1
|
Krueger ABC, Zhu X, Siddiqi S, Whitehead EC, Tang H, Jordan KL, Lerman A, Lerman LO. Mesenchymal Stem/Stromal Cells Reverse Adipose Tissue Inflammation in Pigs with Metabolic Syndrome and Renovascular Hypertension. Cells 2025; 14:40. [PMID: 39791741 PMCID: PMC11720192 DOI: 10.3390/cells14010040] [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: 11/12/2024] [Revised: 12/28/2024] [Accepted: 12/29/2024] [Indexed: 01/12/2025] Open
Abstract
Metabolic syndrome (MetS) is associated with low-grade inflammation, which can be exacerbated by renal artery stenosis (RAS) and renovascular hypertension, potentially worsening outcomes through pro-inflammatory cytokines. This study investigated whether mesenchymal stem/stromal cells (MSCs) could reduce fat inflammation in pigs with MetS and RAS. Twenty-four pigs were divided into Lean (control), MetS, MetS + RAS, and MetS + RAS + MSCs. In the MSC-treated group, autologous adipose-derived MSCs (107 cells) were injected into the renal artery six weeks after RAS induction. After four weeks, fat volumes and inflammatory markers were assessed. MSC treatment reduced levels of pro-inflammatory cytokines (MCP-1, TNF-a, IL-6) in the renal vein blood and in perirenal fat. The MSCs also decreased fat fibrosis, restored adipocyte size, and altered adipogenesis-related gene expression, particularly in the perirenal fat. These effects were less pronounced in subcutaneous fat. The MSC therapy attenuated fat inflammation and improved metabolic outcomes in pigs with MetS + RAS, suggesting that adipose-derived MSCs may offer a promising therapeutic approach for metabolic disorders.
Collapse
Affiliation(s)
- Alexander B. C. Krueger
- Division of Nephrology & Hypertension, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA; (A.B.C.K.); (X.Z.); (E.C.W.); (H.T.); (K.L.J.)
| | - Xiangyang Zhu
- Division of Nephrology & Hypertension, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA; (A.B.C.K.); (X.Z.); (E.C.W.); (H.T.); (K.L.J.)
| | - Sarosh Siddiqi
- Division of Nephrology & Hypertension, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA; (A.B.C.K.); (X.Z.); (E.C.W.); (H.T.); (K.L.J.)
| | - Emma C. Whitehead
- Division of Nephrology & Hypertension, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA; (A.B.C.K.); (X.Z.); (E.C.W.); (H.T.); (K.L.J.)
| | - Hui Tang
- Division of Nephrology & Hypertension, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA; (A.B.C.K.); (X.Z.); (E.C.W.); (H.T.); (K.L.J.)
| | - Kyra L. Jordan
- Division of Nephrology & Hypertension, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA; (A.B.C.K.); (X.Z.); (E.C.W.); (H.T.); (K.L.J.)
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA;
| | - Lilach O. Lerman
- Division of Nephrology & Hypertension, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA; (A.B.C.K.); (X.Z.); (E.C.W.); (H.T.); (K.L.J.)
| |
Collapse
|
2
|
Soták M, Clark M, Suur BE, Börgeson E. Inflammation and resolution in obesity. Nat Rev Endocrinol 2025; 21:45-61. [PMID: 39448830 DOI: 10.1038/s41574-024-01047-y] [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: 09/30/2024] [Indexed: 10/26/2024]
Abstract
Inflammation is an essential physiological defence mechanism, but prolonged or excessive inflammation can cause disease. Indeed, unresolved systemic and adipose tissue inflammation drives obesity-related cardiovascular disease and type 2 diabetes mellitus. Drugs targeting pro-inflammatory cytokine pathways or inflammasome activation have been approved for clinical use for the past two decades. However, potentially serious adverse effects, such as drug-induced weight gain and increased susceptibility to infections, prevented their wider clinical implementation. Furthermore, these drugs do not modulate the resolution phase of inflammation. This phase is an active process orchestrated by specialized pro-resolving mediators, such as lipoxins, and other endogenous resolution mechanisms. Pro-resolving mediators mitigate inflammation and development of obesity-related disease, for instance, alleviating insulin resistance and atherosclerosis in experimental disease models, so mechanisms to modulate their activity are, therefore, of great therapeutic interest. Here, we review current clinical attempts to either target pro-inflammatory mediators (IL-1β, NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome, tumour necrosis factor (TNF) and IL-6) or utilize endogenous resolution pathways to reduce obesity-related inflammation and improve cardiometabolic outcomes. A remaining challenge in the field is to establish more precise biomarkers that can differentiate between acute and chronic inflammation and to assess the functionality of individual leukocyte populations. Such advancements would improve the monitoring of drug effects and support personalized treatment strategies that battle obesity-related inflammation and cardiometabolic disease.
Collapse
Affiliation(s)
- Matúš Soták
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Madison Clark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Bianca E Suur
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Emma Börgeson
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
3
|
Zhang L, Xu F, Hou L. IL-6 and diabetic kidney disease. Front Immunol 2024; 15:1465625. [PMID: 39749325 PMCID: PMC11693507 DOI: 10.3389/fimmu.2024.1465625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Diabetic kidney disease (DKD) is a severe microvascular complication of diabetes associated with high mortality and disability rates. Inflammation has emerged as a key pathological mechanism in DKD, prompting interest in novel therapeutic approaches targeting inflammatory pathways. Interleukin-6 (IL-6), a well-established inflammatory cytokine known for mediating various inflammatory responses, has attracted great attention in the DKD field. Although multiple in vivo and in vitro studies highlight the potential of targeting IL-6 in DKD treatment, its exact roles in the disease remains unclear. This review presents the roles of IL-6 in the pathogenesis of DKD, including immunoinflammation, metabolism, hemodynamics, and ferroptosis. In addition, we summarize the current status of IL-6 inhibitors in DKD-related clinical trials and discuss the potential of targeting IL-6 for treating DKD in the clinic.
Collapse
Affiliation(s)
- Lei Zhang
- Pharmacy Department, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| | - Futian Xu
- Logistics Management Department, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| | - Liyan Hou
- Pharmacy Department, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| |
Collapse
|
4
|
Shang D, Zhao S. Molecular mechanisms of obesity predisposes to atopic dermatitis. Front Immunol 2024; 15:1473105. [PMID: 39564133 PMCID: PMC11574713 DOI: 10.3389/fimmu.2024.1473105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/15/2024] [Indexed: 11/21/2024] Open
Abstract
Obesity is a prevalent metabolic disease that reduces bacterial diversity, colonizes the epidermis with lipophilic bacteria, and increases intestinal pro-inflammatory species, all of which lead to impaired epithelial barriers. Adipose tissue secretes immunomodulatory molecules, such as adipokines, leptin, and adiponectin, which alters the morphology of adipocytes and macrophages as well as modulates T cell differentiation and peripheral Th2-dominated immune responses. Atopic dermatitis (AD) and obesity have similar pathological manifestations, including inflammation as well as insulin and leptin resistance. This review examines the major mechanisms between obesity and AD, which focus on the effect on skin and gut microbiota, immune responses mediated by the toll like receptor (TLR) signaling pathway, and changes in cytokine levels (TNF-a, IL-6, IL-4, and IL13). Moreover, we describe the potential effects of adipokines on AD and finally mechanisms by which PPAR-γ suppresses and regulates type 2 immunity.
Collapse
Affiliation(s)
- Dajin Shang
- School of China Medical University, Shenyang, Liaoning, China
| | - Shengnan Zhao
- School of China Medical University, Shenyang, Liaoning, China
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Sobieska K, Buczyńska A, Krętowski AJ, Popławska-Kita A. Iron homeostasis and insulin sensitivity: unraveling the complex interactions. Rev Endocr Metab Disord 2024; 25:925-939. [PMID: 39287729 PMCID: PMC11470850 DOI: 10.1007/s11154-024-09908-7] [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] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
Diabetes has arisen as a noteworthy global health issue, marked by escalating incidence and mortality rates. Insulin, crucial for preserving euglycemia, acts as a vital energy provider for various tissues. Iron metabolism notably plays a significant role in the development of insulin resistance, a key factor in the onset of various metabolic disorders. The intricate interaction between iron and insulin signaling encompasses complex regulatory mechanisms at the molecular level, thereby impacting cellular reactions to insulin. The intricate interplay between insulin and glucagon, essential for precise regulation of hepatic glucose production and systemic glucose levels, may be influenced by certain microelements for instance zinc, copper, iron, boron, calcium, cobalt, chromium, iodine, magnesium and selenium. While significant progress has been achieved in elucidating the pathophysiological connections between iron overload and glucose metabolism, our understanding of the involvement of the Fenton reaction and oxidative stress in insulin resistance influencing many chronical conditions remains limited. Furthermore, the exploration of the multifaceted roles of insulin in the human body continues to be a subject of active investigation by numerous scientific researchers. This review comprehensively outlines the potential adverse impact of iron overload on insulin function and glucose metabolism. Additionally, we provide a synthesis of findings derived from various research domains, encompassing population studies, animal models, and clinical investigations, to scrutinize the multifaceted relationship between iron and insulin sensitivity. Moreover, we delineate instances of correlations between serum iron levels and various medical conditions, including the diabetes also gestational diabetes and obesity.
Collapse
Affiliation(s)
- Katarzyna Sobieska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Angelika Buczyńska
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Adam Jacek Krętowski
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Anna Popławska-Kita
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
6
|
Sergi D, Melloni M, Passaro A, Neri LM. Influence of Type 2 Diabetes and Adipose Tissue Dysfunction on Breast Cancer and Potential Benefits from Nutraceuticals Inducible in Microalgae. Nutrients 2024; 16:3243. [PMID: 39408212 PMCID: PMC11478231 DOI: 10.3390/nu16193243] [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: 08/31/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Breast cancer (BC) represents the most prevalent cancer in women at any age after puberty. From a pathogenetic prospective, despite a wide array of risk factors being identified thus far, poor metabolic health is emerging as a putative risk factor for BC. In particular, type 2 diabetes mellitus (T2DM) provides a perfect example bridging the gap between poor metabolic health and BC risk. Indeed, T2DM is preceded by a status of hyperinsulinemia and is characterised by hyperglycaemia, with both factors representing potential contributors to BC onset and progression. Additionally, the aberrant secretome of the dysfunctional, hypertrophic adipocytes, typical of obesity, characterised by pro-inflammatory mediators, is a shared pathogenetic factor between T2DM and BC. In this review, we provide an overview on the effects of hyperglycaemia and hyperinsulinemia, hallmarks of type 2 diabetes mellitus, on breast cancer risk, progression, treatment and prognosis. Furthermore, we dissect the role of the adipose-tissue-secreted adipokines as additional players in the pathogenesis of BC. Finally, we focus on microalgae as a novel superfood and a source of nutraceuticals able to mitigate BC risk by improving metabolic health and targeting cellular pathways, which are disrupted in the context of T2DM and obesity.
Collapse
Affiliation(s)
- Domenico Sergi
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
| | - Mattia Melloni
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
| | - Angelina Passaro
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
| | - Luca Maria Neri
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
- Laboratory for Technologies of Advanced Therapies (LTTA)—Electron Microscopy Center, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| |
Collapse
|
7
|
Fraiz GM, Bonifácio DB, Lacerda UV, Cardoso RR, Corich V, Giacomini A, Martino HSD, Echeverría SE, de Barros FAR, Milagro FI, Bressan J. Green Tea Kombucha Impacts Inflammation and Salivary Microbiota in Individuals with Excess Body Weight: A Randomized Controlled Trial. Nutrients 2024; 16:3186. [PMID: 39339787 PMCID: PMC11435194 DOI: 10.3390/nu16183186] [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: 09/04/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Green tea kombucha (GTK) is a fermented beverage with promising health benefits, but few studies proved its impact on human health. Thus, we aimed to investigate the impact of GTK on weight loss, inflammation, and salivary microbiota in individuals with excess body weight. METHODS This is a randomized controlled clinical trial that lasted 10 weeks with two groups of individuals with excess body weight: control (CG; n = 29; caloric restriction) and kombucha (KG; n = 30; caloric restriction + 200 mL GTK). Body composition, anthropometry, saliva, and blood collection were performed in the beginning and end of the intervention. Plasma interleukins were determined by flow cytometry. Salivary microbiota was analyzed by 16S rRNA sequencing. RESULTS Both groups decreased weight, BMI, and body fat (p < 0.001) after the intervention, but there were no differences between groups. The KG reduced lipid accumulation product (LAP) (p = 0.029). Both groups decreased IL-1β and IL-8, but IL-6 increased in the CG (p = 0.023) compared to the kombucha group. Alpha and beta diversity of salivary microbiota increased in the KG. Moreover, the KG presented lower Bacillota/Bacteroidota ratio (p = 0.028), and BMI was positively associated with the Bacillota phylum. CONCLUSIONS GTK did not enhance weight loss, but it decreased the LAP. GTK helped in the inflammatory profile and induced positive changes in oral microbiota composition.
Collapse
Affiliation(s)
- Gabriela Macedo Fraiz
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, Brazil; (G.M.F.); (D.B.B.); (H.S.D.M.)
- Department of Nutrition, Food Science and Physiology, Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain (F.I.M.)
| | - Dandara Baia Bonifácio
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, Brazil; (G.M.F.); (D.B.B.); (H.S.D.M.)
| | - Udielle Vermelho Lacerda
- Department of Food and Technology, Universidade Federal de Viçosa, Viçosa 36570-900, Brazil; (U.V.L.); (R.R.C.); (F.A.R.d.B.)
| | - Rodrigo Rezende Cardoso
- Department of Food and Technology, Universidade Federal de Viçosa, Viçosa 36570-900, Brazil; (U.V.L.); (R.R.C.); (F.A.R.d.B.)
| | - Viviana Corich
- Department of Agronomy, Food Natural Resources, and Environment (DAFNAE), Università degli Studi di Padova, 35020 Legnaro, Italy; (V.C.); (A.G.)
| | - Alessio Giacomini
- Department of Agronomy, Food Natural Resources, and Environment (DAFNAE), Università degli Studi di Padova, 35020 Legnaro, Italy; (V.C.); (A.G.)
| | - Hércia Stampini Duarte Martino
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, Brazil; (G.M.F.); (D.B.B.); (H.S.D.M.)
| | - Sergio Esteban Echeverría
- Department of Nutrition, Food Science and Physiology, Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain (F.I.M.)
| | | | - Fermín I. Milagro
- Department of Nutrition, Food Science and Physiology, Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain (F.I.M.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Institute of Health Carlos III, 28029 Madrid, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Josefina Bressan
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, Brazil; (G.M.F.); (D.B.B.); (H.S.D.M.)
| |
Collapse
|
8
|
Branquinho J, Neves RL, Martin RP, Arata JG, Bittencourt CA, Araújo RC, Icimoto MY, Pesquero JB. Kinin B1 receptor deficiency promotes enhanced adipose tissue thermogenic response to β3-adrenergic stimulation. Inflamm Res 2024; 73:1565-1579. [PMID: 39017739 DOI: 10.1007/s00011-024-01917-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024] Open
Abstract
OBJECTIVE AND DESIGN Kinin B1 receptor (B1R) has a key role in adipocytes to protect against obesity and glycemic metabolism, thus becoming a potential target for regulation of energy metabolism and adipose tissue thermogenesis. MATERIAL OR SUBJECTS Kinin B1 knockout mice (B1KO) were subjected to acute induction with CL 316,243 and chronic cold exposure. METHODS Metabolic and histological analyses, gene and protein expression and RNA-seq were performed on interscapular brown adipose tissue (iBAT) and inguinal white adipose tissue (iWAT) of mice. RESULTS B1KO mice, under acute effect of CL 316,243, exhibited increased energy expenditure and upregulated thermogenic genes in iWAT. They were also protected from chronic cold, showing enhanced non-shivering thermogenesis with increased iBAT mass (~ 90%) and recruitment of beige adipocytes in iWAT (~ 50%). Positive modulation of thermogenic and electron transport chain genes, reaching a 14.5-fold increase for Ucp1 in iWAT. RNA-seq revealed activation of the insulin signaling pathways for iBAT and oxidative phosphorylation, tricarboxylic acid cycle, and browning pathways for iWAT. CONCLUSION B1R deficiency induced metabolic and gene expression alterations in adipose tissue, activating thermogenic pathways and increasing energy metabolism. B1R antagonists emerge as promising therapeutic targets for regulating obesity and associated metabolic disorders, such as inflammation and diabetes.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/drug effects
- Adrenergic beta-3 Receptor Agonists/pharmacology
- Cold Temperature
- Dioxoles/pharmacology
- Energy Metabolism/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Receptor, Bradykinin B1/genetics
- Receptor, Bradykinin B1/metabolism
- Receptors, Adrenergic, beta-3/genetics
- Receptors, Adrenergic, beta-3/metabolism
- Thermogenesis/drug effects
- Thiazoles/pharmacology
- Uncoupling Protein 1/genetics
- Uncoupling Protein 1/metabolism
Collapse
Affiliation(s)
- Jéssica Branquinho
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Raquel L Neves
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Renan P Martin
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Júlia G Arata
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Clarissa A Bittencourt
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Ronaldo C Araújo
- Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Marcelo Y Icimoto
- Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil.
| | - João B Pesquero
- Center for Research and Molecular Diagnostic of Genetic Diseases, Department of Biophysics, Federal University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
9
|
Bosch-Sierra N, Grau-del Valle C, Hermenejildo J, Hermo-Argibay A, Salazar JD, Garrido M, Navajas-Porras B, Sáez G, Morillas C, Bañuls C. The Impact of Weight Loss on Inflammation, Oxidative Stress, and Mitochondrial Function in Subjects with Obesity. Antioxidants (Basel) 2024; 13:870. [PMID: 39061938 PMCID: PMC11273387 DOI: 10.3390/antiox13070870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Inflammation, oxidative stress, and mitochondrial function are implicated in the development of obesity and its comorbidities. The purpose of this study was to assess the impact of weight loss through calorie restriction on the metabolic profile, inflammatory and oxidative stress parameters, and mitochondrial respiration in an obese population. A total of 109 subjects underwent two cycles of a very low-calorie diet alternated with a low-calorie diet (24 weeks). We analyzed biochemical and inflammatory parameters in serum, as well as oxidative stress markers, mRNA antioxidant gene expression, and mitochondrial respiration in peripheral blood mononuclear cells (PBMCs). After the intervention, there was an improvement in both insulin resistance and lipid profiles, including cholesterol subfractions. Weight loss produced a significant reduction in mitochondrial ROSs content and an increase in glutathione levels, coupled with an enhancement in the mRNA expression of antioxidant systems (SOD1, GSR, and CAT). In addition, a significant improvement in basal oxygen consumption, maximal respiration, and ATP production was observed. These findings demonstrate that moderate weight loss can improve insulin resistance, lipid profiles and subfractions, inflammatory and oxidative stress parameters, and mitochondrial respiration. Therefore, we can affirm that dietary intervention can simultaneously achieve significant weight loss and improve metabolic profile and mitochondrial function in obesity.
Collapse
Affiliation(s)
- Neus Bosch-Sierra
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (N.B.-S.); (C.G.-d.V.); (J.H.); (A.H.-A.); (J.D.S.); (M.G.); (B.N.-P.); (C.M.)
| | - Carmen Grau-del Valle
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (N.B.-S.); (C.G.-d.V.); (J.H.); (A.H.-A.); (J.D.S.); (M.G.); (B.N.-P.); (C.M.)
| | - Jonathan Hermenejildo
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (N.B.-S.); (C.G.-d.V.); (J.H.); (A.H.-A.); (J.D.S.); (M.G.); (B.N.-P.); (C.M.)
| | - Alberto Hermo-Argibay
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (N.B.-S.); (C.G.-d.V.); (J.H.); (A.H.-A.); (J.D.S.); (M.G.); (B.N.-P.); (C.M.)
| | - Juan Diego Salazar
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (N.B.-S.); (C.G.-d.V.); (J.H.); (A.H.-A.); (J.D.S.); (M.G.); (B.N.-P.); (C.M.)
| | - Marta Garrido
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (N.B.-S.); (C.G.-d.V.); (J.H.); (A.H.-A.); (J.D.S.); (M.G.); (B.N.-P.); (C.M.)
| | - Beatriz Navajas-Porras
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (N.B.-S.); (C.G.-d.V.); (J.H.); (A.H.-A.); (J.D.S.); (M.G.); (B.N.-P.); (C.M.)
| | - Guillermo Sáez
- Service of Clinical Analysis, University Hospital Dr. Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain;
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
| | - Carlos Morillas
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (N.B.-S.); (C.G.-d.V.); (J.H.); (A.H.-A.); (J.D.S.); (M.G.); (B.N.-P.); (C.M.)
- Department of Medicine, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
| | - Celia Bañuls
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (N.B.-S.); (C.G.-d.V.); (J.H.); (A.H.-A.); (J.D.S.); (M.G.); (B.N.-P.); (C.M.)
| |
Collapse
|
10
|
Li X, Wang R, Hou Z, Sun Q. Urban-rural differences in the prevalence and associated factors of sarcopenia: A systematic review and meta-analysis. Arch Gerontol Geriatr 2024; 122:105390. [PMID: 38460267 DOI: 10.1016/j.archger.2024.105390] [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: 12/12/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND Loss of muscle mass, muscle strength, and/or physical performance due to aging is known as sarcopenia. Regardless of how serious this illness is, no single diagnostic criteria have been established. Much research conducted recently has demonstrated differences between built environment characteristics (i.e., urban and rural) and the occurrence of sarcopenia; however, variations in sarcopenia prevalence in urban-rural areas around the world have been reported by fewer studies. This work sought to determine how sarcopenia prevalence varied between urban and rural areas and to explore the associated influencing factors. METHODS Using the pertinent MESH phrases and free words, PubMed, Web of Science, Embase, and China national knowledge infrastructure databases were scanned for core sarcopenia literature up to February 26, 2023. Observational studies involving urban-rural patients with sarcopenia published in Chinese and English, and assessing muscle mass via computed tomography, bioelectrical impedance, or dual-energy X-ray absorption techniques were considered as inclusion criteria. The meta-analysis involved analysis of the urban-rural prevalence in subgroups by diagnostic criteria, tools for assessing muscle mass and study type, as well as the factors related to urban-rural differences in the occurrence of sarcopenia. STATA version 11.0 was used to perform the statistical analysis. RESULTS Sixty-six articles involving 433,091 participants were included for analysis: of which 27 were analyzed for both prevalence and related factors whereas 39 were for only prevalence. The meta-analysis revealed the prevalence of sarcopenia to be 0.18 (95 % CI 0.14-0.22), with significant heterogeneity (P < 0.001; I2 = 99.9 %). Moreover, the prevalence of sarcopenia in urban group [0.16 (I2 = 99.9 %, 95 % CI 0.1-0.22)] was lower than in rural group [0.2 (I2 = 99.6 %, 95 % CI 0.16-0.25)] and urban-rural group [0.21 (I2 = 97.5 %, 95 % CI 0.16-0.25)]. Besides, the factors significantly associated with sarcopenia in urban-rural areas were age, gender, BMI, malnutrition, physical activity, and polypharmacy. There was significant heterogeneity between these factors and the association of sarcopenia. CONCLUSIONS Sarcopenia is associated with aspects of the built environment, and studies have revealed that sarcopenia is more common in rural than in urban populations with influencing factors including age, gender, BMI, poor nutrition, insufficient physical activity, and polypharmacy. The lack of uniform diagnostic criteria makes a robust and comprehensive assessment difficult. Therefore, the formation of certain universal and standardized diagnostic criteria will help future research on sarcopenia.
Collapse
Affiliation(s)
- Xiaoyan Li
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rongyun Wang
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhuoer Hou
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiuhua Sun
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
11
|
Liu M, Lu F, Feng J. Aging and homeostasis of the hypodermis in the age-related deterioration of skin function. Cell Death Dis 2024; 15:443. [PMID: 38914551 PMCID: PMC11196735 DOI: 10.1038/s41419-024-06818-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 02/01/2024] [Accepted: 06/10/2024] [Indexed: 06/26/2024]
Abstract
Adipose tissues in the hypodermis, the crucial stem cell reservoir in the skin and the endocrine organ for the maintenance of skin homeostasis undergo significant changes during skin aging. Dermal white adipose tissue (dWAT) has recently been recognized as an important organ for both non-metabolic and metabolic health in skin regeneration and rejuvenation. Defective differentiation, adipogenesis, improper adipocytokine production, and immunological dissonance dysfunction in dWAT lead to age-associated clinical changes. Here, we review age-related alterations in dWAT across levels, emphasizing the mechanisms underlying the regulation of aging. We also discuss the pathogenic changes involved in age-related fat dysfunction and the unfavorable consequences of accelerated skin aging, such as chronic inflammaging, immunosenescence, delayed wound healing, and fibrosis. Research has shown that adipose aging is an early initiation event and a potential target for extending longevity. We believe that adipose tissues play an essential role in aging and form a potential therapeutic target for the treatment of age-related skin diseases. Further research is needed to improve our understanding of this phenomenon.
Collapse
Affiliation(s)
- Meiqi Liu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Jingwei Feng
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China.
| |
Collapse
|
12
|
Wystrychowski G, Simka-Lampa K, Witkowska A, Sobecko E, Skubis-Sikora A, Sikora B, Wojtyna E, Golda A, Gwizdek K, Wróbel M, Sędek Ł, Górczyńska-Kosiorz S, Szweda-Gandor N, Trautsolt W, Francuz T, Kruszniewska-Rajs C, Gola J. Selected microRNA Expression and Protein Regulator Secretion by Adipose Tissue-Derived Mesenchymal Stem Cells and Metabolic Syndrome. Int J Mol Sci 2024; 25:6644. [PMID: 38928349 PMCID: PMC11204268 DOI: 10.3390/ijms25126644] [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: 05/02/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
The role of adipose mesenchymal stem cells (Ad-MSCs) in metabolic syndrome remains unclear. We aimed to assess the expression of selected microRNAs in Ad-MSCs of non-diabetic adults in relation to Ad-MSC secretion of protein regulators and basic metabolic parameters. Ten obese, eight overweight, and five normal weight subjects were enrolled: 19 females and 4 males; aged 43.0 ± 8.9 years. Ad-MSCs were harvested from abdominal subcutaneous fat. Ad-MSC cellular expressions of four microRNAs (2-ΔCt values) and concentrations of IL-6, IL-10, VEGF, and IGF-1 in the Ad-MSC-conditioned medium were assessed. The expressions of miR-21, miR-122, or miR-192 did not correlate with clinical parameters (age, sex, BMI, visceral fat, HOMA-IR, fasting glycemia, HbA1c, serum lipids, CRP, and eGFR). Conversely, the expression of miR-155 was lowest in obese subjects (3.69 ± 2.67 × 10-3 vs. 7.07 ± 4.42 × 10-3 in overweight and 10.25 ± 7.05 × 10-3 in normal weight ones, p = 0.04). The expression of miR-155 correlated inversely with BMI (sex-adjusted r = -0.64; p < 0.01), visceral adiposity (r = -0.49; p = 0.03), and serum CRP (r = -0.63; p < 0.01), whereas it correlated positively with serum HDL cholesterol (r = 0.51; p = 0.02). Moreover, miR-155 synthesis was associated marginally negatively with Ad-MSC secretion of IGF-1 (r = -0.42; p = 0.05), and positively with that of IL-10 (r = 0.40; p = 0.06). Ad-MSC expression of miR-155 appears blunted in visceral obesity, which correlates with Ad-MSC IGF-1 hypersecretion and IL-10 hyposecretion, systemic microinflammation, and HDL dyslipidemia. Ad-MSC studies in metabolic syndrome should focus on miR-155.
Collapse
Affiliation(s)
| | - Klaudia Simka-Lampa
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.S.-L.); (E.S.); (T.F.)
| | | | - Ewelina Sobecko
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.S.-L.); (E.S.); (T.F.)
| | - Aleksandra Skubis-Sikora
- Department of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.S.-S.); (B.S.)
| | - Bartosz Sikora
- Department of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.S.-S.); (B.S.)
| | - Ewa Wojtyna
- Institute of Medical Sciences, University of Opole, 45-040 Opole, Poland;
| | - Agnieszka Golda
- Alfamed General Practice, 41-100 Siemianowice Slaskie, Poland;
| | - Katarzyna Gwizdek
- Department of Rehabilitation, Faculty of Health Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Marta Wróbel
- Department of Internal Medicine, Diabetology and Cardiometabolic Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Łukasz Sędek
- Department of Microbiology and Immunology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Sylwia Górczyńska-Kosiorz
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (S.G.-K.); (N.S.-G.); (W.T.)
| | - Nikola Szweda-Gandor
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (S.G.-K.); (N.S.-G.); (W.T.)
| | - Wanda Trautsolt
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (S.G.-K.); (N.S.-G.); (W.T.)
| | - Tomasz Francuz
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (K.S.-L.); (E.S.); (T.F.)
| | - Celina Kruszniewska-Rajs
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (C.K.-R.); (J.G.)
| | - Joanna Gola
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (C.K.-R.); (J.G.)
| |
Collapse
|
13
|
Zhang X, Wang Q, Wang Y, Ma C, Zhao Q, Yin H, Li L, Wang D, Huang Y, Zhao Y, Shi X, Li X, Huang C. Interleukin-6 promotes visceral adipose tissue accumulation during aging via inhibiting fat lipolysis. Int Immunopharmacol 2024; 132:111906. [PMID: 38593501 DOI: 10.1016/j.intimp.2024.111906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/06/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Age-related visceral obesity could contribute to the development of cardiometabolic complications. The pathogenesis of visceral fat mass accumulation during the aging process remains complex and largely unknown. Interleukin-6 (IL-6) has emerged as one of the prominent inflammaging markers which are elevated in circulation during aging. However, the precise role of IL-6 in regulating age-related visceral adipose tissue accumulation remains uncertain. RESULTS A cross-sectional study including 77 older adults (≥65 years of age) was initially conducted. There was a significant positive association between serum IL-6 levels and visceral fat mass. We subsequently validated a modest but significant elevation in serum IL-6 levels in aged mice. Furthermore, we demonstrated that compared to wildtype control, IL-6 deficiency (IL-6 KO) significantly attenuated the accumulation of visceral adipose tissue during aging. Further metabolic characterization suggested that IL-6 deficiency resulted in improved lipid metabolism parameters and energy expenditure in aged mice. Moreover, histological examinations of adipose depots revealed that the absence of IL-6 ameliorated adipocyte hypertrophy in visceral adipose tissue of aged mice. Mechanically, the ablation of IL-6 could promote the PKA-mediated lipolysis and consequently mitigate lipid accumulation in adipose tissue in aged mice. CONCLUSION Our findings identify a detrimental role of IL-6 during the aging process by promoting visceral adipose tissue accumulation through inhibition of lipolysis. Therefore, strategies aimed at preventing or reducing IL-6 levels may potentially ameliorate age-related obesity and improve metabolism during aging.
Collapse
Affiliation(s)
- Xiaofang Zhang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Qingxuan Wang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Yaru Wang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Chen Ma
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Qing Zhao
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Hongyan Yin
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Long Li
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China; Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Dongmei Wang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China; Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen 361023, China
| | - Yinxiang Huang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Yan Zhao
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Xiulin Shi
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Xuejun Li
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China.
| | - Caoxin Huang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China.
| |
Collapse
|
14
|
Taneera J, Khalique A, Mohammed AK, Mussa BM, Sulaiman N, Abu-Gharbieh E, El-Huneidi W, Saber-Ayad MM. Investigating the Impact of IL6 on Insulin Secretion: Evidence from INS-1 Cells, Human Pancreatic Islets, and Serum Analysis. Cells 2024; 13:685. [PMID: 38667300 PMCID: PMC11049194 DOI: 10.3390/cells13080685] [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: 02/26/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Interleukin-6 (IL6) is a pleiotropic cytokine implicated in metabolic disorders and inflammation, yet its precise influence on insulin secretion and glucose metabolism remains uncertain. This study examined IL6 expression in pancreatic islets from individuals with/without diabetes, alongside a series of functional experiments, including siRNA silencing; IL6 treatment; and assessments of glucose uptake, cell viability, apoptosis, and expression of key β-cell genes, which were conducted in both INS-1 cells and human islets to elucidate the effect of IL6 on insulin secretion. Serum levels of IL6 from Emirati patients with type 2 diabetes (T2D) were measured, and the effect of antidiabetic drugs on IL6 levels was studied. The results revealed that IL6 mRNA expression was higher in islets from diabetic and older donors compared to healthy or young donors. IL6 expression correlated negatively with PDX1, MAFB, and NEUROD1 and positively with SOX4, HES1, and FOXA1. Silencing IL6 in INS-1 cells reduced insulin secretion and glucose uptake independently of apoptosis or oxidative stress. Reduced expression of IL6 was associated with the downregulation of Ins, Pdx1, Neurod1, and Glut2 in INS-1 cells. In contrast, IL6 treatment enhanced insulin secretion in INS-1 cells and human islets and upregulated insulin expression. Serum IL6 levels were elevated in patients with T2D and associated with higher glucose, HbA1c, and triglycerides, regardless of glucose-lowering medications. This study provides a new understanding of the role of IL6 in β-cell function and the pathophysiology of T2D. Our data highlight differences in the response to IL6 between INS-1 cells and human islets, suggesting the presence of species-specific variations across different experimental models. Further research is warranted to unravel the precise mechanisms underlying the observed effects of IL-6 on insulin secretion.
Collapse
Affiliation(s)
- Jalal Taneera
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (B.M.M.); (N.S.); (E.A.-G.); (W.E.-H.); (M.M.S.-A.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (A.K.); (A.K.M.)
| | - Anila Khalique
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (A.K.); (A.K.M.)
| | - Abdul Khader Mohammed
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (A.K.); (A.K.M.)
| | - Bashair M. Mussa
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (B.M.M.); (N.S.); (E.A.-G.); (W.E.-H.); (M.M.S.-A.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (A.K.); (A.K.M.)
| | - Nabil Sulaiman
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (B.M.M.); (N.S.); (E.A.-G.); (W.E.-H.); (M.M.S.-A.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (A.K.); (A.K.M.)
| | - Eman Abu-Gharbieh
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (B.M.M.); (N.S.); (E.A.-G.); (W.E.-H.); (M.M.S.-A.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (A.K.); (A.K.M.)
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Waseem El-Huneidi
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (B.M.M.); (N.S.); (E.A.-G.); (W.E.-H.); (M.M.S.-A.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (A.K.); (A.K.M.)
| | - Maha M. Saber-Ayad
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (B.M.M.); (N.S.); (E.A.-G.); (W.E.-H.); (M.M.S.-A.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (A.K.); (A.K.M.)
| |
Collapse
|
15
|
Krupka S, Hoffmann A, Jasaszwili M, Dietrich A, Guiu-Jurado E, Klöting N, Blüher M. Consequences of COVID-19 on Adipose Tissue Signatures. Int J Mol Sci 2024; 25:2908. [PMID: 38474155 DOI: 10.3390/ijms25052908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Since the emergence of coronavirus disease-19 (COVID-19) in 2019, it has been crucial to investigate the causes of severe cases, particularly the higher rates of hospitalization and mortality in individuals with obesity. Previous findings suggest that adipocytes may play a role in adverse COVID-19 outcomes in people with obesity. The impact of COVID-19 vaccination and infection on adipose tissue (AT) is currently unclear. We therefore analyzed 27 paired biopsies of visceral and subcutaneous AT from donors of the Leipzig Obesity BioBank that have been categorized into three groups (1: no infection/no vaccination; 2: no infection but vaccinated; 3: infected and vaccinated) based on COVID-19 antibodies to spike (indicating vaccination) and/or nucleocapsid proteins. We provide additional insights into the impact of COVID-19 on AT biology through a comprehensive histological transcriptome and serum proteome analysis. This study demonstrates that COVID-19 infection is associated with smaller average adipocyte size. The impact of infection on gene expression was significantly more pronounced in subcutaneous than in visceral AT and mainly due to immune system-related processes. Serum proteome analysis revealed the effects of the infection on circulating adiponectin, interleukin 6 (IL-6), and carbonic anhydrase 5A (CA5A), which are all related to obesity and blood glucose abnormalities.
Collapse
Affiliation(s)
- Sontje Krupka
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Mariami Jasaszwili
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Arne Dietrich
- Clinic for Visceral, Transplantation and Thorax and Vascular Surgery, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Esther Guiu-Jurado
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| |
Collapse
|
16
|
Klimczak S, Śliwińska A. Epigenetic regulation of inflammation in insulin resistance. Semin Cell Dev Biol 2024; 154:185-192. [PMID: 36109307 DOI: 10.1016/j.semcdb.2022.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/07/2022] [Accepted: 09/07/2022] [Indexed: 11/20/2022]
Abstract
Epigenetics focuses on the study of changes in gene expression based on modifications that do not interfere with the DNA sequence, such as DNA methylation, post-translational histone modification, and non-coding RNA. Epigenetic changes regulate the expression of many genes, including inflammatory ones. Chronic inflammation is often accompanied by insulin resistance (IR), which is characteristic of inter alia type 2 diabetes. Recently, it has been reported that altered epigenetic signature in the promoter regions of inflammatory genes may contribute to the development of IR. Therefore, the aim of this review is to present the current state of knowledge regarding the epigenetic regulation of inflammation in IR. It includes original papers published from 2014 to 2022. It appears that hypomethylation of the SOCS3 gene increases the risk of IR, while the alteration of H3K4me in the NF-kB promoter promotes changes in inflammatory phenotype. Finally, in hyperglycemic states associated with IR, altered levels of H3K4/K9m3 and H3K9/K14ac result in increased expression of the inflammatory cytokine IL-6. In addition, numerous miRNAs have been identified that may become a target in the fight against diseases related to inflammation and IR. Future studies should examine the epigenetic modifications of IR inflammatory markers associated with environmental factors.
Collapse
Affiliation(s)
- S Klimczak
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland; AllerGen, Center of Personalized Medicine, 97-300 Piotrkow Trybunalski, Poland.
| | - A Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland.
| |
Collapse
|
17
|
Król-Kulikowska M, Urbanowicz I, Kepinska M. The Concentrations of Interleukin-6, Insulin, and Glucagon in the Context of Obesity and Type 2 Diabetes and Single Nucleotide Polymorphisms in IL6 and INS Genes. J Obes 2024; 2024:7529779. [PMID: 38250713 PMCID: PMC10798838 DOI: 10.1155/2024/7529779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/11/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024] Open
Abstract
Obesity and diabetes are a problem of modern medicine. Although the environmental factors contributing to the development of these diseases are widely known, research into genetic factors is still ongoing. At the same time, the role of inflammation in the pathophysiology of obesity and diabetes is increasingly emphasized. Therefore, the purpose of this study was to investigate the influence of two selected polymorphisms (rs1800795 and rs3842729) on the development of obesity and type 2 diabetes. In this study, 118 participants were examined, including a control group (nonobese and nondiabetic group), an obese group, and a diabetic group. Genotype analysis was performed using the PCR-RFLP method. It has been shown that in patients with the G/G genotype within the rs1800795 polymorphism (IL6), the chance of developing type 2 diabetes is several times lower compared to patients with the G/C and C/C genotypes. However, the rs3842729 polymorphism (INS) does not directly affect the risk of obesity or type 2 diabetes (T2D), although elevated insulin concentrations have been observed in obese and diabetic patients. These results confirm the impact of the rs1800795 polymorphism on the development of diabetes; however, this relationship is more complex and requires further research on other factors.
Collapse
Affiliation(s)
- Magdalena Król-Kulikowska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw 50-556, Poland
| | - Iwona Urbanowicz
- Department of Clinical Chemistry and Laboratory Hematology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw 50-556, Poland
| | - Marta Kepinska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw 50-556, Poland
| |
Collapse
|
18
|
Ruhl T, Sessler TM, Keimes JM, Beier JP, Villwock S, Rose M, Dahl E. ITIH5 inhibits proliferation, adipogenic differentiation, and secretion of inflammatory cytokines of human adipose stem cells-A new key in treating obesity? FASEB J 2024; 38:e23352. [PMID: 38095340 DOI: 10.1096/fj.202301366r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/26/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023]
Abstract
Inter-α-trypsin inhibitor heavy chain 5 (ITIH5) is widely expressed in the human body, and it is detected to be particularly abundant in adipose tissue. ITIH5 expression is increased in people with obesity compared to lean persons and is decreased by diet-induced weight loss. This suggests that ITIH5 may be involved in the development of adiposity and clinical metabolic variables, although its exact function remains unknown. We measured the protein concentration of ITIH5 in adipose samples from patients undergoing abdominoplasty and tested for correlation with the subjects' BMI as well as inflammatory mediators. We stimulated human adipose stem cells (ASCs) with recombinant (r)ITIH5 protein and tested for an effect on proliferation, differentiation, and immunosuppressive properties when the cells were exposed to an artificial inflammatory environment. We found positive correlations between ITIH5 levels and the BMI (p < .001) as well as concentrations of inflammatory cytokines (TNF-α, IL-6, and MCP-1) in adipose tissue (p < .01). Application of the rITIH5 protein inhibited both proliferation (p < .001) and differentiation of ASCs. Especially, the development of mature adipocytes was reduced by over 50%. Moreover, rITIH5 decreased the release of IL-6 and MCP-1 when the cells were exposed to TNF-α and IL-1β (p < .001). Our data suggest that ITIH5 is an adipokine that is increasingly released during human adipose tissue development, acting as a regulator that inhibits proliferation and adipogenic differentiation of ASCs. ITIH5 thus presents itself as a positive regulator of adipose tissue homeostasis, possibly protecting against both hyperplasia and hypertrophy of adipose tissue and the associated chronic inflammation.
Collapse
Affiliation(s)
- Tim Ruhl
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Thomas M Sessler
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Jana M Keimes
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Justus P Beier
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Sophia Villwock
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Michael Rose
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Edgar Dahl
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
19
|
Zhang Y, Jia R, Zhang Y, Sun X, Mei Y, Zou R, Niu L, Dong S. Effect of non-surgical periodontal treatment on cytokines/adipocytokines levels among periodontitis patients with or without obesity: a systematic review and meta-analysis. BMC Oral Health 2023; 23:717. [PMID: 37798684 PMCID: PMC10552206 DOI: 10.1186/s12903-023-03383-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 09/04/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND The objective of this systematic review and meta-analysis was to evaluate the effects of non-surgical periodontal therapy (NSPT) on inflammatory-related cytokines/adipocytokines in periodontitis patients with or without obesity. METHODS We followed the preferred reporting items for systematic reviews and meta-analyses statement and registered the study (CRD42022375331) in the Prospective International Register of Systematic Reviews. We screened randomized-controlled trials and controlled clinical trials from six databases up to December 2022. Quality assessment was performed with RoB-2 and ROBINS-I tools for randomized trials and non-randomized trials, respectively. Meta-analysis was carried out using a random-effect model. RESULTS We included seventeen references in the systematic analysis, and sixteen in the meta-analysis. Baseline results of pro-inflammatory biomarkers, including serum interleukin (IL)-6, serum and gingival crevicular fluid (GCF), tumor necrosis factor (TNF)-a, serum C-reactive protein (CRP)/hs-CRP, and serum and GCF resistin, were higher in obesity subjects than in normal weight subjects. The effect of NSPT with respect to levels of cytokines/adipocytokines, including IL-6, TNF-a, CRP/hs-CRP, resistin, adiponectin, leptin and retinol binding protein 4 (RBP4), were then analyzed in the systematic and meta-analysis. After three months of NSPT, serum (MD = -0.54, CI = -0.62 - -0.46), and GCF (MD = -2.70, CI = -4.77 - -0.63) levels of IL-6, along with the serum RBP4 (MD = -0.39, CI = -0.68-0.10) decreased in periodontitis individuals with obesity. NSPT also improved GCF adiponectin levels after three months (MD = 2.37, CI = 0.29 - 4.45) in periodontitis individuals without obesity. CONCLUSIONS Obese status altered the baseline levels of cytokines/adipocytokines (serum IL-6, serum and GCF TNF-a, serum CRP/hs-CRP, and serum and GCF resistin). Then NSPT can shift the levels of specific pro-inflammatory mediators and anti-inflammatory mediators in biological fluids, both in obesity and non-obesity individuals. NSPT can reduce serum and GCF IL-6 levels together with serum RBP4 level in individuals with obesity after 3 months, besides, there is no sufficient evidence to prove that obese patients have a statistically significant decrease in the levels of other cytokines compared to patients with normal weight. NSPT can also increase GCF adiponectin level in normal weight individuals after 3 months. Our findings imply the potential ideal follow-up intervals and sensitive biomarkers for clinical bioanalysis in personalized decision-making of effect of NSPT due to patients' BMI value.
Collapse
Affiliation(s)
- Yuwei Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an, 710004, Shaanxi Province, China
- Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
| | - Ru Jia
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an, 710004, Shaanxi Province, China
- Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
| | - Yifei Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an, 710004, Shaanxi Province, China
- Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
| | - Xuefei Sun
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an, 710004, Shaanxi Province, China
| | - Yukun Mei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an, 710004, Shaanxi Province, China
- Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
| | - Rui Zou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an, 710004, Shaanxi Province, China.
| | - Lin Niu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an, 710004, Shaanxi Province, China.
- Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| | - Shaojie Dong
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an, 710004, Shaanxi Province, China.
- Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| |
Collapse
|
20
|
Caldú X, Prats-Soteras X, García-García I, Prunell-Castañé A, Sánchez-Garre C, Cano N, Tor E, Sender-Palacios MJ, Ottino-González J, Garolera M, Jurado MÁ. Body mass index, systemic inflammation and cognitive performance in adolescents: A cross-sectional study. Psychoneuroendocrinology 2023; 156:106298. [PMID: 37295218 DOI: 10.1016/j.psyneuen.2023.106298] [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: 12/22/2022] [Revised: 05/21/2023] [Accepted: 05/21/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Excessive body weight has been related to lower cognitive performance. One of the mechanisms through which excess body weight may affect cognition is inflammation. HYPOTHESIS Our hypothesis is that both body mass index (BMI) and circulating levels of inflammatory biomarkers will be negatively related to cognitive performance. DESIGN Cross-sectional study. SETTING Users of the public health centres of the Consorci Sanitari de Terrassa (Terrassa, Spain) between 2010 and 2017 aged 12-21 years. PARTICIPANTS One hundred and five adolescents (46 normoweight, 18 overweight, 41 obese). MEASUREMENTS Levels of high sensitivity C-reactive protein, interleukin 6, tumour necrosis factor α (TNFα) and fibrinogen were determined from blood samples. Cognitive performance was evaluated and six cognitive composites were obtained: working memory, cognitive flexibility, inhibitory control, decision-making, verbal memory, and fine motor speed. A single multivariate general lineal model was used to assess the influence of the four inflammatory biomarkers, as well as participants' BMI, sex, and age on the 6 cognitive indexes. RESULTS An inverse relationship between BMI and inhibitory control (F = 5.688, p = .019; β = -0.212, p = .031), verbal memory (F = 5.404, p = .022; β = -0.255, p = .009) and fine motor speed (F = 9.038, p = .003; β = -0.319, p = .001) was observed. Levels of TNFα and fibrinogen were inversely related to inhibitory control (F = 5.055, p = .027; β = -0.226, p = .021) and verbal memory (F = 4.732, p = .032; β = -0.274, p = .005), respectively. LIMITATIONS The cross-sectional nature of the study, the use of cognitive tests designed for clinical purposes, and the use of BMI as a proxy for adiposity are limitations of our study that must be taken into account when interpreting results. CONCLUSIONS Our data indicate that some components of executive functions, together with verbal memory, are sensitive to specific obesity-related inflammatory agents at early ages.
Collapse
Affiliation(s)
- Xavier Caldú
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Pg. Vall d'Hebron, 171, 08035 Barcelona, Spain; Institut de Neurociències, Universitat de Barcelona, Pg. Vall d'Hebron, 171, 08035 Barcelona, Spain; Institut de Recerca Sant Joan de Déu, C/ Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain
| | - Xavier Prats-Soteras
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Pg. Vall d'Hebron, 171, 08035 Barcelona, Spain; Institut de Neurociències, Universitat de Barcelona, Pg. Vall d'Hebron, 171, 08035 Barcelona, Spain; Institut de Recerca Sant Joan de Déu, C/ Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain
| | - Isabel García-García
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Pg. Vall d'Hebron, 171, 08035 Barcelona, Spain; Institut de Neurociències, Universitat de Barcelona, Pg. Vall d'Hebron, 171, 08035 Barcelona, Spain; Clinique la Prairie, Montreux, Rue du Lac 142, 1815 Clarens, Switzerland
| | - Anna Prunell-Castañé
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Pg. Vall d'Hebron, 171, 08035 Barcelona, Spain; Institut de Neurociències, Universitat de Barcelona, Pg. Vall d'Hebron, 171, 08035 Barcelona, Spain; Institut de Recerca Sant Joan de Déu, C/ Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain
| | - Consuelo Sánchez-Garre
- Unitat d'Endocrinologia Pediàtrica, Departament de Pediatria, Hospital de Terrassa, Consorci Sanitari de Terrassa, Ctra Torrebonica s/n, 08227 Terrassa, Spain
| | - Neus Cano
- Unitat de Neuropsicologia, Hospital de Terrassa, Consorci Sanitari de Terrassa, Ctra Torrebonica s/n, 08227 Terrassa, Spain; Brain, Cognition and Behavior Clinical Research Group, Consorci Sanitari de Terrassa, Ctra Torrebonica s/n, 08227 Terrassa, Spain
| | - Encarnació Tor
- Centre d'Atenció Primària Terrassa Nord, Consorci Sanitari de Terrassa, Av del Vallès 451, 08226 Terrassa, Spain
| | - María-José Sender-Palacios
- Centre d'Atenció Primària Terrassa Nord, Consorci Sanitari de Terrassa, Av del Vallès 451, 08226 Terrassa, Spain
| | - Jonatan Ottino-González
- Division of Endocrinology, The Saban Research Institute, Children's Hospital Los Angeles, United States
| | - Maite Garolera
- Unitat de Neuropsicologia, Hospital de Terrassa, Consorci Sanitari de Terrassa, Ctra Torrebonica s/n, 08227 Terrassa, Spain; Brain, Cognition and Behavior Clinical Research Group, Consorci Sanitari de Terrassa, Ctra Torrebonica s/n, 08227 Terrassa, Spain.
| | - María Ángeles Jurado
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Pg. Vall d'Hebron, 171, 08035 Barcelona, Spain; Institut de Neurociències, Universitat de Barcelona, Pg. Vall d'Hebron, 171, 08035 Barcelona, Spain; Institut de Recerca Sant Joan de Déu, C/ Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain
| |
Collapse
|
21
|
Landgraaf RG, Bloem MN, Fumagalli M, Benninga MA, de Lorijn F, Nieuwdorp M. Acupuncture as multi-targeted therapy for the multifactorial disease obesity: a complex neuro-endocrine-immune interplay. Front Endocrinol (Lausanne) 2023; 14:1236370. [PMID: 37795371 PMCID: PMC10545882 DOI: 10.3389/fendo.2023.1236370] [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: 06/07/2023] [Accepted: 08/29/2023] [Indexed: 10/06/2023] Open
Abstract
The prevalence of obesity has reached pandemic dimensions. It is associated with multiple comorbidities and is becoming a clinical and public health threat. Obesity is a multifactorial disease with a complex pathophysiology and interplay of various systems. A strong interplay exists between the neuro-endocrine system, the immune system with systemic chronic low-grade inflammation, and microbiome dysbiosis that can lead to the development of obesity, which in turn can exacerbate each of these factors, hence creating a vicious cycle. The conventional treatment with lifestyle modifications such as diet, physical exercise, pharmacotherapy, and bariatric surgery does not always result in sufficient weight control thus paving the way for other strategies. As one such strategy, acupuncture is increasingly used worldwide to treat obesity. This narrative review outlines the evidence for this neuro-endocrine-immune interplay in the pathophysiology of obesity. Furthermore, the existing experimental and clinical evidence of acupuncture as a multi-targeted therapy for obesity is explained and future research perspectives are discussed.
Collapse
Affiliation(s)
- Raymond Guy Landgraaf
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Amsterdam, Netherlands
- Sinomedica Gui Sheng Tang, Scientific Department, Lugano, Switzerland
| | - Michelle Nicté Bloem
- Emma Children’s Hospital, Amsterdam University Medical Center (UMC), Pediatric Gastroenterology, University of Amsterdam, Amsterdam, Netherlands
| | - Massimo Fumagalli
- Sinomedica Gui Sheng Tang, Scientific Department, Lugano, Switzerland
| | - Marc Alexander Benninga
- Emma Children’s Hospital, Amsterdam University Medical Center (UMC), Pediatric Gastroenterology, University of Amsterdam, Amsterdam, Netherlands
| | - Fleur de Lorijn
- Emma Children’s Hospital, Amsterdam University Medical Center (UMC), Pediatric Gastroenterology, University of Amsterdam, Amsterdam, Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Amsterdam, Netherlands
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
22
|
Mazitova AM, Márquez-Sánchez AC, Koltsova EK. Fat and inflammation: adipocyte-myeloid cell crosstalk in atherosclerosis. Front Immunol 2023; 14:1238664. [PMID: 37781401 PMCID: PMC10540690 DOI: 10.3389/fimmu.2023.1238664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Adipose tissue inflammation has been implicated in various chronic inflammatory diseases and cancer. Perivascular adipose tissue (PVAT) surrounds the aorta as an extra layer and was suggested to contribute to atherosclerosis development. PVAT regulates the function of endothelial and vascular smooth muscle cells in the aorta and represent a reservoir for various immune cells which may participate in aortic inflammation. Recent studies demonstrate that adipocytes also express various cytokine receptors and, therefore, may directly respond to inflammatory stimuli. Here we will summarize current knowledge on immune mechanisms regulating adipocyte activation and the crosstalk between myeloid cells and adipocytes in pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Aleksandra M. Mazitova
- Cedars-Sinai Cancer, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ana Cristina Márquez-Sánchez
- Cedars-Sinai Cancer, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ekaterina K. Koltsova
- Cedars-Sinai Cancer, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
23
|
Ren J, Wang XQ, Nakao T, Libby P, Shi GP. Differential Roles of Interleukin-6 in Severe Acute Respiratory Syndrome-Coronavirus-2 Infection and Cardiometabolic Diseases. CARDIOLOGY DISCOVERY 2023; 3:166-182. [PMID: 38152628 PMCID: PMC10750760 DOI: 10.1097/cd9.0000000000000096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection can lead to a cytokine storm, unleashed in part by pyroptosis of virus-infected macrophages and monocytes. Interleukin-6 (IL-6) has emerged as a key participant in this ominous complication of COVID-19. IL-6 antagonists have improved outcomes in patients with COVID-19 in some, but not all, studies. IL-6 signaling involves at least 3 distinct pathways, including classic-signaling, trans-signaling, and trans-presentation depending on the localization of IL-6 receptor and its binding partner glycoprotein gp130. IL-6 has become a therapeutic target in COVID-19, cardiovascular diseases, and other inflammatory conditions. However, the efficacy of inhibition of IL-6 signaling in metabolic diseases, such as obesity and diabetes, may depend in part on cell type-dependent actions of IL-6 in controlling lipid metabolism, glucose uptake, and insulin sensitivity owing to complexities that remain to be elucidated. The present review sought to summarize and discuss the current understanding of how and whether targeting IL-6 signaling ameliorates outcomes following SARS-CoV-2 infection and associated clinical complications, focusing predominantly on metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Jingjing Ren
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Xiao-Qi Wang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Tetsushi Nakao
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Peter Libby
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
24
|
Lundqvist MH, Pereira MJ, Eriksson JW. Glucose-dependent inflammatory responses in obese compared to lean individuals. Endocrine 2023; 81:464-476. [PMID: 37400734 PMCID: PMC10403442 DOI: 10.1007/s12020-023-03433-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/15/2023] [Indexed: 07/05/2023]
Abstract
PURPOSE Obesity is characterized by chronic inflammation that may contribute to insulin resistance and promote type 2 diabetes. We have investigated whether inflammatory responses to glycemic and insulinemic variations are altered in obese individuals. METHODS Eight obese and eight lean individuals without diabetes had undergone hyperinsulinemic-euglycemic-hypoglycemic and hyperglycemic clamps in a previous study. Using Proximity Extension Assay, 92 inflammatory markers were analyzed from plasma samples at fasting, hyperinsulinemia-euglycemia, hypoglycemia and hyperglycemia. RESULTS In all participants, hyperinsulinemia, hypoglycemia and hyperglycemia led to reductions of 11, 19 and 62 out of the 70 fully evaluable biomarkers, respectively. FGF-21 increased during both hypoglycemia and hyperglycemia while IL-6 and IL-10 increased during hypoglycemia. In obese vs lean participants, Oncostatin-M, Caspase-8 and 4E-BP1 were more markedly suppressed during hypoglycemia, whereas VEGF-A was more markedly suppressed during hyperglycemia. BMI correlated inversely with changes of PD-L1 and CD40 during hyperinsulinemia, Oncostatin-M, TNFSF14, FGF-21 and 4EBP-1 during hypoglycemia and CCL23, VEGF-A and CDCP1 during hyperglycemia (Rho ≤ -0.50). HbA1c correlated positively with changes of MCP-2 and IL-15-RA during hyperinsulinemia (Rho ≥ 0.51) and inversely with changes of CXCL1, MMP-1 and Axin-1 during hypoglycemia (Rho ≤ -0.55). M-value correlated positively with changes of IL-12B and VEGF-A during hyperglycemia (Rho ≥ 0.51). Results above were significant (p < 0.05). CONCLUSION Overall, hyperinsulinemia, hypo- and hyperglycemia led to suppression of several inflammatory markers and this tended to be more marked in individuals with obesity, insulin resistance and dysglycemia. Thus, acute glycemic or insulinemic variations do not seem to potentiate possible inflammatory pathways in the development of insulin resistance and disturbed glucose metabolism.
Collapse
Affiliation(s)
- Martin H Lundqvist
- Clinical Diabetology and Metabolism, Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| | - Maria J Pereira
- Clinical Diabetology and Metabolism, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
25
|
Fu X, Wang Y, Zhao F, Cui R, Xie W, Liu Q, Yang W. Shared biological mechanisms of depression and obesity: focus on adipokines and lipokines. Aging (Albany NY) 2023; 15:5917-5950. [PMID: 37387537 PMCID: PMC10333059 DOI: 10.18632/aging.204847] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
Depression and obesity are both common disorders currently affecting public health, frequently occurring simultaneously within individuals, and the relationship between these disorders is bidirectional. The association between obesity and depression is highly co-morbid and tends to significantly exacerbate metabolic and related depressive symptoms. However, the neural mechanism under the mutual control of obesity and depression is largely inscrutable. This review focuses particularly on alterations in systems that may mechanistically explain the in vivo homeostatic regulation of the obesity and depression link, such as immune-inflammatory activation, gut microbiota, neuroplasticity, HPA axis dysregulation as well as neuroendocrine regulators of energy metabolism including adipocytokines and lipokines. In addition, the review summarizes potential and future treatments for obesity and depression and raises several questions that need to be answered in future research. This review will provide a comprehensive description and localization of the biological connection between obesity and depression to better understand the co-morbidity of obesity and depression.
Collapse
Affiliation(s)
- Xiying Fu
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun 130041, P.R. China
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yicun Wang
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Fangyi Zhao
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Ranji Cui
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Wei Xie
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Qianqian Liu
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Wei Yang
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
- Department of Neurology, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| |
Collapse
|
26
|
Štšepetova J, Rätsep M, Gerulis O, Jõesaar A, Mikelsaar M, Songisepp E. Impact of Lactiplantibacillus plantarum Inducia on metabolic and antioxidative response in cholesterol and BMI variable indices: randomised, double-blind, placebo-controlled trials. Benef Microbes 2023; 14:1-16. [PMID: 36437811 DOI: 10.3920/bm2022.0030] [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] [Indexed: 11/29/2022]
Abstract
Probiotics may have potential in reducing cardiovascular disease (CVD) risk in middle-aged persons with borderline metabolic indices. The ability of potential probiotic Lactiplantibacillus plantarum Inducia to reduce CVD risk factors in persons with variable cholesterol and body mass indices (BMI) was assessed. In two parallel-armed double-blind placebo-controlled interventions (n=136) and (n=104), participants daily received either test yoghurt (Inducia) or placebo yoghurt. BMI, blood pressure, plasma glucose, cholesterol, high-sensitivity C-reactive protein (hs-CRP), oxidative stress and immunological markers were measured. Total counts of lactobacilli and L. plantarum Inducia were evaluated using real-time PCR. Significant reduction of total cholesterol, low density lipoprotein cholesterol (LDL-c) and non-high-density cholesterol occurred in both trials. The change in cholesterol (P=0.023) in persons with normal BMI and borderline cholesterol levels after four weeks of yoghurt consumption was detected. A difference was also found between placebo and test yoghurt groups (P=0.042) in LDL-c with normal BMI. Blood glucose reduction (P=0.01) and antioxidative effect was detected in overweight volunteers of the test yoghurt group. The suppression of oxidised LDL was associated with lowered oxidative stress index and total peroxide concentration values and faecal recovery of Inducia. The Inducia strain expresses antioxidative effect on blood lipids and has anti-glycaemic impact that allow to apply it as dietary probiotic supplement for the management of CVD risks in humans.
Collapse
Affiliation(s)
- J Štšepetova
- BioCC OÜ, 1 Kreutzwaldi, 51006 Tartu, Estonia
- Institute of Biomedicine and Translational Medicine, Department of Microbiology, University of Tartu, 19 Ravila, 50411 Tartu, Estonia
| | - M Rätsep
- BioCC OÜ, 1 Kreutzwaldi, 51006 Tartu, Estonia
| | - O Gerulis
- BioCC OÜ, 1 Kreutzwaldi, 51006 Tartu, Estonia
| | - A Jõesaar
- BioCC OÜ, 1 Kreutzwaldi, 51006 Tartu, Estonia
| | - M Mikelsaar
- BioCC OÜ, 1 Kreutzwaldi, 51006 Tartu, Estonia
- Institute of Biomedicine and Translational Medicine, Department of Microbiology, University of Tartu, 19 Ravila, 50411 Tartu, Estonia
| | - E Songisepp
- BioCC OÜ, 1 Kreutzwaldi, 51006 Tartu, Estonia
| |
Collapse
|
27
|
Xiao L, Mochizuki M, Wang D, Shimamura N, Sunada K, Nakahara T. Types of cell culture inserts affect cell crosstalk between co-cultured macrophages and adipocytes. Biochem Biophys Res Commun 2023; 658:10-17. [PMID: 37011478 DOI: 10.1016/j.bbrc.2023.03.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023]
Abstract
Cell culture inserts offer an in vivo-like microenvironment to investigate cell-cell interactions between co-cultivated cells. However, it is unclear if types of inserts affect cell crosstalk. Here, we developed an environment-friendly cell culture insert, XL-insert, which can reduce plastic waste with lower cost. We compared XL insert with two types of commercial disposable culture inserts, Koken® insert with atelocollagen membrane (Col-inserts) and Falcon® inserts with plastic membrane (PET-inserts) on cell-cell interactions in co-cultivated THP-1 macrophages and OP9 adipocytes. Scanning electron microscope, immunoassay and imaging analysis showed that among three types of inserts, XL-inserts allowed cytokines from co-cultivated macrophages and adipocytes to diffuse freely and offered preferable in vivo-like microenvironment for cell-cell interactions. PET-inserts showed limitations for intercellular communication due to some pores being blocked by somas on the membrane that caused much lower permeability for cytokines passing through. Col-inserts blocked large sized cytokines but allowed small sized molecules to permeate resulting in improved lipid accumulation and adiponectin secretion in OP9 adipocytes. Taken together, our data demonstrated that membrane type and pore size on the membrane affect the cross-talk between co-cultivated cells very differently. Some previous co-culture studies might have different results if the inserts were changed.
Collapse
Affiliation(s)
- Li Xiao
- Department of Pharmacology, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan.
| | - Mai Mochizuki
- Department of Life Science Dentistry, The Nippon Dental University, Tokyo, Japan; Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan.
| | - Dongliang Wang
- Beijing Xiaoxiandun Biotechnology Co., Ltd., No. 150, Guanzhuang Road, Changying Town, Chaoyang District, Beijing, 100020, China; Hebei Edible Bird's Nest Fresh Stew Technology Innovation Center, Bazhou Economic Development Zone, Langfang, 065700, China.
| | - Naohiro Shimamura
- Department of Dental Anesthesiology, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan.
| | - Katsuhisa Sunada
- Department of Dental Anesthesiology, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan.
| | - Taka Nakahara
- Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan.
| |
Collapse
|
28
|
Li W, Chen W. Weight cycling based on altered immune microenvironment as a result of metaflammation. Nutr Metab (Lond) 2023; 20:13. [PMID: 36814270 PMCID: PMC9945679 DOI: 10.1186/s12986-023-00731-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
As a result of the obesity epidemic, more people are concerned about losing weight; however, weight regain is common, leading to repeated weight loss and weight cycling. The health benefits of early weight loss are nullified by weight regain after weight cycling, which has much more severe metabolic consequences. Weight cycling alters body composition, resulting in faster fat recovery and slower muscle reconstruction. This evident fat accumulation, muscle loss, and ectopic fat deposition destroy the intestinal barrier, increase the permeability of the small intestinal epithelium, and cause the lipotoxicity of lipid metabolites and toxins to leak into extraintestinal tissues and circulation. It causes oxidative stress and hypoxia in local tissues and immune cell infiltration in various tissues, all contributing to the adaptation to this metabolic change. Immune cells transmit inflammatory responses in adipose and skeletal muscle tissue by secreting cytokines and adipokines, which mediate immune cell pathways and cause metaflammation and inefficient metabolic degradation. In this review, we focus on the regulatory function of the immunological microenvironment in the final metabolic outcome, with a particular emphasis on the cellular and molecular processes of local and systemic metaflammation induced by weight cycling-induced changes in body composition. Metaflammation in adipose and muscle tissues that is difficult to relieve may cause weight cycling. As this chronic low-grade inflammation spreads throughout the body, metabolic complications associated with weight cycling are triggered. Inhibiting the onset and progression of metabolic inflammation and enhancing the immune microenvironment of adipose and muscle tissues may be the first step in addressing weight cycling.
Collapse
Affiliation(s)
- Wanyang Li
- grid.413106.10000 0000 9889 6335Department of Clinical Nutrition, Chinese Academy of Medical Sciences - Peking Union Medical College, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730 China
| | - Wei Chen
- Department of Clinical Nutrition, Chinese Academy of Medical Sciences - Peking Union Medical College, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
29
|
Long F, Bhatti MR, Kellenberger A, Sun W, Modica S, Höring M, Liebisch G, Krieger JP, Wolfrum C, Challa TD. A low-carbohydrate diet induces hepatic insulin resistance and metabolic associated fatty liver disease in mice. Mol Metab 2023; 69:101675. [PMID: 36682412 PMCID: PMC9900440 DOI: 10.1016/j.molmet.2023.101675] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVES Metabolic-associated fatty liver disease (MAFLD) is the most common chronic liver disease that can range from hepatic steatosis to non-alcoholic steatohepatitis (NASH), which can lead to fibrosis and cirrhosis. Recently, ketogenic diet (KD), a low carbohydrate diet, gained popularity as a weight-loss approach, although it has been reported to induce hepatic insulin resistance and steatosis in animal model systems via an undefined mechanism. Herein, we investigated the KD metabolic benefits and its contribution to the pathogenesis of NASH. METHODS Using metabolic, biochemical and omics approaches, we identified the effects of a KD on NASH and investigated the mechanisms by which KD induces hepatic insulin resistance and steatosis. RESULTS We demonstrate that KD can induce fibrosis and NASH regardless of body weight loss compared to high-fat diet (HFD) fed mice at thermoneutrality. At ambient temperature (23 °C), KD-fed mice develop a severe hepatic injury, inflammation, and steatosis. In addition, KD increases liver cholesterol, IL-6, and p-JNK and aggravates diet induced-glucose intolerance and hepatic insulin resistance compared to HFD. Pharmacological inhibition of IL-6 and JNK reverses KD-induced glucose intolerance, and hepatic steatosis and restores insulin sensitivity. CONCLUSIONS Our studies uncover a new mechanism for KD-induced hepatic insulin resistance and NASH potentially via IL-6-JNK signaling and provide a new NASH mouse model.
Collapse
Affiliation(s)
- Fen Long
- Institute of Food Nutrition and Health and Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), CH-8603 Schwerzenbach, Switzerland
| | - Memoona R. Bhatti
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 75/B1-7503, Brussels 1200, Belgium
| | - Alexandra Kellenberger
- Institute of Food Nutrition and Health and Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), CH-8603 Schwerzenbach, Switzerland
| | - Wenfei Sun
- Institute of Food Nutrition and Health and Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), CH-8603 Schwerzenbach, Switzerland
| | - Salvatore Modica
- Institute of Food Nutrition and Health and Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), CH-8603 Schwerzenbach, Switzerland
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, 93053 Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, 93053 Regensburg, Germany
| | - Jean-Philippe Krieger
- Department of Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Christian Wolfrum
- Institute of Food Nutrition and Health and Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), CH-8603 Schwerzenbach, Switzerland.
| | - Tenagne D. Challa
- Institute of Food Nutrition and Health and Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), CH-8603 Schwerzenbach, Switzerland,Corresponding author. Eidgenössische Technische Hochschule Zürich (ETH, Zürich), Department of Health Sciences and Technology, Schorenstrasse 16, CH-8603 Schwerzenbach, Switzerland.
| |
Collapse
|
30
|
Pita-Grisanti V, Dubay K, Lahooti A, Badi N, Ueltschi O, Gumpper-Fedus K, Hsueh HY, Lahooti I, Chavez-Tomar M, Terhorst S, Knoblaugh SE, Cao L, Huang W, Coss CC, Mace TA, Choueiry F, Hinton A, Mitchell JM, Schmandt R, Grinsfelder MO, Basen-Engquist K, Cruz-Monserrate Z. Physical Activity Delays Obesity-Associated Pancreatic Ductal Adenocarcinoma in Mice and Decreases Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.03.521203. [PMID: 36711764 PMCID: PMC9881853 DOI: 10.1101/2023.01.03.521203] [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] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Obesity is a risk factor for pancreatic ductal adenocarcinoma (PDAC), a deadly disease with limited preventive strategies. Lifestyle interventions to decrease obesity might prevent obesity-associated PDAC. Here, we examined whether decreasing obesity by increased physical activity (PA) and/or dietary changes would decrease inflammation in humans and prevent PDAC in mice. METHODS Circulating inflammatory-associated cytokines of overweight and obese subjects before and after a PA intervention were compared. PDAC pre-clinical models were exposed to PA and/or dietary interventions after obesity-associated cancer initiation. Body composition, tumor progression, growth, fibrosis, inflammation, and transcriptomic changes in the adipose tissue were evaluated. RESULTS PA decreased the levels of systemic inflammatory cytokines in overweight and obese subjects. PDAC mice on a diet-induced obesity (DIO) and PA intervention, had delayed weight gain, decreased systemic inflammation, lower grade pancreatic intraepithelial neoplasia lesions, reduced PDAC incidence, and increased anti-inflammatory signals in the adipose tissue compared to controls. PA had additional cancer prevention benefits when combined with a non-obesogenic diet after DIO. However, weight loss through PA alone or combined with a dietary intervention did not prevent tumor growth in an orthotopic PDAC model. Adipose-specific targeting of interleukin (IL)-15, an anti-inflammatory cytokine induced by PA in the adipose tissue, slowed PDAC growth. CONCLUSIONS PA alone or combined with diet-induced weight loss delayed the progression of PDAC and reduced systemic and adipose inflammatory signals. Therefore, obesity management via dietary interventions and/or PA, or modulating weight loss related pathways could prevent obesity-associated PDAC in high-risk obese individuals.
Collapse
Affiliation(s)
- Valentina Pita-Grisanti
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
- The Ohio State University Interdisciplinary Nutrition Program, The Ohio State University, Columbus, OH
| | - Kelly Dubay
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Ali Lahooti
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Niharika Badi
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Olivia Ueltschi
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Kristyn Gumpper-Fedus
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Hsiang-Yin Hsueh
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
- The Ohio State University Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH
| | - Ila Lahooti
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Myrriah Chavez-Tomar
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Samantha Terhorst
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Sue E. Knoblaugh
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH
| | - Lei Cao
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Wei Huang
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Thomas A. Mace
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Fouad Choueiry
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Alice Hinton
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH
| | - Jennifer M Mitchell
- Department of Veterinary Medicine and Surgery, UT MD Anderson Cancer Center, Houston, TX
| | - Rosemarie Schmandt
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, UT MD Anderson Cancer Center, Houston, TX
| | - Michaela Onstad Grinsfelder
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, UT MD Anderson Cancer Center, Houston, TX
| | - Karen Basen-Engquist
- Department of Behavioral Science, Center for Energy Balance, The University of Texas MD Anderson Cancer Center, UT MD Anderson Cancer Center, Houston, TX
| | - Zobeida Cruz-Monserrate
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| |
Collapse
|
31
|
Barros D, Marques EA, Magalhães J, Carvalho J. Energy metabolism and frailty: The potential role of exercise-induced myokines - A narrative review. Ageing Res Rev 2022; 82:101780. [PMID: 36334911 DOI: 10.1016/j.arr.2022.101780] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Frailty is a complex condition that emerges from dysregulation in multiple physiological systems. Increasing evidence suggests the potential role of age-related energy dysregulation as a key driver of frailty. Exercise is considered the most efficacious intervention to prevent and even ameliorate frailty as it up-tunes and improves the function of several related systems. However, the mechanisms and molecules responsible for these intersystem benefits are not fully understood. The skeletal muscle is considered a secretory organ with endocrine functions that can produce and secrete exercise-related molecules such as myokines. These molecules are cytokines and other peptides released by muscle fibers in response to acute and/or chronic exercise. The available evidence supports that several myokines can elicit autocrine, paracrine, or endocrine effects, partly mediating inter-organ crosstalk and also having a critical role in improving cardiovascular, metabolic, immune, and neurological health. This review describes the current evidence about the potential link between energy metabolism dysregulation and frailty and provides a theoretical framework for the potential role of myokines (via exercise) in counteracting frailty. It also summarizes the physiological role of selected myokines and their response to different acute and chronic exercise protocols in older adults.
Collapse
Affiliation(s)
- Duarte Barros
- The Research Centre in Physical Activity, Health and Leisure, CIAFEL, University of Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal.
| | - Elisa A Marques
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University of Maia (ISMAI), Portugal; School of Sport and Exercise Sciences, Loughborough University, Loughborough, UK
| | - José Magalhães
- The Research Centre in Physical Activity, Health and Leisure, CIAFEL, University of Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Joana Carvalho
- The Research Centre in Physical Activity, Health and Leisure, CIAFEL, University of Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| |
Collapse
|
32
|
Gutierrez AD, Gao Z, Hamidi V, Zhu L, Saint Andre KB, Riggs K, Ruscheinsky M, Wang H, Yu Y, Miller C, Vasquez H, Taegtmeyer H, Kolonin MG. Anti-diabetic effects of GLP1 analogs are mediated by thermogenic interleukin-6 signaling in adipocytes. Cell Rep Med 2022; 3:100813. [PMID: 36384099 PMCID: PMC9729831 DOI: 10.1016/j.xcrm.2022.100813] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 09/06/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022]
Abstract
Mechanisms underlying anti-diabetic effects of GLP1 analogs remain incompletely understood. We observed that in prediabetic humans exenatide treatment acutely induces interleukin-6 (IL-6) secretion by monocytes and IL-6 in systemic circulation. We hypothesized that GLP1 analogs signal through IL-6 in adipose tissue (AT) and used the mouse model to test if IL-6 receptor (IL-6R) signaling underlies the effects of the GLP1-IL-6 axis. We show that liraglutide transiently increases IL-6 in mouse circulation and IL-6R signaling in AT. Metronomic liraglutide treatment resulted in AT browning and thermogenesis linked with STAT3 activation. IL-6-blocking antibody treatment inhibited STAT3 activation in AT and suppressed liraglutide-induced increase in thermogenesis and glucose utilization. We show that adipose IL-6R knockout mice still display liraglutide-induced weight loss but lack thermogenic adipocyte browning and metabolism activation. We conclude that the anti-diabetic effects of GLP1 analogs are mediated by transient upregulation of IL-6, which activates canonical IL-6R signaling and thermogenesis.
Collapse
Affiliation(s)
- Absalon D Gutierrez
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, The University of Texas Health Science Center, Houston, TX 77030, USA.
| | - Zhanguo Gao
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Vala Hamidi
- Department of Medicine, Division of Endocrinology, University of California San Diego, La Jolla, CA 92093, USA
| | - Liang Zhu
- Department of Internal Medicine, Division of Clinical and Translational Sciences, The University of Texas Health Science Center, Houston, TX 77030, USA
| | | | - Kayla Riggs
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern, Dallas, TX 75225, USA
| | - Monika Ruscheinsky
- Department of Pathology, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Hongyu Wang
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Charles Miller
- Department of Cardiothoracic and Vascular Surgery, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Hernan Vasquez
- Department of Internal Medicine, Division of Cardiovascular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Heinrich Taegtmeyer
- Department of Internal Medicine, Division of Cardiovascular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA.
| |
Collapse
|
33
|
The association of measures of body shape and adiposity with incidence of cardiometabolic disease from an ageing perspective. GeroScience 2022; 45:463-476. [PMID: 36129566 PMCID: PMC9886769 DOI: 10.1007/s11357-022-00654-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/31/2022] [Indexed: 02/03/2023] Open
Abstract
While obesity increases the risk of developing cardiometabolic diseases (CMDs), these associations seem to attenuate with increasing age, albeit studied poorly. The present study aimed to investigate the associations between adiposity and CMDs in sex-specific groups of chronological age and leukocyte telomere length (LTL) as a measure of biological age. We investigated the associations between BMI, a body shape index, waist-to-hip ratio (adjusted for BMI) and total body fat, and incident coronary artery disease (CAD), type 2 diabetes (T2D) and ischemic stroke (IS) in 413,017 European-ancestry participants of the UK Biobank without CMD at baseline. We assessed the change in the associations between adiposity and CMD over strata of increasing chronological age or decreasing LTL. Participants (56% women) had a median (IQR) age of 57.0 (50.0-63.0) years. The median follow-up time was 12 years. People with higher BMI had a higher risk of incident CAD (HR 1.14 (95% confidence interval [CI] 1.13, 1.16)), T2D (HR 1.70 (95% CI 1.68, 1.72)) and IS (HR 1.09 (95% CI 1.06, 1.12)). In groups based on chronological age and LTL, adiposity measures were associated with higher risk of CAD and T2D in both men and women, but these associations attenuated with increasing chronological age (Pinteractions < 0.001), but not with decreasing LTL (Pinteraction men = 0.85; Pinteraction women = 0.27). Increased (abdominal) adiposity was associated with higher risk of incident CMDs, which attenuated with increasing chronological age but not with decreasing LTL. Future research may validate these findings using different measures of biological age.
Collapse
|
34
|
To Explore the Molecular Mechanism of Acupuncture Alleviating Inflammation and Treating Obesity Based on Text Mining. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3133096. [PMID: 36105933 PMCID: PMC9467717 DOI: 10.1155/2022/3133096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/03/2022] [Accepted: 08/24/2022] [Indexed: 11/29/2022]
Abstract
Objective To explore the related mechanism of acupuncture affecting obesity by regulating inflammation using bioinformatics methods. Methods The genes related to obesity, inflammation, and acupuncture and inflammation were mined using GenCLiP 3, and the intersecting genes were extracted using Venn diagram. The DAVID database was employed for pathway enrichment analysis and functional annotation of coexpressed genes. Then, the protein-protein interaction (PPI) network was constructed with the STRING database and visualized by the Cytoscape software and screened out important hub genes. Finally, the Boxplot and Survival Analysis of the hub genes in various cancers were performed by GEPIA. Results 755 genes related to obesity and inflammation and 38 genes related to acupuncture and inflammation were identified, and 24 coexpressed genes related to obesity, inflammation, and acupuncture were extracted from the Venn diagram. Eight hub genes including interleukin-6 (IL-6), interleukin-10 (IL-10), Toll-like receptor 4 (TLR4), signal transduction and transcriptional activation factor 3 (STAT3), C-X-C motif chemokine 10 (CXCL10), interleukin-17A (IL-17A), prostaglandin peroxide synthesis-2 (PTGS2), signal transistors, and transcriptional activation factor 6 (STAT6) were identified by gene ontology (GO), Kyoto Encyclopedia of Genes (KEGG), and PPI network analysis. Among them, IL-6 is suggested to play an essential role in the treatment of obesity and inflammation by acupuncture, and IL-6 was significant in both Boxplot and Survival Analysis of pancreatic cancer (PAAD). Therefore, in this study, the core gene, IL-6 was used as the breakthrough point to explore the possible mechanism of acupuncture in treating obesity and pancreatic cancer by regulating IL-6. Conclusion (1) Acupuncture can regulate the expression of IL-6 through the TLR4/nuclear factor-κB (NF-κB) pathway, thereby alleviating inflammation, which can be used as a potential strategy for the treatment of obesity. (2) IL-6/STAT3 is closely related to the occurrence, development, and metastasis of pancreatic cancer. Acupuncture affecting pancreatic cancer through TLR4/NF-κB/IL-6/STAT3 pathway may be a potential method for the treatment of pancreatic cancer.
Collapse
|
35
|
Selman A, Burns S, Reddy AP, Culberson J, Reddy PH. The Role of Obesity and Diabetes in Dementia. Int J Mol Sci 2022; 23:ijms23169267. [PMID: 36012526 PMCID: PMC9408882 DOI: 10.3390/ijms23169267] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 12/06/2022] Open
Abstract
Chronic conditions such as obesity, diabetes, and dementia are increasing in the United States (US) population. Knowledge of these chronic conditions, preventative measures, and proper management tactics is important and critical to preventing disease. The overlap between obesity, diabetes, and dementia is becoming further elucidated. These conditions share a similar origin through the components of increasing age, gender, genetic and epigenetic predispositions, depression, and a high-fat Western diet (WD) that all contribute to the inflammatory state associated with the development of obesity, diabetes, and dementia. This inflammatory state leads to the dysregulation of food intake and insulin resistance. Obesity is often the cornerstone that leads to the development of diabetes and, subsequently, in the case of type 2 diabetes mellitus (T2DM), progression to “type 3 diabetes mellitus (T3DM)”. Obesity and depression are closely associated with diabetes. However, dementia can be avoided with lifestyle modifications, by switching to a plant-based diet (e.g., a Mediterranean diet (MD)), and increasing physical activity. Diet and exercise are not the only treatment options. There are several surgical and pharmacological interventions available for prevention. Current and future research within each of these fields is warranted and offers the chance for new treatment options and a better understanding of the pathogenesis of each condition.
Collapse
Affiliation(s)
- Ashley Selman
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Scott Burns
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Arubala P. Reddy
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - John Culberson
- Department of Family Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Correspondence: ; Tel.: +1-806-743-3194
| |
Collapse
|
36
|
Rogal J, Roosz J, Teufel C, Cipriano M, Xu R, Eisler W, Weiss M, Schenke‐Layland K, Loskill P. Autologous Human Immunocompetent White Adipose Tissue-on-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104451. [PMID: 35466539 PMCID: PMC9218765 DOI: 10.1002/advs.202104451] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/03/2022] [Indexed: 05/07/2023]
Abstract
Obesity and associated diseases, such as diabetes, have reached epidemic proportions globally. In this era of "diabesity", white adipose tissue (WAT) has become a target of high interest for therapeutic strategies. To gain insights into mechanisms of adipose (patho-)physiology, researchers traditionally relied on animal models. Leveraging Organ-on-Chip technology, a microphysiological in vitro model of human WAT is introduced: a tailored microfluidic platform featuring vasculature-like perfusion that integrates 3D tissues comprising all major WAT-associated cellular components (mature adipocytes, organotypic endothelial barriers, stromovascular cells including adipose tissue macrophages) in an autologous manner and recapitulates pivotal WAT functions, such as energy storage and mobilization as well as endocrine and immunomodulatory activities. A precisely controllable bottom-up approach enables the generation of a multitude of replicates per donor circumventing inter-donor variability issues and paving the way for personalized medicine. Moreover, it allows to adjust the model's degree of complexity via a flexible mix-and-match approach. This WAT-on-Chip system constitutes the first human-based, autologous, and immunocompetent in vitro adipose tissue model that recapitulates almost full tissue heterogeneity and can become a powerful tool for human-relevant research in the field of metabolism and its associated diseases as well as for compound testing and personalized- and precision medicine applications.
Collapse
Affiliation(s)
- Julia Rogal
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGBNobelstr. 12Stuttgart70569Germany
| | - Julia Roosz
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
| | - Claudia Teufel
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| | - Madalena Cipriano
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- 3R‐Center for In vitro Models and Alternatives to Animal TestingEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| | - Raylin Xu
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGBNobelstr. 12Stuttgart70569Germany
- Harvard Medical School (HMS)25 Shattuck StBostonMA02115USA
| | - Wiebke Eisler
- Clinic for PlasticReconstructiveHand and Burn SurgeryBG Trauma CenterEberhard Karls University TübingenSchnarrenbergstraße 95Tübingen72076Germany
| | - Martin Weiss
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- Department of Women's HealthEberhard Karls University TübingenCalwerstrasse 7Tübingen72076Germany
| | - Katja Schenke‐Layland
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- Department of Medicine/CardiologyCardiovascular Research LaboratoriesDavid Geffen School of Medicine at UCLA675 Charles E. Young Drive South, MRL 3645Los AngelesCA90095USA
- Cluster of Excellence iFIT (EXC2180) “Image‐Guided and Functionally Instructed Tumor Therapies”Eberhard Karls University TuebingenRöntgenweg 11Tuebingen72076Germany
- Department for Medical Technologies and Regenerative MedicineInstitute of Biomedical EngineeringEberhard Karls University TübingenSilcherstr. 7/1Tübingen72076Germany
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- 3R‐Center for In vitro Models and Alternatives to Animal TestingEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| |
Collapse
|
37
|
Folick A, Cheang RT, Valdearcos M, Koliwad SK. Metabolic factors in the regulation of hypothalamic innate immune responses in obesity. Exp Mol Med 2022; 54:393-402. [PMID: 35474339 PMCID: PMC9076660 DOI: 10.1038/s12276-021-00666-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/13/2021] [Indexed: 12/14/2022] Open
Abstract
The hypothalamus is a central regulator of body weight and energy homeostasis. There is increasing evidence that innate immune activation in the mediobasal hypothalamus (MBH) is a key element in the pathogenesis of diet-induced obesity. Microglia, the resident immune cells in the brain parenchyma, have been shown to play roles in diverse aspects of brain function, including circuit refinement and synaptic pruning. As such, microglia have also been implicated in the development and progression of neurological diseases. Microglia express receptors for and are responsive to a wide variety of nutritional, hormonal, and immunological signals that modulate their distinct functions across different brain regions. We showed that microglia within the MBH sense and respond to a high-fat diet and regulate the function of hypothalamic neurons to promote food intake and obesity. Neurons, glia, and immune cells within the MBH are positioned to sense and respond to circulating signals that regulate their capacity to coordinate aspects of systemic energy metabolism. Here, we review the current knowledge of how these peripheral signals modulate the innate immune response in the MBH and enable microglia to regulate metabolic control.
Collapse
Affiliation(s)
- Andrew Folick
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA
| | - Rachel T Cheang
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA
| | - Martin Valdearcos
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA.
| | - Suneil K Koliwad
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
38
|
Salas-Venegas V, Flores-Torres RP, Rodríguez-Cortés YM, Rodríguez-Retana D, Ramírez-Carreto RJ, Concepción-Carrillo LE, Pérez-Flores LJ, Alarcón-Aguilar A, López-Díazguerrero NE, Gómez-González B, Chavarría A, Konigsberg M. The Obese Brain: Mechanisms of Systemic and Local Inflammation, and Interventions to Reverse the Cognitive Deficit. Front Integr Neurosci 2022; 16:798995. [PMID: 35422689 PMCID: PMC9002268 DOI: 10.3389/fnint.2022.798995] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Overweight and obesity are now considered a worldwide pandemic and a growing public health problem with severe economic and social consequences. Adipose tissue is an organ with neuroimmune-endocrine functions, which participates in homeostasis. So, adipocyte hypertrophy and hyperplasia induce a state of chronic inflammation that causes changes in the brain and induce neuroinflammation. Studies with obese animal models and obese patients have shown a relationship between diet and cognitive decline, especially working memory and learning deficiencies. Here we analyze how obesity-related peripheral inflammation can affect central nervous system physiology, generating neuroinflammation. Given that the blood-brain barrier is an interface between the periphery and the central nervous system, its altered physiology in obesity may mediate the consequences on various cognitive processes. Finally, several interventions, and the use of natural compounds and exercise to prevent the adverse effects of obesity in the brain are also discussed.
Collapse
Affiliation(s)
- Verónica Salas-Venegas
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana - Unidad Iztapalapa, Mexico City, Mexico
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Rosa Pamela Flores-Torres
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana - Unidad Iztapalapa, Mexico City, Mexico
- Departamento de Biología de la Reproducción, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México (CDMX), Mexico City, Mexico
| | - Yesica María Rodríguez-Cortés
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Diego Rodríguez-Retana
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Ricardo Jair Ramírez-Carreto
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Luis Edgar Concepción-Carrillo
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Laura Josefina Pérez-Flores
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Adriana Alarcón-Aguilar
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Norma Edith López-Díazguerrero
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Beatriz Gómez-González
- Departamento de Biología de la Reproducción, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México (CDMX), Mexico City, Mexico
| | - Anahí Chavarría
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Mina Konigsberg
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
- *Correspondence: Mina Konigsberg,
| |
Collapse
|
39
|
Kreiner FF, Kraaijenhof JM, von Herrath M, Hovingh GKK, von Scholten BJ. Interleukin 6 in diabetes, chronic kidney disease and cardiovascular disease: mechanisms and therapeutic perspectives. Expert Rev Clin Immunol 2022; 18:377-389. [PMID: 35212585 DOI: 10.1080/1744666x.2022.2045952] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Diabetes, chronic kidney disease (CKD) and cardiovascular disease (CVD) are cardiometabolic diseases that remain amongst the leading causes of morbidity and premature mortality. Here, we review the current understanding of how anti-inflammatory intervention via inhibition of the pro-inflammatory but pleiotropic cytokine interleukin (IL) 6 may benefit patients with these or related diseases or complications. AREAS COVERED Based on a PubMed literature search, this review integrates and contextualizes evidence regarding the clinical utility of anti-IL-6 intervention in the treatment of cardiometabolic diseases, as well as of the associated condition non-alcoholic hepatosteatosis. EXPERT OPINION Evidence implicates the pro-inflammatory effects of IL-6 in the pathophysiology of diabetes, CKD and CVD. Thus, targeting the IL-6 pathway holds a therapeutic potential in these cardiometabolic disorders. However, because IL-6 has multiple homeostatic roles, antagonizing this cytokine may be associated with side effects such as increased risk of infection as seen with other anti-inflammatory drugs. Additional studies are required to establish the benefit-risk profile of anti-IL-6 intervention in the cardiometabolic diseases, whilst also considering alternative interventions such as lifestyle changes. IL-6 is also elevated in NASH, but the clinical usefulness of targeting IL-6 in this hepatic disorder remains largely unexplored.
Collapse
Affiliation(s)
| | - Jordan M Kraaijenhof
- Global Chief Medical Office, Novo Nordisk A/S, Søborg, Denmark.,Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Matthias von Herrath
- Global Chief Medical Office, Novo Nordisk A/S, Søborg, Denmark.,La Jolla Institute for Immunology, La Jolla, California, United States
| | - G Kees Kornelis Hovingh
- Global Chief Medical Office, Novo Nordisk A/S, Søborg, Denmark.,Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
40
|
Kistner TM, Pedersen BK, Lieberman DE. Interleukin 6 as an energy allocator in muscle tissue. Nat Metab 2022; 4:170-179. [PMID: 35210610 DOI: 10.1038/s42255-022-00538-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/21/2022] [Indexed: 12/31/2022]
Abstract
Extensive research has shown that interleukin 6 (IL-6) is a multifunctional molecule that is both proinflammatory and anti-inflammatory, depending on the context. Here, we combine an evolutionary perspective with physiological data to propose that IL-6's context-dependent effects on metabolism reflect its adaptive role for short-term energy allocation. This energy-allocation role is especially salient during physical activity, when skeletal muscle releases large amounts of IL-6. We predict that during bouts of physical activity, myokine IL-6 fulfills the three main characteristics of a short-term energy allocator: it is secreted from muscle in response to an energy deficit, it liberates somatic energy through lipolysis and it enhances muscular energy uptake and transiently downregulates immune function. We then extend this model of energy allocation beyond myokine IL-6 to reinterpret the roles that IL-6 plays in chronic inflammation, as well as during COVID-19-associated hyperinflammation and multiorgan failure.
Collapse
Affiliation(s)
- Timothy M Kistner
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
| | - Bente K Pedersen
- Centre of Inflammation and Metabolism/Centre for Physical Activity Research (CIM/CFAS), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | - Daniel E Lieberman
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
41
|
Poledne R, Kralova Lesna I. Adipose tissue macrophages and atherogenesis – a synergy with cholesterolaemia. Physiol Res 2021. [DOI: 10.33549//physiolres.934745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Excessive LDL cholesterol concentration together with subclinical inflammation, in which macrophages play a central role, are linked pathologies. The process starts with the accumulation of macrophages in white adipose tissue and the switch of their polarization toward a pro-inflammatory phenotype. The proportion of pro-inflammatory macrophages in adipose tissue is related to the main risk predictors of cardiovascular disease. The cholesterol content of phospholipids of cell membranes seems to possess a crucial role in the regulation of membrane signal transduction and macrophage polarization. Also, different fatty acids of membrane phospholipids influence phenotypes of adipose tissue macrophages with saturated fatty acids stimulating pro-inflammatory whereas ω3 fatty acids anti-inflammatory changes. The inflammatory status of white adipose tissue, therefore, reflects not only adipose tissue volume but also adipose tissue macrophages feature. The beneficial dietary change leading to an atherogenic lipoprotein decrease may therefore synergically reduce adipose tissue driven inflammation.
Collapse
Affiliation(s)
- R Poledne
- Laboratory for Atherosclerosis Research, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | |
Collapse
|
42
|
Maccarone MC, Magro G, Tognolo L, Masiero S. Post COVID-19 persistent fatigue: a proposal for rehabilitative interventions in the spa setting. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2021; 65:2241-2243. [PMID: 34086142 PMCID: PMC8175920 DOI: 10.1007/s00484-021-02158-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/19/2021] [Accepted: 05/31/2021] [Indexed: 05/09/2023]
Abstract
With this letter to Editor, we aim at offering our viewpoint on the potential use of the spa resources to contribute to the treatment of persistent COVID-19 sequelae. In particular, our argumentations focus on the comprehensive management of fatigue persisting after the resolution of the acute infection. This sequela seems to be the most frequent disabling outcome, leading to a delay in social reintegration and return to working life.We suggest that spa facilities including multidisciplinary interventions and trained staffs could be an appropriate setting for providing rehabilitative protocols to treat chronic fatigue in post COVID-19 subjects. Spa treatments may improve the physical symptoms of chronic fatigue and modulate the immune imbalance, reducing persisting inflammation. Moreover, in the spa setting, specific respiratory rehabilitation and neuropsychological interventions could be provided. The rehabilitative protocol could also properly address comorbidities.In conclusion, respecting the hygiene and preventive measures, the spa setting could represent the appropriate environment to take care of post-COVID chronic fatigue, offering adequate and multidisciplinary rehabilitation protocols. Further, prospective trials on this topic are needed to fully evaluate the positive impact of a comprehensive rehabilitative treatment in the spa environment for subjects with post-COVID-19 fatigue.
Collapse
Affiliation(s)
- Maria Chiara Maccarone
- Physical Medicine and Rehabilitation School, University of Padova, Via Giustiniani 3, 35128, Padua, Italy.
| | - Giacomo Magro
- Physical Medicine and Rehabilitation School, University of Padova, Padua, Italy
| | - Lucrezia Tognolo
- Rehabilitation Unit, Department of Neuroscience, University of Padova, Padua, Italy
| | - Stefano Masiero
- Rehabilitation Unit, Department of Neuroscience, University of Padova, Padua, Italy
| |
Collapse
|
43
|
Santos GSP, Costa AC, Picoli CC, Rocha BGS, Sulaiman SO, Radicchi DC, Pinto MCX, Batista ML, Amorim JH, Azevedo VAC, Resende RR, Câmara NOS, Mintz A, Birbrair A. Sympathetic nerve-adipocyte interactions in response to acute stress. J Mol Med (Berl) 2021; 100:151-165. [PMID: 34735579 PMCID: PMC8567732 DOI: 10.1007/s00109-021-02157-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022]
Abstract
Psychological stress predisposes our body to several disorders. Understanding the cellular and molecular mechanisms involved in the physiological responses to psychological stress is essential for the success of therapeutic applications. New studies show, by using in vivo inducible Cre/loxP-mediated approaches in combination with pharmacological blockage, that sympathetic nerves, activated by psychological stress, induce brown adipocytes to produce IL-6. Strikingly, this cytokine promotes gluconeogenesis in hepatocytes, that results in the decline of tolerance to inflammatory organ damage. The comprehension arising from this research will be crucial for the handling of many inflammatory diseases. Here, we review recent advances in our comprehension of the sympathetic nerve-adipocyte axis in the tissue microenvironment.
Collapse
Affiliation(s)
- Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alinne C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Beatriz G S Rocha
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sheu O Sulaiman
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Debora C Radicchi
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro C X Pinto
- Laboratory of Neuropharmacology, Federal University of Goiás, Goiânia, GO, Brazil
| | - Miguel L Batista
- Laboratory of Adipose Tissue Biology, University of Mogi das Cruzes, Mogi das Cruzes, SP, Brazil.,Department of Biochemistry, Boston University School of Medicine, Boston, USA
| | - Jaime H Amorim
- Center of Biological Sciences and Health, Federal University of Western Bahia, BA, Barreiras, Brazil
| | - Vasco A C Azevedo
- Cellular and Molecular Genetics Laboratory, Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Niels O S Câmara
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, SP, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
44
|
Feraco A, Gorini S, Armani A, Camajani E, Rizzo M, Caprio M. Exploring the Role of Skeletal Muscle in Insulin Resistance: Lessons from Cultured Cells to Animal Models. Int J Mol Sci 2021; 22:ijms22179327. [PMID: 34502235 PMCID: PMC8430804 DOI: 10.3390/ijms22179327] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle is essential to maintain vital functions such as movement, breathing, and thermogenesis, and it is now recognized as an endocrine organ. Muscles release factors named myokines, which can regulate several physiological processes. Moreover, skeletal muscle is particularly important in maintaining body homeostasis, since it is responsible for more than 75% of all insulin-mediated glucose disposal. Alterations of skeletal muscle differentiation and function, with subsequent dysfunctional expression and secretion of myokines, play a key role in the pathogenesis of obesity, type 2 diabetes, and other metabolic diseases, finally leading to cardiometabolic complications. Hence, a deeper understanding of the molecular mechanisms regulating skeletal muscle function related to energy metabolism is critical for novel strategies to treat and prevent insulin resistance and its cardiometabolic complications. This review will be focused on both cellular and animal models currently available for exploring skeletal muscle metabolism and endocrine function.
Collapse
Affiliation(s)
- Alessandra Feraco
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Stefania Gorini
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Elisabetta Camajani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
- PhD Programme in Endocrinological Sciences, Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Manfredi Rizzo
- Promise Department, School of Medicine, University of Palermo, 90127 Palermo, Italy;
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
- Correspondence: ; Tel.: +39-065-225-3419
| |
Collapse
|
45
|
Sonsalla MM, Love SL, Hoh LJ, Summers LN, Follett HM, Bojang A, Duddleston KN, Kurtz CC. Development of metabolic inflammation during pre-hibernation fattening in 13-lined ground squirrels (Ictidomys tridecemlineatus). J Comp Physiol B 2021; 191:941-953. [PMID: 34165591 DOI: 10.1007/s00360-021-01384-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/07/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023]
Abstract
Obesity is a worldwide pandemic with significant comorbidities. It is often accompanied by mild inflammation of the intestine followed by inflammation of metabolic tissues such as liver, adipose tissue, and skeletal muscle. Several laboratory models of obesity exist, but seasonal models like hibernators may be valuable for understanding the pathogenesis of obesity independent of genetic or high-fat diet-induced changes. As part of their annual cycle, obligate hibernators, like the 13-lined ground squirrel (Ictidomys tridecemlineatus), undergo a rapid shift from a lean to an obese state to store energy in the form of fat for their prolonged winter fast. Here, we show that ground squirrels gained mass steadily throughout the active season despite a drop in energy intake starting around 9 weeks post-hibernation. Glucose tolerance tests revealed a significant decrease in tolerance late in the active season. In visceral adipose, we found increases in adipocyte size, tumor necrosis factor (TNF)-α and interleukin (IL)-6 levels. IL-6 levels also increased in liver and muscle and TNF-α increased in the ileum late in the active season. Levels of the anti-inflammatory cytokine, IL-10, decreased in visceral adipose and colon tissues around the same time. These data suggest metabolic inflammation develops along with adiposity late in the squirrels' active season.
Collapse
Affiliation(s)
- Michelle M Sonsalla
- Department of Biology, College of Letters and Science, University of Wisconsin-Oshkosh, 800 Algoma Boulevard, Oshkosh, WI, 54901, USA
| | - Santidra L Love
- Department of Biology, College of Letters and Science, University of Wisconsin-Oshkosh, 800 Algoma Boulevard, Oshkosh, WI, 54901, USA
| | - Laurana J Hoh
- Department of Biology, College of Letters and Science, University of Wisconsin-Oshkosh, 800 Algoma Boulevard, Oshkosh, WI, 54901, USA
| | - Lauren N Summers
- Department of Biology, College of Letters and Science, University of Wisconsin-Oshkosh, 800 Algoma Boulevard, Oshkosh, WI, 54901, USA
| | - Hannah M Follett
- Department of Biology, College of Letters and Science, University of Wisconsin-Oshkosh, 800 Algoma Boulevard, Oshkosh, WI, 54901, USA
| | - Aminata Bojang
- Department of Biology, College of Letters and Science, University of Wisconsin-Oshkosh, 800 Algoma Boulevard, Oshkosh, WI, 54901, USA
| | - Khrystyne N Duddleston
- Department of Biological Sciences, College of Arts and Sciences, University of Alaska Anchorage, 3211 Providence Dr., Anchorage, AK, 99508, USA
| | - Courtney C Kurtz
- Department of Biology, College of Letters and Science, University of Wisconsin-Oshkosh, 800 Algoma Boulevard, Oshkosh, WI, 54901, USA.
| |
Collapse
|
46
|
Michailidou Z, Gomez-Salazar M, Alexaki VI. Innate Immune Cells in the Adipose Tissue in Health and Metabolic Disease. J Innate Immun 2021; 14:4-30. [PMID: 33849008 DOI: 10.1159/000515117] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/09/2021] [Indexed: 11/19/2022] Open
Abstract
Metabolic disorders, such as obesity, type 2 diabetes mellitus, and nonalcoholic fatty liver disease, are characterized by chronic low-grade tissue and systemic inflammation. During obesity, the adipose tissue undergoes immunometabolic and functional transformation. Adipose tissue inflammation is driven by innate and adaptive immune cells and instigates insulin resistance. Here, we discuss the role of innate immune cells, that is, macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid type 2 cells, dendritic cells, and mast cells, in the adipose tissue in the healthy (lean) and diseased (obese) state and describe how their function is shaped by the obesogenic microenvironment, and humoral, paracrine, and cellular interactions. Moreover, we particularly outline the role of hypoxia as a central regulator in adipose tissue inflammation. Finally, we discuss the long-lasting effects of adipose tissue inflammation and its potential reversibility through drugs, caloric restriction, or exercise training.
Collapse
Affiliation(s)
- Zoi Michailidou
- Centre for Cardiovascular Sciences, Edinburgh University, Edinburgh, United Kingdom
| | - Mario Gomez-Salazar
- Centre for Cardiovascular Sciences, Edinburgh University, Edinburgh, United Kingdom
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
47
|
IL-6 Receptor Blockade Increases Circulating Adiponectin Levels in People with Obesity: An Explanatory Analysis. Metabolites 2021; 11:metabo11020079. [PMID: 33572989 PMCID: PMC7911215 DOI: 10.3390/metabo11020079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/18/2021] [Accepted: 01/25/2021] [Indexed: 11/17/2022] Open
Abstract
Human obesity is associated with decreased circulating adiponectin and elevated leptin levels. In vitro experiments and studies in high fat diet (HFD)-fed mice suggest that interleukin-6 (IL-6) may regulate adiponectin and leptin release from white adipose tissue (WAT). Herein, we aimed to investigate whether IL-6 receptor blockade affects the levels of circulating adiponectin and leptin in obese human individuals. To this end, serum samples collected during a multicenter, double-blind clinical trial were analyzed. In the latter study, obese human subjects with or without type 2 diabetes were randomly assigned to recurrent placebo or intravenous tocilizumab (an IL-6 receptor antibody) administration during a 12-week exercise training intervention. Twelve weeks of tocilizumab administration (in combination with exercise training) trend wise enhanced the decrease in circulating leptin levels (−2.7 ± 8.2% in the placebo vs. −20.6 ± 5.6% in tocilizumab, p = 0.08) and significantly enhanced the increase in circulating adiponectin (3.4 ± 3.7% in the placebo vs. 27.0 ± 6.6% in tocilizumab, p = 0.01). In addition, circulating adiponectin levels were negatively correlated with the homeostatic model assessment of insulin resistance (HOMA-IR), indicating that increased adiponectin levels positively affect insulin sensitivity in people with obesity. In conclusion, IL-6 receptor blockade increases circulating adiponectin levels in people with obesity.
Collapse
|