GLP-1 RA may play a significant role in preventing HFpEF in patients with obesity, MS, or obese T2D. This effect may be due to activating cardioprotective mechanisms (the endogenous counter-regulatory renin angiotensin system and the AMPK/mTOR pathway) and by inhibiting deleterious remodeling mechanisms (the PKA/RhoA/ROCK pathway, aldosterone levels, and microinflammation). However, there is still a need for further research to validate the impact of these mechanisms on humans.
Collapse
Affiliation(s)
- Jorge E. Jalil
- Pontificia Universidad Católica de Chile, School of Medicine, Division of Cardiovascular Diseases, Santiago 8330055, Chile; (L.G.); (P.M.); (R.F.); (H.V.); (M.A.); (S.C.); (L.S.); (D.G.)
| | - Luigi Gabrielli
- Pontificia Universidad Católica de Chile, School of Medicine, Division of Cardiovascular Diseases, Santiago 8330055, Chile; (L.G.); (P.M.); (R.F.); (H.V.); (M.A.); (S.C.); (L.S.); (D.G.)
| | - María Paz Ocaranza
- Pontificia Universidad Católica de Chile, School of Medicine, Division of Cardiovascular Diseases, Santiago 8330055, Chile; (L.G.); (P.M.); (R.F.); (H.V.); (M.A.); (S.C.); (L.S.); (D.G.)
| | - Paul MacNab
- Pontificia Universidad Católica de Chile, School of Medicine, Division of Cardiovascular Diseases, Santiago 8330055, Chile; (L.G.); (P.M.); (R.F.); (H.V.); (M.A.); (S.C.); (L.S.); (D.G.)
| | - Rodrigo Fernández
- Pontificia Universidad Católica de Chile, School of Medicine, Division of Cardiovascular Diseases, Santiago 8330055, Chile; (L.G.); (P.M.); (R.F.); (H.V.); (M.A.); (S.C.); (L.S.); (D.G.)
| | - Bruno Grassi
- Pontificia Universidad Católica de Chile, School of Medicine, Department of Nutrition and Diabetes, Santiago 8330055, Chile; (B.G.); (P.J.)
| | - Paulina Jofré
- Pontificia Universidad Católica de Chile, School of Medicine, Department of Nutrition and Diabetes, Santiago 8330055, Chile; (B.G.); (P.J.)
| | - Hugo Verdejo
- Pontificia Universidad Católica de Chile, School of Medicine, Division of Cardiovascular Diseases, Santiago 8330055, Chile; (L.G.); (P.M.); (R.F.); (H.V.); (M.A.); (S.C.); (L.S.); (D.G.)
| | - Monica Acevedo
- Pontificia Universidad Católica de Chile, School of Medicine, Division of Cardiovascular Diseases, Santiago 8330055, Chile; (L.G.); (P.M.); (R.F.); (H.V.); (M.A.); (S.C.); (L.S.); (D.G.)
| | - Samuel Cordova
- Pontificia Universidad Católica de Chile, School of Medicine, Division of Cardiovascular Diseases, Santiago 8330055, Chile; (L.G.); (P.M.); (R.F.); (H.V.); (M.A.); (S.C.); (L.S.); (D.G.)
| | - Luis Sanhueza
- Pontificia Universidad Católica de Chile, School of Medicine, Division of Cardiovascular Diseases, Santiago 8330055, Chile; (L.G.); (P.M.); (R.F.); (H.V.); (M.A.); (S.C.); (L.S.); (D.G.)
| | - Douglas Greig
- Pontificia Universidad Católica de Chile, School of Medicine, Division of Cardiovascular Diseases, Santiago 8330055, Chile; (L.G.); (P.M.); (R.F.); (H.V.); (M.A.); (S.C.); (L.S.); (D.G.)
| |
Collapse
|