1
|
Arner P, Sørensen TI, Andersson DP. Adipose cellularity as a measurement of long-term changes in body weight: a Swedish cohort study spanning 1988-2016. EClinicalMedicine 2025; 82:103165. [PMID: 40235948 PMCID: PMC11997358 DOI: 10.1016/j.eclinm.2025.103165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 04/17/2025] Open
Abstract
Background Adipocyte size and number (cellularity) determine the adipose mass and may relate to long-term body weight changes. Methods We investigated 1014 healthy participants at Karolinska Institutet in Sweden 1988-2016 for body weight and size/number of subcutaneous adipocytes, and 273 for visceral adipocyte size. We measured body weight on 281 subjects about 16 years later. We analysed the association of baseline adipocyte size and number with body weight changes by linear regression including relevant co-factors, and the associations of cellularity (low or high number of either large or small adipocytes) regarding body weight changes by analysis of variance. Findings Subcutaneous adipocyte size and number and visceral adipocyte size showed strong relationships with body weight changes irrespective of its mode of expression (adjusted r2 ≥0·15). The relationships were significant (p ≤ 0·027) independent of co-factors (age, sex, initial body weight or height, body fat, obesity, nicotine use, physical activity, and observation time). Interventions (lifestyle change or bariatric surgery) did not influence the associations (p = 0·86). A low or high number of large adipocytes associated with body weight loss, whereas a low or high number of small cells associated with weight stability or weight gain. Interpretation Adipose cellularity is associated with long-term changes in body weight, following interventions to decrease body weight. Patients with a high number of large fat cells experienced the most pronounced weight reduction. Funding The Stockholm County Council (963296, 994175, 986118), the Center for Innovative Medicine at Karolinska Institutet (986109) and the Swedish Society of Medicine (1001156). None of the funding sources had any involvement in the study.
Collapse
Affiliation(s)
- Peter Arner
- Department of Medicine-H7 at Karolinska Institutet, C2:94 Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| | - Thorkild I.A. Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Public Health Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen, 2200N, Denmark
- Center for Childhood Health, Islands Brygge 41, Copenhagen, 2300S, Denmark
| | - Daniel P. Andersson
- Department of Medicine-H7 at Karolinska Institutet, C2:94 Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
- Department of Endocrinology, C2:94 Karolinska University Hospital Huddinge, 14186, Stockholm, Sweden
| |
Collapse
|
2
|
Wang Z, Liu X, Sheng L, Xie Y, Feng W, Yu L. Effects of duration of high-fat diet on adipocyte hyperplasia in rat epididymis. Obes Res Clin Pract 2025; 19:54-62. [PMID: 39922761 DOI: 10.1016/j.orcp.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 06/25/2024] [Accepted: 02/01/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND High-fat diet (HFD) contributes to obesity and enhances the expression of mature adipocyte markers. However, the effect of HFD on adipocyte hyperplasia remains controversial. This may be due to variations in the duration of HFD. This study aimed to investigate the effects of different durations of HFD on adipocyte hyperplasia and the expression of mature adipocyte-related markers in obese rats. METHODS We divided 32 Sprague-Dawley rats into four groups: B (standard diet control), H1 (HFD for four weeks), H2 (HFD for eight weeks), and H3 (HFD for 12 weeks). We evaluated the morphological changes in epididymal fat cells, measured serum inflammatory markers using enzyme-linked immunosorbent assay (ELISA) kits, and quantified adipocyte hyperplasia and maturation markers using western blotting. RESULTS We observed progressive increases in body weight, epididymal fat weight, serum leptin, TNF-α, IL-6, irisin, PPARγ, adiponectin, and FNDC5 protein expression over 8 weeks of HFD. 12 weeks of HFD intervention resulted in significant decreases in irisin, PPARγ, adiponectin, and FNDC5. Concurrently, the expression of perilipin A and ATGL declined with prolonged HFD. CONCLUSIONS Our results suggest that the duration of HFD significantly affects adipocyte ability to undergo hyperplasia in the epididymis of obese rats. Specifically, 4 weeks of HFD did not change the capacity for adipocyte hyperplasia, while 8 weeks of the diet enhanced this capacity. Interestingly, a longer diet duration (12 weeks) led to a decrease in adipocyte hyperplasia.
Collapse
Affiliation(s)
- Zhaoxin Wang
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Xiujuan Liu
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China.
| | - Lei Sheng
- Department of Scientific Research, Nanjing Sport Institute, Nanjing, China.
| | - Yuting Xie
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Wanyu Feng
- Department of Science Experiment Center, Nanjing Sport Institute, Nanjing, China
| | - Li Yu
- Department of Sports and Health, Nanjing Sport Institute, Nanjing, China
| |
Collapse
|
3
|
Popa AD, Gherasim A, Caba L, Niță O, Graur M, Mihalache L, Arhire LI. Cathelicidin: Insights into Its Impact on Metabolic Syndrome and Chronic Inflammation. Metabolites 2024; 14:672. [PMID: 39728453 DOI: 10.3390/metabo14120672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/23/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: LL-37 is associated with metabolic syndrome (MetS), a constellation of risk factors comprising obesity, insulin resistance (IR), dyslipidemia, and hypertension, which elevates the risk of cardiovascular disease and type 2 diabetes. Methods: In this narrative review, we analyzed the literature focusing on recent developments in the relationship between cathelicidin and various components of MetS to provide a comprehensive overview. Results: Studies have shown that LL-37 is linked to inflammation in adipose tissue (AT) and the development of IR in obesity. Cathelicidin can enhance inflammation by activating pro-inflammatory genes, as well as modulate the inflammatory response. The mechanisms of IR include the activation of complex signaling pathways that induce inflammation and reduce insulin signaling in adipocytes. The activation of Toll-like receptors (TLRs) by cathelicidin stimulates the secretion of pro-inflammatory cytokines, contributing to the disruption of insulin function in adipose cells. Cathelicidin also influences lipid metabolism, with recent research showing a negative relationship between LL-37 levels and HDL cholesterol. Therefore, LL-37 is involved not only in the regulation of inflammation but also in lipid metabolism, potentially aggravating the cardiovascular complications associated with MetS. Conclusions: Cathelicidin plays a crucial role in regulating the balance between inflammatory and anti-inflammatory responses in MetS. Understanding the impact of LL-37 on these mechanisms may unveil novel approaches for addressing MetS and its associated complications.
Collapse
Affiliation(s)
- Alina Delia Popa
- Internal Medicine II Department, Faculty of Medicine, University of Medicine, and Pharmacy "Grigore T. Popa", 700115 Iasi, Romania
| | - Andreea Gherasim
- Internal Medicine II Department, Faculty of Medicine, University of Medicine, and Pharmacy "Grigore T. Popa", 700115 Iasi, Romania
| | - Lavinia Caba
- Department of Medical Genetics, Faculty of Medicine, University of Medicine and Pharmacy "Grigore T. Popa", 16 University Street, 700115 Iasi, Romania
| | - Otilia Niță
- Internal Medicine II Department, Faculty of Medicine, University of Medicine, and Pharmacy "Grigore T. Popa", 700115 Iasi, Romania
| | - Mariana Graur
- Faculty of Medicine and Biological Sciences, University "Ștefan cel Mare" of Suceava, 720229 Suceava, Romania
| | - Laura Mihalache
- Internal Medicine II Department, Faculty of Medicine, University of Medicine, and Pharmacy "Grigore T. Popa", 700115 Iasi, Romania
| | - Lidia Iuliana Arhire
- Internal Medicine II Department, Faculty of Medicine, University of Medicine, and Pharmacy "Grigore T. Popa", 700115 Iasi, Romania
| |
Collapse
|
4
|
Gao YP, Hu C, Hu M, Dong WS, Li K, Ye YJ, Hu YX, Zhang X. CREB3 protein family: the promising therapeutic targets for cardiovascular and metabolic diseases. Cell Biol Toxicol 2024; 40:103. [PMID: 39581923 PMCID: PMC11586310 DOI: 10.1007/s10565-024-09939-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/17/2024] [Indexed: 11/26/2024]
Abstract
Significant advancements in cardiovascular and metabolic disease research have been made with the CREB3 protein family. Studies have revealed that members of this family are crucial in the development of these diseases, contributing to the regulation of lipid metabolism, inflammation, and vascular function. These studies provide useful information for future therapeutic strategies in cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Yi-Peng Gao
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Can Hu
- Department of Ultrasound, Clinical Research Center for Medical Imaging in Hubei Province, Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Min Hu
- Department of Cardiology, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wen-Sheng Dong
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Kang Li
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yun-Jia Ye
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu-Xin Hu
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xin Zhang
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
5
|
Pinette JA, Myers JW, Park WY, Bryant HG, Eddie AM, Wilson GA, Montufar C, Shaikh Z, Vue Z, Nunn ER, Bessho R, Cottam MA, Haase VH, Hinton AO, Spinelli JB, Cartailler JP, Zaganjor E. Disruption of nucleotide biosynthesis reprograms mitochondrial metabolism to inhibit adipogenesis. J Lipid Res 2024; 65:100641. [PMID: 39245323 PMCID: PMC11913791 DOI: 10.1016/j.jlr.2024.100641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/13/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024] Open
Abstract
A key organismal response to overnutrition involves the development of new adipocytes through the process of adipogenesis. Preadipocytes sense changes in the systemic nutrient status and metabolites can directly modulate adipogenesis. We previously identified a role of de novo nucleotide biosynthesis in adipogenesis induction, whereby inhibition of nucleotide biosynthesis suppresses the expression of the transcriptional regulators PPARγ and C/EBPα. Here, we set out to identify the global transcriptomic changes associated with the inhibition of nucleotide biosynthesis. Through RNA sequencing (RNAseq), we discovered that mitochondrial signatures were the most altered in response to inhibition of nucleotide biosynthesis. Blocking nucleotide biosynthesis induced rounded mitochondrial morphology, and altered mitochondrial function, and metabolism, reducing levels of tricarboxylic acid cycle intermediates, and increasing fatty acid oxidation (FAO). The loss of mitochondrial function induced by suppression of nucleotide biosynthesis was rescued by exogenous expression of PPARγ. Moreover, inhibition of FAO restored PPARγ expression, mitochondrial protein expression, and adipogenesis in the presence of nucleotide biosynthesis inhibition, suggesting a regulatory role of nutrient oxidation in differentiation. Collectively, our studies shed light on the link between substrate oxidation and transcription in cell fate determination.
Collapse
Affiliation(s)
- Julia A Pinette
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jacob W Myers
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Woo Yong Park
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Heather G Bryant
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Alex M Eddie
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Genesis A Wilson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Claudia Montufar
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Zayedali Shaikh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Elizabeth R Nunn
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Ryoichi Bessho
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew A Cottam
- Creative Data Solutions, Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | - Volker H Haase
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Research and Medical Services, Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Antentor O Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jessica B Spinelli
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jean-Philippe Cartailler
- Creative Data Solutions, Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Vanderbilt Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Diabetes Research Center, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
6
|
Del Bianco V, Ferreira GDS, Bochi APG, Pinto PR, Rodrigues LG, Furukawa LNS, Okamoto MM, Almeida JA, da Silveira LKR, Santos AS, Bispo KCS, Capelozzi VL, Correa-Giannella ML, da Silva AA, Velosa APP, Nakandakare ER, Machado UF, Teodoro WPR, Passarelli M, Catanozi S. Aerobic Exercise Training Protects Against Insulin Resistance, Despite Low-Sodium Diet-Induced Increased Inflammation and Visceral Adiposity. Int J Mol Sci 2024; 25:10179. [PMID: 39337664 PMCID: PMC11432465 DOI: 10.3390/ijms251810179] [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: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Dietary sodium restriction increases plasma triglycerides (TG) and total cholesterol (TC) concentrations as well as causing insulin resistance and stimulation of the renin-angiotensin-aldosterone system (RAAS) and the sympathetic nervous system. Stimulation of the angiotensin II type-1 receptor (AT1) is associated with insulin resistance, inflammation, and the inhibition of adipogenesis. The current study investigated whether aerobic exercise training (AET) mitigates or inhibits the adverse effects of dietary sodium restriction on adiposity, inflammation, and insulin sensitivity in periepididymal adipose tissue. LDL receptor knockout mice were fed either a normal-sodium (NS; 1.27% NaCl) or a low-sodium (LS; 0.15% NaCl) diet and were either subjected to AET for 90 days or kept sedentary. Body mass, blood pressure (BP), hematocrit, plasma TC, TG, glucose and 24-hour urinary sodium (UNa) concentrations, insulin sensitivity, lipoprotein profile, histopathological analyses, and gene and protein expression were determined. The results were evaluated using two-way ANOVA. Differences were not observed in BP, hematocrit, diet consumption, and TC. The LS diet was found to enhance body mass, insulin resistance, plasma glucose, TG, LDL-C, and VLDL-TG and reduce UNa, HDL-C, and HDL-TG, showing a pro-atherogenic lipid profile. In periepididymal adipose tissue, the LS diet increased tissue mass, TG, TC, AT1 receptor, pro-inflammatory macro-phages contents, and the area of adipocytes; contrarily, the LS diet decreased anti-inflammatory macrophages, protein contents and the transcription of genes related to insulin sensitivity. The AET prevented insulin resistance, but did not protect against dyslipidemia, adipose tissue pro-inflammatory profile, increased tissue mass, AT1 receptor expression, TG, and TC induced by the LS diet.
Collapse
Affiliation(s)
- Vanessa Del Bianco
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Guilherme da Silva Ferreira
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Ana Paula Garcia Bochi
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Paula Ramos Pinto
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Letícia Gomes Rodrigues
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Luzia Naoko Shinohara Furukawa
- Laboratory of Renal Pathophysiology, Department of Internal Medicine, School of Medicine, University of São Paulo, São Paulo 01246 000, Brazil;
| | - Maristela Mitiko Okamoto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508 000, Brazil; (M.M.O.); (U.F.M.)
| | - Jaíne Alves Almeida
- Rheumatology Division of the Hospital das Clinicas, University of São Paulo Medical School, São Paulo 01246 000, Brazil; (J.A.A.); (L.K.R.d.S.); (A.P.P.V.); (W.P.R.T.)
| | - Lizandre Keren Ramos da Silveira
- Rheumatology Division of the Hospital das Clinicas, University of São Paulo Medical School, São Paulo 01246 000, Brazil; (J.A.A.); (L.K.R.d.S.); (A.P.P.V.); (W.P.R.T.)
| | - Aritania Sousa Santos
- Laboratorio de Carboidratos e Radioimunoensaios (Laboratorio de Investigações Médicas, LIM-18), Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo 01246 000, Brazil; (A.S.S.); (M.L.C.-G.)
| | - Kely Cristina Soares Bispo
- Department of Pathology of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, Sao Paulo 01246 000, Brazil; (K.C.S.B.); (V.L.C.)
| | - Vera Luiza Capelozzi
- Department of Pathology of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, Sao Paulo 01246 000, Brazil; (K.C.S.B.); (V.L.C.)
| | - Maria Lucia Correa-Giannella
- Laboratorio de Carboidratos e Radioimunoensaios (Laboratorio de Investigações Médicas, LIM-18), Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo 01246 000, Brazil; (A.S.S.); (M.L.C.-G.)
| | - Alexandre Alves da Silva
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Ana Paula Pereira Velosa
- Rheumatology Division of the Hospital das Clinicas, University of São Paulo Medical School, São Paulo 01246 000, Brazil; (J.A.A.); (L.K.R.d.S.); (A.P.P.V.); (W.P.R.T.)
| | - Edna Regina Nakandakare
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| | - Ubiratan Fabres Machado
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508 000, Brazil; (M.M.O.); (U.F.M.)
| | - Walcy Paganelli Rosolia Teodoro
- Rheumatology Division of the Hospital das Clinicas, University of São Paulo Medical School, São Paulo 01246 000, Brazil; (J.A.A.); (L.K.R.d.S.); (A.P.P.V.); (W.P.R.T.)
| | - Marisa Passarelli
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
- Programa de Pós Graduação em Medicina, Universidade Nove de Julho, Sao Paulo 01525 000, Brazil
| | - Sergio Catanozi
- Laboratorio de Lipides (LIM-10), Hospital das Clinicas (HCFMUSP) da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo 01246 000, Brazil; (V.D.B.); (G.d.S.F.); (A.P.G.B.); (P.R.P.); (L.G.R.); (E.R.N.); (M.P.)
| |
Collapse
|
7
|
Hofwimmer K, de Paula Souza J, Subramanian N, Vujičić M, Rachid L, Méreau H, Zhao C, Dror E, Barreby E, Björkström NK, Wernstedt Asterholm I, Böni-Schnetzler M, Meier DT, Donath MY, Laurencikiene J. IL-1β promotes adipogenesis by directly targeting adipocyte precursors. Nat Commun 2024; 15:7957. [PMID: 39261467 PMCID: PMC11390900 DOI: 10.1038/s41467-024-51938-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 08/21/2024] [Indexed: 09/13/2024] Open
Abstract
Postprandial IL-1β surges are predominant in the white adipose tissue (WAT), but its consequences are unknown. Here, we investigate the role of IL-1β in WAT energy storage and show that adipocyte-specific deletion of IL-1 receptor 1 (IL1R1) has no metabolic consequences, whereas ubiquitous lack of IL1R1 reduces body weight, WAT mass, and adipocyte formation in mice. Among all major WAT-resident cell types, progenitors express the highest IL1R1 levels. In vitro, IL-1β potently promotes adipogenesis in murine and human adipose-derived stem cells. This effect is exclusive to early-differentiation-stage cells, in which the adipogenic transcription factors C/EBPδ and C/EBPβ are rapidly upregulated by IL-1β and enriched near important adipogenic genes. The pro-adipogenic, but not pro-inflammatory effect of IL-1β is potentiated by acute treatment and blocked by chronic exposure. Thus, we propose that transient postprandial IL-1β surges regulate WAT remodeling by promoting adipogenesis, whereas chronically elevated IL-1β levels in obesity blunts this physiological function.
Collapse
Affiliation(s)
- Kaisa Hofwimmer
- Lipid Laboratory, Unit of Endocrinology, Department of Medicine Huddinge, Karolinska Institutet, SE-141 52, Huddinge, Sweden
| | - Joyce de Paula Souza
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Narmadha Subramanian
- Lipid Laboratory, Unit of Endocrinology, Department of Medicine Huddinge, Karolinska Institutet, SE-141 52, Huddinge, Sweden
| | - Milica Vujičić
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Leila Rachid
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Hélène Méreau
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Cheng Zhao
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Erez Dror
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Emelie Barreby
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, SE-141 52, Huddinge, Sweden
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, SE-141 52, Huddinge, Sweden
| | - Ingrid Wernstedt Asterholm
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Marianne Böni-Schnetzler
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Daniel T Meier
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland.
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland.
| | - Marc Y Donath
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Jurga Laurencikiene
- Lipid Laboratory, Unit of Endocrinology, Department of Medicine Huddinge, Karolinska Institutet, SE-141 52, Huddinge, Sweden.
| |
Collapse
|
8
|
Song X, Song Y, Zhang J, Hu Y, Zhang L, Huang Z, Abbas Raza SH, Jiang C, Ma Y, Ma Y, Wu H, Wei D. Regulatory role of exosome-derived miRNAs and other contents in adipogenesis. Exp Cell Res 2024; 441:114168. [PMID: 39004201 DOI: 10.1016/j.yexcr.2024.114168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Intramuscular fat (IMF) content significantly impacts meat quality. influenced by complex interactions between skeletal muscle cells and adipocytes. Adipogenesis plays a pivotal role in IMF formation. Exosomes, extracellular membranous nanovesicles, facilitate intercellular communication by transporting proteins, nucleic acids (DNA and RNA), and other biomolecules into target cells, thereby modulating cellular behaviors. Recent studies have linked exosome-derived microRNAs (miRNAs) and other cargo to adipogenic processes. Various cell types, including skeletal muscle cells, interact with adipocytes via exosome secretion and uptake. Exosomes entering adipocytes regulate adipogenesis by modulating key signaling pathways, thereby influencing the extent and distribution of IMF deposition. This review comprehensively explores the origin, formation, and mechanisms of exosome action, along with current research and their applications in adipogenesis. Emphasis is placed on exosome-mediated regulation of miRNAs, non-coding RNAs (ncRNAs), proteins, lipids, and other biomolecules during adipogenesis. Leveraging exosomal contents for genetic breeding and treating obesity-related disorders is discussed. Insights gathered contribute to advancing understanding and potential therapeutic applications of exosome-regulated adipogenesis mechanisms.
Collapse
Affiliation(s)
- Xiaoyu Song
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Yaping Song
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Jiupan Zhang
- Institute of Animal Science, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan, 750021, China
| | - Yamei Hu
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Lingkai Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | | | - Sayed Haidar Abbas Raza
- Xichang University, Xichang, 615000, China; Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, 510642, China
| | - Chao Jiang
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Yanfen Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Yun Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Hao Wu
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Dawei Wei
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China.
| |
Collapse
|
9
|
McLaren J, Gao X, Ghouri N, Freeman DJ, Richardson J, Sattar N, Gill JMR. Weight gain leads to greater adverse metabolic responses in South Asian compared with white European men: the GlasVEGAS study. Nat Metab 2024; 6:1632-1645. [PMID: 39152223 PMCID: PMC11349579 DOI: 10.1038/s42255-024-01101-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 07/09/2024] [Indexed: 08/19/2024]
Abstract
South Asians (SAs) develop type 2 diabetes at lower body mass index values than white Europeans (WEs). This basic human experimental study aimed to compare the metabolic consequences of weight gain in SA and WE men without overweight or obesity. Fourteen SAs and 21 WEs had assessments of body composition, metabolic responses to mixed-meal ingestion, cardiorespiratory fitness and physical activity, and a subcutaneous abdominal adipose tissue biopsy, before and after 4-6 weeks of overfeeding to induce 5-7% weight gain. Here we show that body mass index and whole-body adipose tissue volume increases similarly between ethnic groups, but SAs gain less lean tissue. SAs experience a substantially greater decrease in insulin sensitivity compared with WEs (38% versus 7% decrease, P = 0.009), have fewer small (37.1% versus 60.0%, P = 0.003) and more large (26.2% versus 9.1%, P = 0.005) adipocytes at baseline and have a smaller decrease in very small adipocytes with weight gain (-0.1% versus -1.9%, P < 0.0001). Ethnic differences in adipocyte morphology are associated with SA's greater adverse metabolic changes with weight gain. ClinicalTrials.gov registration: NCT02399423 .
Collapse
Affiliation(s)
- James McLaren
- School of Cardiovascular and Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Xuan Gao
- School of Cardiovascular and Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Nazim Ghouri
- School of Cardiovascular and Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of General Medicine, Queen Elizabeth University Hospital, Glasgow, UK
| | - Dilys J Freeman
- School of Cardiovascular and Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Janice Richardson
- School of Cardiovascular and Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jason M R Gill
- School of Cardiovascular and Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
10
|
Wang G, Muñoz-Rojas AR, Spallanzani RG, Franklin RA, Benoist C, Mathis D. Adipose-tissue Treg cells restrain differentiation of stromal adipocyte precursors to promote insulin sensitivity and metabolic homeostasis. Immunity 2024; 57:1345-1359.e5. [PMID: 38692280 PMCID: PMC11188921 DOI: 10.1016/j.immuni.2024.04.002] [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: 05/23/2023] [Revised: 02/08/2024] [Accepted: 04/04/2024] [Indexed: 05/03/2024]
Abstract
Regulatory T (Treg) cells in epidydimal visceral adipose tissue (eVAT) of lean mice and humans regulate metabolic homeostasis. We found that constitutive or punctual depletion of eVAT-Treg cells reined in the differentiation of stromal adipocyte precursors. Co-culture of these precursors with conditional medium from eVAT-Treg cells limited their differentiation in vitro, suggesting a direct effect. Transcriptional comparison of adipocyte precursors, matured in the presence or absence of the eVAT-Treg-conditioned medium, identified the oncostatin-M (OSM) signaling pathway as a key distinction. Addition of OSM to in vitro cultures blocked the differentiation of adipocyte precursors, while co-addition of anti-OSM antibodies reversed the ability of the eVAT-Treg-conditioned medium to inhibit in vitro adipogenesis. Genetic depletion of OSM (specifically in Treg) cells or of the OSM receptor (specifically on stromal cells) strongly impaired insulin sensitivity and related metabolic indices. Thus, Treg-cell-mediated control of local progenitor cells maintains adipose tissue and metabolic homeostasis, a regulatory axis seemingly conserved in humans.
Collapse
Affiliation(s)
- Gang Wang
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | | | | | - Ruth A Franklin
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | | | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Puig N, Rives J, Gil-Millan P, Miñambres I, Ginel A, Tauron M, Bonaterra-Pastra A, Hernández-Guillamon M, Pérez A, Sánchez-Quesada JL, Benitez S. Apolipoprotein J protects cardiomyocytes from lipid-mediated inflammation and cytotoxicity induced by the epicardial adipose tissue of diabetic patients. Biomed Pharmacother 2024; 175:116779. [PMID: 38776681 DOI: 10.1016/j.biopha.2024.116779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Diabetic patients present increased volume and functional alterations in epicardial adipose tissue (EAT). We aimed to analyze EAT from type 2 diabetic patients and the inflammatory and cytotoxic effects induced on cardiomyocytes. Furthermore, we analyzed the cardioprotective role of apolipoprotein J (apoJ). EAT explants were obtained from nondiabetic patients (ND), diabetic patients without coronary disease (DM), and DM patients with coronary disease (DM-C) after heart surgery. Morphological characteristics and gene expression were evaluated. Explants were cultured for 24 h and the content of nonesterified fatty acids (NEFA) and sphingolipid species in secretomes was evaluated by lipidomic analysis. Afterwards, secretomes were added to AC16 human cardiomyocytes for 24 h in the presence or absence of cardioprotective molecules (apoJ and HDL). Cytokine release and apoptosis/necrosis were assessed by ELISA and flow cytometry. The EAT from the diabetic samples showed altered expression of genes related to lipid accumulation, insulin resistance, and inflammation. The secretomes from the DM samples presented an increased ratio of pro/antiatherogenic ceramide (Cer) species, while those from DM-C contained the highest concentration of saturated NEFA. DM and DM-C secretomes promoted inflammation and cytotoxicity on AC16 cardiomyocytes. Exogenous Cer16:0, Cer24:1, and palmitic acid reproduced deleterious effects in AC16 cells. These effects were attenuated by exogenous apoJ. Diabetic secretomes promoted inflammation and cytotoxicity in cardiomyocytes. This effect was exacerbated in the secretomes of the DM-C samples. The increased content of specific NEFA and ceramide species seems to play a key role in inducing such deleterious effects, which are attenuated by apoJ.
Collapse
Affiliation(s)
- Núria Puig
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), Barcelona, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - José Rives
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), Barcelona, Spain; Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - Pedro Gil-Millan
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, and IR-Sant Pau, Barcelona, Spain
| | - Inka Miñambres
- Endocrinology Department, Hospital de la Santa Creu i Sant Pau, and IR-Sant Pau, Barcelona, Spain
| | - Antonino Ginel
- Cardiology Department, Hospital de la Santa Creu i Sant Pau, and IR-Sant Pau, Barcelona, Spain
| | - Manel Tauron
- Cardiology Department, Hospital de la Santa Creu i Sant Pau, and IR-Sant Pau, Barcelona, Spain
| | - Anna Bonaterra-Pastra
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antonio Pérez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona 08193, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - José Luís Sánchez-Quesada
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain.
| | - Sonia Benitez
- Cardiovascular Biochemistry, Institut de Recerca Sant Pau (IR-Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Madrid, Spain.
| |
Collapse
|
12
|
Tricò D, Chiriacò M, Nouws J, Vash-Margita A, Kursawe R, Tarabra E, Galderisi A, Natali A, Giannini C, Hellerstein M, Ferrannini E, Caprio S. Alterations in Adipose Tissue Distribution, Cell Morphology, and Function Mark Primary Insulin Hypersecretion in Youth With Obesity. Diabetes 2024; 73:941-952. [PMID: 37870826 PMCID: PMC11109779 DOI: 10.2337/db23-0450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023]
Abstract
Excessive insulin secretion independent of insulin resistance, defined as primary hypersecretion, is associated with obesity and an unfavorable metabolic phenotype. We examined the characteristics of adipose tissue of youth with primary insulin hypersecretion and the longitudinal metabolic alterations influenced by the complex adipo-insular interplay. In a multiethnic cohort of adolescents with obesity but without diabetes, primary insulin hypersecretors had enhanced model-derived β-cell glucose sensitivity and rate sensitivity but worse glucose tolerance, despite similar demographics, adiposity, and insulin resistance measured by both oral glucose tolerance test and euglycemic-hyperinsulinemic clamp. Hypersecretors had greater intrahepatic and visceral fat depots at abdominal MRI, hypertrophic abdominal subcutaneous adipocytes, higher free fatty acid and leptin serum levels per fat mass, and faster in vivo lipid turnover assessed by a long-term 2H2O labeling protocol. At 2-year follow-up, hypersecretors had greater fat accrual and a threefold higher risk for abnormal glucose tolerance, while individuals with hypertrophic adipocytes or higher leptin levels showed enhanced β-cell glucose sensitivity. Primary insulin hypersecretion is associated with marked alterations in adipose tissue distribution, cellularity, and lipid dynamics, independent of whole-body adiposity and insulin resistance. Pathogenetic insight into the metabolic crosstalk between β-cell and adipocyte may help to identify individuals at risk for chronic hyperinsulinemia, body weight gain, and glucose intolerance. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Domenico Tricò
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Laboratory of Metabolism, Nutrition, and Atherosclerosis, University of Pisa, Pisa, Italy
| | - Martina Chiriacò
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Laboratory of Metabolism, Nutrition, and Atherosclerosis, University of Pisa, Pisa, Italy
| | - Jessica Nouws
- Department of Pediatrics, Yale School of Medicine, New Haven, CT
| | - Alla Vash-Margita
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
| | | | | | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Laboratory of Metabolism, Nutrition, and Atherosclerosis, University of Pisa, Pisa, Italy
| | - Cosimo Giannini
- Department of Pediatrics, University of Chieti “G. d’Annunzio,” Chieti, Italy
| | - Marc Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Ele Ferrannini
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Sonia Caprio
- Department of Pediatrics, Yale School of Medicine, New Haven, CT
| |
Collapse
|
13
|
Cabeza de Baca T, Parrington S, Votruba S, Piaggi P, Krakoff J, Chang DC. Adipocyte size, adipose tissue calories, and circulating adipokines, before and after diet-induced weight loss in humans. Endocrine 2024; 84:490-499. [PMID: 38172345 DOI: 10.1007/s12020-023-03666-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Abstract
OBJECTIVE Adipose tissue (AT) contains a bimodal population of large and small adipocytes. Changes in fat cell size (FCS) distribution and AT caloric density (kcal/g) with weight loss are unclear. We aimed to evaluate changes in FCS and AT calories in weight loss and determine associations with anthropometrics. MATERIALS AND METHODS Healthy adults (6 men/4 women; age 33 ± 11 years; BMI 35 ± 6 kg/m2) underwent DXA and subcutaneous abdominal/thigh fat biopsies, before and after 6 weeks of caloric restriction. AT calories (bomb calorimetry) and hormones (adiponectin, leptin, FGF21) were measured. RESULTS Abdominal large cell diameter (LCD; Δ = -13.2 μm, p = 0.01) and nadir (Δ = -7.3 μm, p = 0.03) decreased. In repeated measures correlations (rrm), abdominal and thigh LCD and nadir were associated with fat mass (FM) loss (rrm = 0.68; rrm = 0.63; rrm = 0.66; rrm = 0.62, p's < 0.05, respectively) and waist circumference decrease (rrm = 0.70; rrm = 0.60, p's ≤ 0.05). Small cell percentage did not change and was not associated with FM changes. Abdominal AT calories were unchanged with weight loss. Change in leptin was associated with change in abdominal LCD (rrm = 0.77, p = 0.01). CONCLUSIONS Caloric restriction reduces adipocyte LCD and nadir. These changes are associated with FM loss. Larger fat cells should be considered as phenotypic targets for weight loss. CLINICAL TRIALS REGISTRATION clinicaltrials.gov identifier: NCT00687115, May 29, 2008.
Collapse
Affiliation(s)
- Tomás Cabeza de Baca
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA.
| | - Shannon Parrington
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Susanne Votruba
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Paolo Piaggi
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Jonathan Krakoff
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Douglas C Chang
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| |
Collapse
|
14
|
Iacobini C, Vitale M, Haxhi J, Menini S, Pugliese G. Impaired Remodeling of White Adipose Tissue in Obesity and Aging: From Defective Adipogenesis to Adipose Organ Dysfunction. Cells 2024; 13:763. [PMID: 38727299 PMCID: PMC11083890 DOI: 10.3390/cells13090763] [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: 04/02/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
The adipose organ adapts and responds to internal and environmental stimuli by remodeling both its cellular and extracellular components. Under conditions of energy surplus, the subcutaneous white adipose tissue (WAT) is capable of expanding through the enlargement of existing adipocytes (hypertrophy), followed by de novo adipogenesis (hyperplasia), which is impaired in hypertrophic obesity. However, an impaired hyperplastic response may result from various defects in adipogenesis, leading to different WAT features and metabolic consequences, as discussed here by reviewing the results of the studies in animal models with either overexpression or knockdown of the main molecular regulators of the two steps of the adipogenesis process. Moreover, impaired WAT remodeling with aging has been associated with various age-related conditions and reduced lifespan expectancy. Here, we delve into the latest advancements in comprehending the molecular and cellular processes underlying age-related changes in WAT function, their involvement in common aging pathologies, and their potential as therapeutic targets to influence both the health of elderly people and longevity. Overall, this review aims to encourage research on the mechanisms of WAT maladaptation common to conditions of both excessive and insufficient fat tissue. The goal is to devise adipocyte-targeted therapies that are effective against both obesity- and age-related disorders.
Collapse
|
15
|
Hagberg CE, Spalding KL. White adipocyte dysfunction and obesity-associated pathologies in humans. Nat Rev Mol Cell Biol 2024; 25:270-289. [PMID: 38086922 DOI: 10.1038/s41580-023-00680-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 02/10/2024]
Abstract
The prevalence of obesity and associated chronic diseases continues to increase worldwide, negatively impacting on societies and economies. Whereas the association between excess body weight and increased risk for developing a multitude of diseases is well established, the initiating mechanisms by which weight gain impairs our metabolic health remain surprisingly contested. In order to better address the myriad of disease states associated with obesity, it is essential to understand adipose tissue dysfunction and develop strategies for reinforcing adipocyte health. In this Review we outline the diverse physiological functions and pathological roles of human white adipocytes, examining our current knowledge of why white adipocytes are vital for systemic metabolic control, yet poorly adapted to our current obesogenic environment.
Collapse
Affiliation(s)
- Carolina E Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kirsty L Spalding
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
16
|
Giacobbi AS, Meyer L, Ribot M, Yvinec R, Soula H, Audebert C. Mathematical modeling of adipocyte size distributions: Identifiability and parameter estimation from rat data. J Theor Biol 2024; 581:111747. [PMID: 38278344 DOI: 10.1016/j.jtbi.2024.111747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/05/2023] [Accepted: 01/20/2024] [Indexed: 01/28/2024]
Abstract
Fat cells, called adipocytes, are designed to regulate energy homeostasis by storing energy in the form of lipids. Adipocyte size distribution is assumed to play a role in the development of obesity-related diseases. These cells that do not have a characteristic size, indeed a bimodal size distribution is observed in adipose tissue. We propose a model based on a partial differential equation to describe adipocyte size distribution. The model includes a description of the lipid fluxes and the cell size fluctuations and using a formulation of a stationary solution fast computation of bimodal distribution is achieved. We investigate the parameter identifiability and estimate parameter values with CMA-ES algorithm. We first validate the procedure on synthetic data, then we estimate parameter values with experimental data of 32 rats. We discuss the estimated parameter values and their variability within the population, as well as the relation between estimated values and their biological significance. Finally, a sensitivity analysis is performed to specify the influence of parameters on cell size distribution and explain the differences between the model and the measurements. The proposed framework enables the characterization of adipocyte size distribution with four parameters and can be easily adapted to measurements of cell size distribution in different health conditions.
Collapse
Affiliation(s)
- Anne-Sophie Giacobbi
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratory of Computational and Quantitative Biology UMR 7238, 75005 Paris, France.
| | - Leo Meyer
- Institut Denis Poisson, Université d'Orléans, CNRS, Université de Tours, 45067 Orléans, France
| | - Magali Ribot
- Institut Denis Poisson, Université d'Orléans, CNRS, Université de Tours, 45067 Orléans, France
| | - Romain Yvinec
- PRC, INRAE, CNRS, Université de Tours, 37380 Nouzilly, France; Université Paris-Saclay, Inria, Centre Inria de Saclay, 91120 Palaiseau, France
| | - Hedi Soula
- Nutriomics, La Pitié-Salpétrière, Sorbonne Université, CNRS, 75013 Paris, France
| | - Chloe Audebert
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratory of Computational and Quantitative Biology UMR 7238, 75005 Paris, France; Sorbonne Université, CNRS, Université de Paris, Laboratoire Jacques-Louis Lions UMR 7598, 75005 Paris, France.
| |
Collapse
|
17
|
Elguezabal Rodelo RG, Porchia LM, Torres‐Rasgado E, López-Bayghen E, Gonzalez-Mejia ME. Visceral and subcutaneous abdominal fat is associated with non-alcoholic fatty liver disease while augmenting Metabolic Syndrome's effect on non-alcoholic fatty liver disease: A cross-sectional study of NHANES 2017-2018. PLoS One 2024; 19:e0298662. [PMID: 38394065 PMCID: PMC10889905 DOI: 10.1371/journal.pone.0298662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The aim was to evaluate the effect different types of abdominal fat have on NAFLD development and the effects of abdominal fat has on the association between Metabolic Syndrome (MetS) and NALFD. METHODS Data was collected from the cross-sectional NHANES dataset (2017-2018 cycle). Using the controlled attenuation parameter (USG CAP, dB/m), which measures the level of steatosis, the cohort was stratified into two groups: NAFLD(+) (≥274 dB/m) and NAFLD(-). Using complex samples analyses, associations between liver steatosis or NAFLD and types of abdominal fat area [Total abdominal (TAFA), subcutaneous (SAT), and visceral (VAT)] were determined. Pearson's correlation coefficient (r) was calculated to evaluate the associations between adipose tissues and NAFLD. Logistic regression was used to determine the risk [odds ratio (OR) and 95% confidence interval (95%CI)]. Participants were also classified by MetS, using the Harmonizing Definition criteria. RESULTS Using 1,980 participants (96,282,896 weighted), there was a significant (p<0.001) correlation between USG CAP and TAFA (r = 0.569), VAT (r = 0.645), and SAT (r = 0.479). Additionally, the risk of developing NAFLD was observed for total abdominal obesity (OR = 19.9, 95%CI: 5.1-77.8, p<0.001), visceral obesity (OR = 9.1, 95%CI: 6.2-13.5, p<0.001) and subcutaneous obesity (OR = 4.8, 95%CI: 3.2-6.9, p<0.001). Using 866 participants (44,399,696 weighted), for visceral obesity, participants with MetS and visceral obesity (OR = 18.1, 95%CI: 8.0-41.3, p<0.001) were shown to have a greater risk than participants with MetS only (OR = 6.3, 95%CI: 2.6-15.2, p<0.001). For subcutaneous obesity, again, participants with MetS and subcutaneous obesity (OR = 18.3, 95%CI: 8.0-41.9, p<0.001) were shown to have a greater risk than the MetS-only group (OR = 10.3, 95%CI: 4.8-22.4, p<0.001). CONCLUSION TAFA, VAT, and SAT were positively associated with USG CAP values and increased the risk of developing NAFLD. Also, the type of abdominal fat depots did affect the association between MetS and NAFLD.
Collapse
Affiliation(s)
| | - Leonardo M. Porchia
- Facultad de Medicina, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla, México
| | | | - Esther López-Bayghen
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | | |
Collapse
|
18
|
Mitchelson KAJ, O’Connell F, O’Sullivan J, Roche HM. Obesity, Dietary Fats, and Gastrointestinal Cancer Risk-Potential Mechanisms Relating to Lipid Metabolism and Inflammation. Metabolites 2024; 14:42. [PMID: 38248845 PMCID: PMC10821017 DOI: 10.3390/metabo14010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Obesity is a major driving factor in the incidence, progression, and poor treatment response in gastrointestinal cancers. Herein, we conducted a comprehensive analysis of the impact of obesity and its resulting metabolic perturbations across four gastrointestinal cancer types, namely, oesophageal, gastric, liver, and colorectal cancer. Importantly, not all obese phenotypes are equal. Obese adipose tissue heterogeneity depends on the location, structure, cellular profile (including resident immune cell populations), and dietary fatty acid intake. We discuss whether adipose heterogeneity impacts the tumorigenic environment. Dietary fat quality, in particular saturated fatty acids, promotes a hypertrophic, pro-inflammatory adipose profile, in contrast to monounsaturated fatty acids, resulting in a hyperplastic, less inflammatory adipose phenotype. The purpose of this review is to examine the impact of obesity, including dietary fat quality, on adipose tissue biology and oncogenesis, specifically focusing on lipid metabolism and inflammatory mechanisms. This is achieved with a particular focus on gastrointestinal cancers as exemplar models of obesity-associated cancers.
Collapse
Affiliation(s)
- Kathleen A. J. Mitchelson
- Nutrigenomics Research Group, UCD Conway Institute, UCD Institute of Food and Health, and School of Public Health, Physiotherapy and Sports Science, University College Dublin, D04 H1W8 Dublin, Ireland
| | - Fiona O’Connell
- Department of Surgery, Trinity St. James’s Cancer Institute and Trinity Translational Medicine Institute, St. James’s Hospital and Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Jacintha O’Sullivan
- Department of Surgery, Trinity St. James’s Cancer Institute and Trinity Translational Medicine Institute, St. James’s Hospital and Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Helen M. Roche
- Nutrigenomics Research Group, UCD Conway Institute, UCD Institute of Food and Health, and School of Public Health, Physiotherapy and Sports Science, University College Dublin, D04 H1W8 Dublin, Ireland
- Institute for Global Food Security, School of Biological Sciences, Queens University Belfast, Belfast BT9 5DL, UK
| |
Collapse
|
19
|
Adeva-Andany MM, Domínguez-Montero A, Adeva-Contreras L, Fernández-Fernández C, Carneiro-Freire N, González-Lucán M. Body Fat Distribution Contributes to Defining the Relationship between Insulin Resistance and Obesity in Human Diseases. Curr Diabetes Rev 2024; 20:e160823219824. [PMID: 37587805 DOI: 10.2174/1573399820666230816111624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/28/2023] [Accepted: 05/31/2023] [Indexed: 08/18/2023]
Abstract
The risk for metabolic and cardiovascular complications of obesity is defined by body fat distribution rather than global adiposity. Unlike subcutaneous fat, visceral fat (including hepatic steatosis) reflects insulin resistance and predicts type 2 diabetes and cardiovascular disease. In humans, available evidence indicates that the ability to store triglycerides in the subcutaneous adipose tissue reflects enhanced insulin sensitivity. Prospective studies document an association between larger subcutaneous fat mass at baseline and reduced incidence of impaired glucose tolerance. Case-control studies reveal an association between genetic predisposition to insulin resistance and a lower amount of subcutaneous adipose tissue. Human peroxisome proliferator-activated receptorgamma (PPAR-γ) promotes subcutaneous adipocyte differentiation and subcutaneous fat deposition, improving insulin resistance and reducing visceral fat. Thiazolidinediones reproduce the effects of PPAR-γ activation and therefore increase the amount of subcutaneous fat while enhancing insulin sensitivity and reducing visceral fat. Partial or virtually complete lack of adipose tissue (lipodystrophy) is associated with insulin resistance and its clinical manifestations, including essential hypertension, hypertriglyceridemia, reduced HDL-c, type 2 diabetes, cardiovascular disease, and kidney disease. Patients with Prader Willi syndrome manifest severe subcutaneous obesity without insulin resistance. The impaired ability to accumulate fat in the subcutaneous adipose tissue may be due to deficient triglyceride synthesis, inadequate formation of lipid droplets, or defective adipocyte differentiation. Lean and obese humans develop insulin resistance when the capacity to store fat in the subcutaneous adipose tissue is exhausted and deposition of triglycerides is no longer attainable at that location. Existing adipocytes become large and reflect the presence of insulin resistance.
Collapse
Affiliation(s)
- María M Adeva-Andany
- Nephrology Division, Department of Internal Medicine, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406 Ferrol, Spain
| | - Alberto Domínguez-Montero
- Nephrology Division, Department of Internal Medicine, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406 Ferrol, Spain
| | | | - Carlos Fernández-Fernández
- Nephrology Division, Department of Internal Medicine, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406 Ferrol, Spain
| | - Natalia Carneiro-Freire
- Nephrology Division, Department of Internal Medicine, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406 Ferrol, Spain
| | - Manuel González-Lucán
- Nephrology Division, Department of Internal Medicine, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406 Ferrol, Spain
| |
Collapse
|
20
|
Kudo T, Zhao ML, Jeknić S, Kovary KM, LaGory EL, Covert MW, Teruel MN. Context-dependent regulation of lipid accumulation in adipocytes by a HIF1α-PPARγ feedback network. Cell Syst 2023; 14:1074-1086.e7. [PMID: 37995680 PMCID: PMC11251692 DOI: 10.1016/j.cels.2023.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 12/03/2022] [Accepted: 10/26/2023] [Indexed: 11/25/2023]
Abstract
Hypoxia-induced upregulation of HIF1α triggers adipose tissue dysfunction and insulin resistance in obese patients. HIF1α closely interacts with PPARγ, the master regulator of adipocyte differentiation and lipid accumulation, but there are conflicting results regarding how this interaction controls the excessive lipid accumulation that drives adipocyte dysfunction. To directly address these conflicts, we established a differentiation system that recapitulated prior seemingly opposing observations made across different experimental settings. Using single-cell imaging and coarse-grained mathematical modeling, we show how HIF1α can both promote and repress lipid accumulation during adipogenesis. Our model predicted and our experiments confirmed that the opposing roles of HIF1α are isolated from each other by the positive-feedback-mediated upregulation of PPARγ that drives adipocyte differentiation. Finally, we identify three factors: strength of the differentiation cue, timing of hypoxic perturbation, and strength of HIF1α expression changes that, when considered together, provide an explanation for many of the previous conflicting reports.
Collapse
Affiliation(s)
- Takamasa Kudo
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Michael L Zhao
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Stevan Jeknić
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kyle M Kovary
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Edward L LaGory
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Markus W Covert
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| | - Mary N Teruel
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Biochemistry and the Drukier Institute of Children's Health, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
21
|
Lecoutre S, Maqdasy S, Lambert M, Breton C. The Impact of Maternal Obesity on Adipose Progenitor Cells. Biomedicines 2023; 11:3252. [PMID: 38137473 PMCID: PMC10741630 DOI: 10.3390/biomedicines11123252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
The concept of Developmental Origin of Health and Disease (DOHaD) postulates that adult-onset metabolic disorders may originate from suboptimal conditions during critical embryonic and fetal programming windows. In particular, nutritional disturbance during key developmental stages may program the set point of adiposity and its associated metabolic diseases later in life. Numerous studies in mammals have reported that maternal obesity and the resulting accelerated growth in neonates may affect adipocyte development, resulting in persistent alterations in adipose tissue plasticity (i.e., adipocyte proliferation and storage) and adipocyte function (i.e., insulin resistance, impaired adipokine secretion, reduced thermogenesis, and higher inflammation) in a sex- and depot-specific manner. Over recent years, adipose progenitor cells (APCs) have been shown to play a crucial role in adipose tissue plasticity, essential for its development, maintenance, and expansion. In this review, we aim to provide insights into the developmental timeline of lineage commitment and differentiation of APCs and their role in predisposing individuals to obesity and metabolic diseases. We present data supporting the possible implication of dysregulated APCs and aberrant perinatal adipogenesis through epigenetic mechanisms as a primary mechanism responsible for long-lasting adipose tissue dysfunction in offspring born to obese mothers.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, F-75013 Paris, France
| | - Salwan Maqdasy
- Department of Medicine (H7), Karolinska Institutet Hospital, C2-94, 14186 Stockholm, Sweden;
| | - Mélanie Lambert
- U978 Institut National de la Santé et de la Recherche Médicale, F-93022 Bobigny, France;
- Université Sorbonne Paris Nord, Alliance Sorbonne Paris Cité, Labex Inflamex, F-93000 Bobigny, France
| | - Christophe Breton
- Maternal Malnutrition and Programming of Metabolic Diseases, Université de Lille, EA4489, F-59000 Lille, France
- U1283-UMR8199-EGID, Université de Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, F-59000 Lille, France
| |
Collapse
|
22
|
Hernández-Quiles M, Martinez Campesino L, Morris I, Ilyas Z, Reynolds S, Soon Tan N, Sobrevals Alcaraz P, Stigter ECA, Varga Á, Varga J, van Es R, Vos H, Wilson HL, Kiss-Toth E, Kalkhoven E. The pseudokinase TRIB3 controls adipocyte lipid homeostasis and proliferation in vitro and in vivo. Mol Metab 2023; 78:101829. [PMID: 38445671 PMCID: PMC10663684 DOI: 10.1016/j.molmet.2023.101829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 03/07/2024] Open
Abstract
OBJECTIVE In vivo studies in humans and mice have implicated the pseudokinase Tribbles 3 (TRIB3) in various aspects of energy metabolism. Whilst cell-based studies indicate a role for TRIB3 in adipocyte differentiation and function, it is unclear if and how these cellular functions may contribute to overall metabolic health. METHODS We investigated the metabolic phenotype of whole-body Trib3 knockout (Trib3KO) mice, focusing on adipocyte and adipose tissue functions. In addition, we combined lipidomics, transcriptomics, interactomics and phosphoproteomics analyses to elucidate cell-intrinsic functions of TRIB3 in pre- and mature adipocytes. RESULTS Trib3KO mice display increased adiposity, but their insulin sensitivity remains unaltered. Trib3KO adipocytes are smaller and display higher Proliferating Cell Nuclear Antigen (PCNA) levels, indicating potential alterations in either i) proliferation-differentiation balance, ii) impaired expansion after cell division, or iii) an altered balance between lipid storage and release, or a combination thereof. Lipidome analyses suggest TRIB3 involvement in the latter two processes, as triglyceride storage is reduced and membrane composition, which can restrain cellular expansion, is altered. Integrated interactome, phosphoproteome and transcriptome analyses support a role for TRIB3 in all three cellular processes through multiple cellular pathways, including Mitogen Activated Protein Kinase- (MAPK/ERK), Protein Kinase A (PKA)-mediated signaling and Transcription Factor 7 like 2 (TCF7L2) and Beta Catenin-mediated gene expression. CONCLUSIONS Our findings support TRIB3 playing multiple distinct regulatory roles in the cytoplasm, nucleus and mitochondria, ultimately controlling adipose tissue homeostasis, rather than affecting a single cellular pathway.
Collapse
Affiliation(s)
- Miguel Hernández-Quiles
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3C584 CG Utrecht, The Netherlands
| | - Laura Martinez Campesino
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2TN, UK
| | - Imogen Morris
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3C584 CG Utrecht, The Netherlands
| | - Zabran Ilyas
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2TN, UK
| | - Steve Reynolds
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2TN, UK
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, 308232 Singapore, Singapore; School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, 637551 Singapore, Singapore
| | - Paula Sobrevals Alcaraz
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3C584 CG Utrecht, The Netherlands
| | - Edwin C A Stigter
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3C584 CG Utrecht, The Netherlands
| | - Ákos Varga
- Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - János Varga
- Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - Robert van Es
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3C584 CG Utrecht, The Netherlands
| | - Harmjan Vos
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3C584 CG Utrecht, The Netherlands
| | - Heather L Wilson
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2TN, UK
| | - Endre Kiss-Toth
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2TN, UK
| | - Eric Kalkhoven
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3C584 CG Utrecht, The Netherlands.
| |
Collapse
|
23
|
Palacios-Marin I, Serra D, Jimenez-Chillarón J, Herrero L, Todorčević M. Adipose Tissue Dynamics: Cellular and Lipid Turnover in Health and Disease. Nutrients 2023; 15:3968. [PMID: 37764752 PMCID: PMC10535304 DOI: 10.3390/nu15183968] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 08/30/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
The alarming increase in obesity and its related metabolic health complications, such as type 2 diabetes, has evolved into a global pandemic. Obesity is mainly characterized by excessive accumulation of adipose tissue, primarily due to an imbalance between energy intake and expenditure. Prolonged positive energy balance leads to the expansion of existing adipocytes (hypertrophy) and/or an increase in preadipocyte and adipocyte number (hyperplasia) to accommodate excess energy intake. However, obesity is not solely defined by increases in adipocyte size and number. The turnover of adipose tissue cells also plays a crucial role in the development and progression of obesity. Cell turnover encompasses the processes of cell proliferation, differentiation, and apoptosis, which collectively regulate the overall cell population within adipose tissue. Lipid turnover represents another critical factor that influences how adipose tissue stores and releases energy. Our understanding of adipose tissue lipid turnover in humans remains limited due to the slow rate of turnover and methodological constraints. Nonetheless, disturbances in lipid metabolism are strongly associated with altered adipose tissue lipid turnover. In obesity, there is a decreased rate of triglyceride removal (lipolysis followed by oxidation), leading to the accumulation of triglycerides over time. This review provides a comprehensive summary of findings from both in vitro and in vivo methods used to study the turnover of adipose cells and lipids in metabolic health and disease. Understanding the mechanisms underlying cellular and lipid turnover in obesity is essential for developing strategies to mitigate the adverse effects of excess adiposity.
Collapse
Affiliation(s)
- Ivonne Palacios-Marin
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, E-08950 Barcelona, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Josep Jimenez-Chillarón
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, Esplugues, E-08950 Barcelona, Spain
- Department of Physiological Sciences, School of Medicine, University of Barcelona, E-08907 L’Hospitalet, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Marijana Todorčević
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| |
Collapse
|
24
|
Ahmad S, Drag MH, Mohamad Salleh S, Cai Z, Nielsen MO. Gene coexpression network analysis reveals perirenal adipose tissue as an important target of prenatal malnutrition in sheep. Physiol Genomics 2023; 55:392-413. [PMID: 37458462 PMCID: PMC10642927 DOI: 10.1152/physiolgenomics.00128.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 08/24/2023] Open
Abstract
We have previously demonstrated that pre- and early postnatal malnutrition in sheep induced depot- and sex-specific changes in adipose morphological features, metabolic outcomes, and transcriptome in adulthood, with perirenal (PER) as the major target followed by subcutaneous (SUB) adipose tissue. We aimed to identify coexpressed and hub genes in SUB and PER to identify the underlying molecular mechanisms contributing to the early nutritional programming of adipose-related phenotypic outcomes. Transcriptomes of SUB and PER of male and female adult sheep with different pre- and early postnatal nutrition histories were used to construct networks of coexpressed genes likely to be functionally associated with pre- and early postnatal nutrition histories and phenotypic traits using weighted gene coexpression network analysis. The modules from PER showed enrichment of cell cycle regulation, gene expression, transmembrane transport, and metabolic processes associated with both sexes' prenatal nutrition. In SUB (only males), a module of enriched adenosine diphosphate metabolism and development correlated with prenatal nutrition. Sex-specific module enrichments were found in PER, such as chromatin modification in the male network but histone modification and mitochondria- and oxidative phosphorylation-related functions in the female network. These sex-specific modules correlated with prenatal nutrition and adipocyte size distribution patterns. Our results point to PER as a primary target of prenatal malnutrition compared to SUB, which played only a minor role. The prenatal programming of gene expression and cell cycle, potentially through epigenetic modifications, might be underlying mechanisms responsible for observed changes in PER expandability and adipocyte-size distribution patterns in adulthood in both sexes.
Collapse
Affiliation(s)
- Sharmila Ahmad
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Research Unit of Nutrition, Department of Animal and Veterinary Sciences, Aarhus University, Tjele, Denmark
| | - Markus Hodal Drag
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Copenhagen Zoo, Frederiksberg, Denmark
| | - Suraya Mohamad Salleh
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Department of Animal Nutrition and Management, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Zexi Cai
- Centre for Quantitative Genetics and Genomics, Aarhus University, Tjele, Denmark
| | - Mette Olaf Nielsen
- Research Unit of Nutrition, Department of Animal and Veterinary Sciences, Aarhus University, Tjele, Denmark
| |
Collapse
|
25
|
Abstract
In this review, we provide a brief synopsis of the connections between adipose tissue and metabolic health and highlight some recent developments in understanding and exploiting adipocyte biology. Adipose tissue plays critical roles in the regulation of systemic glucose and lipid metabolism and secretes bioactive molecules possessing endocrine, paracrine, and autocrine functions. Dysfunctional adipose tissue has a detrimental impact on metabolic health and is intimately involved in key aspects of metabolic diseases such as insulin resistance, lipid overload, inflammation, and organelle stress. Differences in the distribution of fat depots and adipose characteristics relate to divergent degrees of metabolic dysfunction found in metabolically healthy and unhealthy obese individuals. Thermogenic adipocytes increase energy expenditure via mitochondrial uncoupling or adenosine triphosphate-consuming futile substrate cycles, while functioning as a metabolic sink and participating in crosstalk with other metabolic organs. Manipulation of adipose tissue provides a wealth of opportunities to intervene and combat the progression of associated metabolic diseases. We discuss current treatment modalities for obesity including incretin hormone analogs and touch upon emerging strategies with therapeutic potential including exosome-based therapy, pharmacological activation of brown and beige adipocyte thermogenesis, and administration or inhibition of adipocyte-derived factors.
Collapse
Affiliation(s)
- Sung-Min An
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| | - Seung-Hee Cho
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| | - John C. Yoon
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
26
|
Ballesteros-Pla C, Sánchez-Alonso MG, Pizarro-Delgado J, Zuccaro A, Sevillano J, Ramos-Álvarez MP. Pleiotrophin and metabolic disorders: insights into its role in metabolism. Front Endocrinol (Lausanne) 2023; 14:1225150. [PMID: 37484951 PMCID: PMC10360176 DOI: 10.3389/fendo.2023.1225150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
Pleiotrophin (PTN) is a cytokine which has been for long studied at the level of the central nervous system, however few studies focus on its role in the peripheral organs. The main aim of this review is to summarize the state of the art of what is known up to date about pleiotrophin and its implications in the main metabolic organs. In summary, pleiotrophin promotes the proliferation of preadipocytes, pancreatic β cells, as well as cells during the mammary gland development. Moreover, this cytokine is important for the structural integrity of the liver and the neuromuscular junction in the skeletal muscle. From a metabolic point of view, pleiotrophin plays a key role in the maintenance of glucose and lipid as well as whole-body insulin homeostasis and favors oxidative metabolism in the skeletal muscle. All in all, this review proposes pleiotrophin as a druggable target to prevent from the development of insulin-resistance-related pathologies.
Collapse
|
27
|
Valenzuela PL, Carrera-Bastos P, Castillo-García A, Lieberman DE, Santos-Lozano A, Lucia A. Obesity and the risk of cardiometabolic diseases. Nat Rev Cardiol 2023; 20:475-494. [PMID: 36927772 DOI: 10.1038/s41569-023-00847-5] [Citation(s) in RCA: 162] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 03/18/2023]
Abstract
The prevalence of obesity has reached pandemic proportions, and now approximately 25% of adults in Westernized countries have obesity. Recognized as a major health concern, obesity is associated with multiple comorbidities, particularly cardiometabolic disorders. In this Review, we present obesity as an evolutionarily novel condition, summarize the epidemiological evidence on its detrimental cardiometabolic consequences and discuss the major mechanisms involved in the association between obesity and the risk of cardiometabolic diseases. We also examine the role of potential moderators of this association, with evidence for and against the so-called 'metabolically healthy obesity phenotype', the 'fatness but fitness' paradox or the 'obesity paradox'. Although maintenance of optimal cardiometabolic status should be a primary goal in individuals with obesity, losing body weight and, particularly, excess visceral adiposity seems to be necessary to minimize the risk of cardiometabolic diseases.
Collapse
Affiliation(s)
- Pedro L Valenzuela
- Physical Activity and Health Research Group (PaHerg), Research Institute of Hospital 12 de Octubre ("i + 12"), Madrid, Spain.
- Department of Systems Biology, University of Alcalá, Alcalá de Henares, Spain.
| | - Pedro Carrera-Bastos
- Center for Primary Health Care Research, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain
| | | | - Daniel E Lieberman
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Alejandro Santos-Lozano
- Physical Activity and Health Research Group (PaHerg), Research Institute of Hospital 12 de Octubre ("i + 12"), Madrid, Spain
- Department of Health Sciences, European University Miguel de Cervantes, Valladolid, Spain
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain.
- CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain.
| |
Collapse
|
28
|
Pourdashti S, Faridi N, Monem-Homaie F, Yaghooti SH, Soroush A, Bathaie SZ. The size of human subcutaneous adipocytes, but not adiposity, is associated with inflammation, endoplasmic reticulum stress, and insulin resistance markers. Mol Biol Rep 2023; 50:5755-5765. [PMID: 37219669 PMCID: PMC10289932 DOI: 10.1007/s11033-023-08460-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/13/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND The fat storage capacity of the adipose tissue prevents ectopic lipid deposition, which is one of the risk factors for metabolic abnormalities in obesity. This capacity depends upon the adipogenic gene expression and blood supply provision for tissue expansion through angiogenesis. Here, we studied hyperplasia/hypertrophy of subcutaneous white adipose tissue (scWAT) concerning adipogenic gene expression, angiogenic status, and metabolic parameters in non-obese and different classes of obese individuals. METHODS The scWAT samples were collected from 80 individuals. The anthropometric parameters, adipose tissue cell size, serum biochemistry, ER stress-induced XBP1 splicing, PPARγ2, SFRP1, WNT10B, and VEGFA gene expression levels were studied. In addition, the CD31 level was investigated by Western blotting. RESULTS The obese individuals had greater waist circumferences and higher serum TG, TC, insulin, and HOMA-IR than the non-obese group. However, the largest adipocyte size, increased TNFα, insulin, and HOMA-IR, and the highest expression level of sXBP1, WNT10B, and VEGFA were observed in Class I obese individuals. It means that inflammation, insulin resistance, and ER stress accompany hypertrophic scWAT adipocytes with limited adipose tissue expansion ability. Furthermore, the Class II + III obese individuals showed high PPARγ2 expression and CD31 levels. There is adipogenesis through hyperplasia in this group. The SFRP1 expression was not significantly different in the studied groups. CONCLUSION The results suggest that the capability of adipogenesis with inadequate angiogenesis is related to the metabolic status, inflammation, and ER function. Therefore, therapeutic strategies that support both angiogenesis and adipogenesis can effectively prevent the complications of obesity.
Collapse
Affiliation(s)
- Sara Pourdashti
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), P.O. Box: 14155-331, Tehran, Iran
| | - Nassim Faridi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), P.O. Box: 14155-331, Tehran, Iran
| | - Forouzandeh Monem-Homaie
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), P.O. Box: 14155-331, Tehran, Iran
| | - S Hamid Yaghooti
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), P.O. Box: 14155-331, Tehran, Iran
| | - Ahmadreza Soroush
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular- Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - S Zahra Bathaie
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University (TMU), P.O. Box: 14155-331, Tehran, Iran.
- UCLA-DOE Institute, University of California, Los Angeles (UCLA), CA, USA.
| |
Collapse
|
29
|
Jialal I, Adams-Huet B, Devaraj S. Increased Adipocyte Hypertrophy in Patients with Nascent Metabolic Syndrome. J Clin Med 2023; 12:4247. [PMID: 37445281 DOI: 10.3390/jcm12134247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Background and Aims: Metabolic Syndrome (MetS), a global problem, predisposes to an increased risk for type 2 diabetes and premature cardiovascular disease. While MetS is associated with central obesity, there is scanty data on adipocyte hypertrophy, increased fat cell size (FCS), in MetS. The aim of this study was to investigate FCS status in adipose tissue (AT) biopsy of patients with nascent MetS without the confounding of diabetes, cardiovascular disease, smoking, or lipid therapy. Methods and Results: Fasting blood and subcutaneous gluteal AT biopsies were obtained in MetS (n = 20) and controls (n = 19). Cardio-metabolic features, FFA levels, hsCRP, and HOMA-IR were significantly increased in patients with MetS. Waist-circumference (WC) adjusted-FCS was significantly increased in patients with MetS and increased with increasing severity of MetS. Furthermore, there were significant correlations between FCS with glucose, HDL-C, and the ratio of TG: HDL-C. There were significant correlations between FCS and FFA, as well as endotoxin and monocyte TLR4 abundance. Additionally, FCS correlated with readouts of NLRP3 Inflammasome activity. Most importantly, FCS correlated with markers of fibrosis and angiogenesis. Conclusions: In conclusion, in patients with nascent MetS, we demonstrate WC-adjusted increase in FCS from gluteal adipose tissue which correlated with cellular inflammation, fibrosis, and angiogenesis. While these preliminary observations were in gluteal fat, future studies are warranted to confirm these findings in visceral and other fat depots.
Collapse
Affiliation(s)
- Ishwarlal Jialal
- Veterans Affairs Medical Center, Mather, CA 95655, USA
- UCDavis School of Medicine and VA Medical Center, 10535 Hospital Way, Mather, CA 95655, USA
| | | | - Sridevi Devaraj
- Texas Children's Hospital and Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
30
|
Ma Y, Zhen Y, Wang M, Gao L, Dang Y, Shang J, Chen X, Ma S, Zhou K, Feng K, Xin Y, Hou Y, Guo C. Associations between the serum triglyceride-glucose index and pericoronary adipose tissue attenuation and plaque features using dual-layer spectral detector computed tomography: a cross-sectional study. Front Endocrinol (Lausanne) 2023; 14:1166117. [PMID: 37234808 PMCID: PMC10206237 DOI: 10.3389/fendo.2023.1166117] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND AND AIMS The triglyceride-glucose (TyG) index is a reliable alternative marker for insulin resistance (IR). Pericoronary adipose tissue (PCAT) can indirectly reflect coronary inflammation. IR and coronary inflammation play a key role in the development and progression of coronary atherosclerosis. Therefore, this study investigated the relationships between the TyG index, PCAT and atherosclerotic plaque characteristics to explore whether IR might lead to coronary artery atherosclerosis progression by inducing coronary inflammation. METHODS We retrospectively collected data on patients with chest pain who underwent coronary computed tomography angiography using spectral detector computed tomography at our institution from June to December 2021. The patients were grouped based on their TyG index levels: T1 (low), T2 (medium), and T3 (high). Each patient was assessed for total plaque volume, plaque load, maximum stenosis, the plaque component volume proportion, high-risk plaques(HRPs), and plaque characteristics (including low attenuation plaques, positive remodeling, a napkin ring sign, and spot calcification). PCAT quantification was performed on the proximal right coronary artery using the fat attenuation index (FAI) measured from a conventional multicolor computed tomography image (FAI120kVp), a spectral virtual single-energy image (FAI40keV), and the slope of the spectral HU curve (λHU). RESULTS We enrolled 201 patients. The proportion of patients with maximum plaque stenosis, positive remodeling, low-density plaques, and HRPs increased as the TyG index level increased. Moreover, the FAI40keV and λHU significantly differed among the three groups, and we identified good positive correlations between FAI40keV and λHU and the TyG index (r = 0.319, P <0.01 and r = 0.325, P <0.01, respectively). FAI120kVp did not significantly differ among the groups. FAI40keV had the highest area under the curve, with an optimal cutoff value of -130.5 HU for predicting a TyG index value of ≥9.13. The multivariate linear regression analysis demonstrated that FAI40keV and λHU were independently positively related to a high TyG index level (standardized regression coefficients: 0.117 [P <0.001] and 0.134 [P <0.001], respectively). CONCLUSIONS Patients with chest pain and a higher TyG index level were more likely to have severe stenosis and HRPs. Moreover, FAI40keV and λHU had good correlations with the serum TyG index, which may noninvasively reflect PCAT inflammation under insulin resistance. These results could help explain the mechanism of plaque progression and instability in patients with insulin resistance might be related to IR-induced coronary inflammation.
Collapse
Affiliation(s)
- Yue Ma
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanhua Zhen
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Min Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lingfeng Gao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuxue Dang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jin Shang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xujiao Chen
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shaowei Ma
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ke Zhou
- Department of Cardiac Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kai Feng
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Xin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Hou
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chuanji Guo
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
31
|
Ibáñez CA, Lira-León G, Reyes-Castro LA, Rodríguez-González GL, Lomas-Soria C, Hernández-Rojas A, Bravo-Flores E, Solis-Paredes JM, Estrada-Gutierrez G, Zambrano E. Programming Mechanism of Adipose Tissue Expansion in the Rat Offspring of Obese Mothers Occurs in a Sex-Specific Manner. Nutrients 2023; 15:nu15102245. [PMID: 37242132 DOI: 10.3390/nu15102245] [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: 03/11/2023] [Revised: 04/19/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
We investigated whether excessive retroperitoneal adipose tissue (AT) expansion programmed by maternal obesity (MO) affects adipocyte size distribution and gene expression in relation to adipocyte proliferation and differentiation in male and female offspring (F1) from control (F1C) and obese (F1MO) mothers. Female Wistar rats (F0) ate a control or high-fat diet from weaning through pregnancy and lactation. F1 were weaned onto a control diet and euthanized at 110 postnatal days. Fat depots were weighed to estimate the total AT. Serum glucose, triglyceride, leptin, insulin, and the insulin resistance index (HOMA-IR) were determined. Adipocyte size and adipogenic gene expression were examined in retroperitoneal fat. Body weight, retroperitoneal AT and adipogenesis differed between male and female F1Cs. Retroperitoneal AT, glucose, triglyceride, insulin, HOMA-IR and leptin were higher in male and female F1MO vs. F1C. Small adipocytes were reduced in F1MO females and absent in F1MO males; large adipocytes were increased in F1MO males and females vs. F1C. Wnt, PI3K-Akt, and insulin signaling pathways in F1MO males and Egr2 in F1MO females were downregulated vs. F1C. MO induced metabolic dysfunction in F1 through different sex dimorphism mechanisms, including the decreased expression of pro-adipogenic genes and reduced insulin signaling in males and lipid mobilization-related genes in females.
Collapse
Affiliation(s)
- Carlos A Ibáñez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Gabriela Lira-León
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Luis A Reyes-Castro
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Guadalupe L Rodríguez-González
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Consuelo Lomas-Soria
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
- CONACyT-Cátedras, Investigador por México, Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City 14080, Mexico
| | - Alejandra Hernández-Rojas
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Eyerahí Bravo-Flores
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Juan Mario Solis-Paredes
- Departamento de Investigación en Salud Reproductiva y Perinatal, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Guadalupe Estrada-Gutierrez
- Dirección de Investigación, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Elena Zambrano
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| |
Collapse
|
32
|
White U. Adipose tissue expansion in obesity, health, and disease. Front Cell Dev Biol 2023; 11:1188844. [PMID: 37181756 PMCID: PMC10169629 DOI: 10.3389/fcell.2023.1188844] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/14/2023] [Indexed: 05/16/2023] Open
Abstract
White adipose tissue (WAT) expands under physiological conditions via an increase in adipocyte size (hypertrophy) and/or number (hyperplasia; adipogenesis), and the ability of WAT to expand to accommodate energy demands is a significant determinant of metabolic health status. Obesity is associated with impaired WAT expansion and remodeling, which results in the deposition of lipids to other non-adipose organs, leading to metabolic derangements. Although increased hyperplasia has been implicated as a cornerstone in promoting healthy WAT expansion, recent developments suggest that the role of adipogenesis as a contributing factor in the transition from impaired subcutaneous WAT expansion to impaired metabolic health remains up for debate. This mini-review will summarize recent developments and highlight emerging concepts on the features of WAT expansion and turnover, and the significance in obesity, health, and disease.
Collapse
Affiliation(s)
- Ursula White
- Clinical Science Division, LSU Pennington Biomedical Research Center, Baton Rouge, LA, United States
| |
Collapse
|
33
|
Maniyadath B, Zhang Q, Gupta RK, Mandrup S. Adipose tissue at single-cell resolution. Cell Metab 2023; 35:386-413. [PMID: 36889280 PMCID: PMC10027403 DOI: 10.1016/j.cmet.2023.02.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 03/09/2023]
Abstract
Adipose tissue exhibits remarkable plasticity with capacity to change in size and cellular composition under physiological and pathophysiological conditions. The emergence of single-cell transcriptomics has rapidly transformed our understanding of the diverse array of cell types and cell states residing in adipose tissues and has provided insight into how transcriptional changes in individual cell types contribute to tissue plasticity. Here, we present a comprehensive overview of the cellular atlas of adipose tissues focusing on the biological insight gained from single-cell and single-nuclei transcriptomics of murine and human adipose tissues. We also offer our perspective on the exciting opportunities for mapping cellular transitions and crosstalk, which have been made possible by single-cell technologies.
Collapse
Affiliation(s)
- Babukrishna Maniyadath
- Center for Functional Genomics and Tissue Plasticity, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Qianbin Zhang
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rana K Gupta
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark.
| |
Collapse
|
34
|
Nikołajuk A, Stefanowicz M, Strączkowski M, Karczewska-Kupczewska M. Changes in Adipose Tissue Gene Expression of the Core Components of the Hippo Signaling Pathway in Young Adults with Uncomplicated Overweight or Obesity Following Weight Loss. J Nutr 2023; 153:665-672. [PMID: 36805181 DOI: 10.1016/j.tjnut.2023.01.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Appropriate adipogenesis leads to the "healthy" expansion of adipose tissue and is a crucial component in maintaining metabolic homeostasis. The Hippo signaling network may balance adipocyte proliferation/differentiation regulating adipogenic footpath. OBJECTIVES Our study aimed to assess subcutaneous adipose tissue (SAT) expression of genes involved in Hippo signaling network in subjects with marked overweight or obesity after dietary intervention (DI) in relation to obesity and insulin sensitivity. METHODS Forty overweight or obese subjects (O/O) [mean ± SD age 33 ± 7 y, 45% men, BMI (in kg/m2) 32.9 ± 3.1] completed DI [low-calorie diet (20 kcal/kg of proper body weight) for 12 wks]. The control group comprising 20 normal-weight subjects (mean ± SD age: 24 ± 2 y, 40% men, BMI: 22.4 ± 2.3 ) was examined at baseline only. Hyperinsulinemic-euglycemic clamp and SAT biopsy with gene expression analysis were performed. Student's t-test for unpaired and paired samples and Pearson correlation analysis were applied. This is an exploratory analysis of the DI program. RESULTS SAT mRNA expression of mammalian sterile 20-like kinase 2 (MST2) encoded by serine/threonine kinase 3 gene (STK3)-->, large tumor suppressor kinase 2 (LATS2), and salvador family WW domain containing protein 1 (SAV1), the upstream members of the Hippo pathway, were decreased (21%, 40%, and 36%, respectively) in O/O in comparison with weight subjects individuals before DI (all P < 0.05). At baseline, positive correlations between SAT SAV1, LATS2 expression and adiponectin (ADIPOQ) (r = 0.50, P < 0.001; r = 0.53, P = 0.004, respectively) and solute carrier family 2 member 4 (SLC2A4) (r = 0.35, P = 0.007; r = 0.28, P = 0.03, respectively) expression were observed in the entire study group. Body weight of the O/O group decreased during DI (11.2 ± 3.8 kg, P < 0.001), and there was an increase in insulin sensitivity (by 27%) and SAT expression of STK3, LATS2 (both by 19%), and SAV1 (by 26%) (all P < 0.05). After DI, SAT SLC2A4 expression was correlated with STK3 (r = 0.47, P = 0.003), LATS2 (r = 0.56, P < 0.001), and yes-associated protein (r = 0.50, P = 0.001) expression. CONCLUSIONS Obesity is associated with altered mRNA expression of upstream effectors of the Hippo pathway in SAT in young adults. DI may improve adipogenic capacity. J Nutr 20XX;xx:xx-xx.
Collapse
Affiliation(s)
- Agnieszka Nikołajuk
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland.
| | - Magdalena Stefanowicz
- Department of Metabolic Diseases, Medical University of Białystok, Białystok, Poland
| | - Marek Strączkowski
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | | |
Collapse
|
35
|
Kologrivova IV, Naryzhnaya NV, Koshelskaya OA, Suslova TE, Kravchenko ES, Kharitonova OA, Evtushenko VV, Boshchenko AA. Association of Epicardial Adipose Tissue Adipocytes Hypertrophy with Biomarkers of Low-Grade Inflammation and Extracellular Matrix Remodeling in Patients with Coronary Artery Disease. Biomedicines 2023; 11:biomedicines11020241. [PMID: 36830779 PMCID: PMC9953115 DOI: 10.3390/biomedicines11020241] [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: 12/23/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
The aim of the study was to compare the morphological features of epicardial adipose tissue (EAT) adipocyte with the circulating inflammatory biomarkers and parameters of extracellular matrix remodeling in patients with coronary artery disease (CAD). We recruited 42 patients with CAD (m/f 28/14) who were scheduled for coronary artery bypass graft surgery (CABG). EAT adipocytes were obtained by the enzymatic method from intraoperative adipose tissue samples. Concentrations of secreted and lipoprotein-associated phospholipase A2 (sPLA2 and LpPLA2), TNF-α, IL-1β, IL-6, IL-10, high-sensitive C-reactive protein (hsCRP), metalloproteinase-9 (MMP-9), MMP-2, C-terminal cross-linking telopeptide of type I collagen (CTX-I), and tissue inhibitor of metalloproteinase 1 (TIMP-1) were measured in blood serum. Patients were divided into two groups: group 1-with mean EAT adipocytes' size ≤ 87.32 μm; group 2-with mean EAT adipocytes' size > 87.32 μm. Patients of group 2 had higher concentrations of triglycerides, hsCRP, TNF-α, and sPLA2 and a lower concentration of CTX-I. A multiple logistic regression model was created (RN2 = 0.43, p = 0.0013). Concentrations of TNF-α, sPLA2 and CTX-I appeared to be independent determinants of the EAT adipocyte hypertrophy. ROC analysis revealed the 78% accuracy, 71% sensitivity, and 85% specificity of the model, AUC = 0.82. According to our results, chronic low-grade inflammation and extracellular matrix remodeling are closely associated with the development of hypertrophy of EAT adipocytes, with serum concentrations of TNF-α, sPLA2 and CTX-I being the key predictors, describing the variability of epicardial adipocytes' size.
Collapse
Affiliation(s)
- Irina V. Kologrivova
- Correspondence: (I.V.K.); (N.V.N.); Tel.: +79-131-053-869 (I.V.K.); +79-039-542-139 (N.V.N.)
| | - Natalia V. Naryzhnaya
- Correspondence: (I.V.K.); (N.V.N.); Tel.: +79-131-053-869 (I.V.K.); +79-039-542-139 (N.V.N.)
| | | | | | | | | | | | | |
Collapse
|
36
|
Bushman T, Lin TY, Chen X. Depot-Dependent Impact of Time-Restricted Feeding on Adipose Tissue Metabolism in High Fat Diet-Induced Obese Male Mice. Nutrients 2023; 15:238. [PMID: 36615895 PMCID: PMC9823673 DOI: 10.3390/nu15010238] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/04/2023] Open
Abstract
Time-restricted feeding (TRF) is known to be an effective strategy for weight loss and metabolic health. TRF's effect on metabolism is complex and likely acts on various pathways within multiple tissues. Adipose tissue plays a key role in systemic homeostasis of glucose and lipid metabolism. Adipose tissue dysregulation has been causally associated with metabolic disorders in obesity. However, it is largely unknown how TRF impacts metabolic pathways such as lipolysis, lipogenesis, and thermogenesis within different in adipose tissue depots in obesity. To determine this, we conducted a 10-week TRF regimen in male mice, previously on a long-term high fat diet (HFD) and subjected the mice to TRF of a HFD for 10 h per day or ad libitum. The TRF regimen showed reduction in weight gain. TRF restored HFD-induced impairment of adipogenesis and increased lipid storage in white adipose tissues. TRF also showed a depot-dependent effect in lipid metabolism and restored ATP-consuming futile cycle of lipogenesis and lipolysis that is impaired by HFD within epididymal adipose tissue, but not inguinal fat depot. We demonstrate that TRF may be a beneficial option as a dietary and lifestyle intervention in lowering bodyweight and improving adipose tissue metabolism.
Collapse
Affiliation(s)
| | | | - Xiaoli Chen
- Department of Food Science and Nutrition, University of Minnesota, Twin Cities, MN 55108, USA
| |
Collapse
|
37
|
Dickson E, Fryklund C, Soylu-Kucharz R, Sjögren M, Stenkula KG, Björkqvist M. Altered Adipocyte Cell Size Distribution Prior to Weight Loss in the R6/2 Model of Huntington's Disease. J Huntingtons Dis 2023; 12:253-266. [PMID: 37718850 DOI: 10.3233/jhd-230587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
BACKGROUND Metabolic alterations contribute to disease onset and prognosis of Huntington's disease (HD). Weight loss in the R6/2 mouse model of HD is a consistent feature, with onset in mid-to-late stage of disease. OBJECTIVE In the present study, we aimed to investigate molecular and functional changes in white adipose tissue (WAT) that occur at weight loss in R6/2 mice. We further elaborated on the effect of leptin-deficiency and early obesity in R6/2 mice. METHODS We performed analyses at 12 weeks of age; a time point that coincides with the start of weight loss in our R6/2 mouse colony. Gonadal (visceral) and inguinal (subcutaneous) WAT depot weights were monitored, as well as adipocyte size distribution. Response to isoprenaline-stimulated glycerol release and insulin-stimulated glucose uptake in adipocytes from gonadal WAT was assessed. RESULTS In R6/2 mice, WAT depot weights were comparable to wildtype (WT) mice, and the response to insulin and isoprenaline in gonadal adipocytes was unaltered. Leptin-deficient R6/2 mice exhibited distinct changes compared to leptin-deficient WT mice. At 12 weeks, female leptin-deficient R6/2 mice had reduced body weight accompanied by an increased proportion of smaller adipocytes, while in contrast; male mice displayed a shift towards larger adipocyte sizes without a significant body weight reduction at this timepoint. CONCLUSIONS We here show that there are early sex-specific changes in adipocyte cell size distribution in WAT of R6/2 mice and leptin-deficient R6/2 mice.
Collapse
Affiliation(s)
- Elna Dickson
- Brain Disease Biomarker Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Claes Fryklund
- Glucose Transport and Protein Trafficking, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Rana Soylu-Kucharz
- Brain Disease Biomarker Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Marie Sjögren
- Brain Disease Biomarker Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Karin G Stenkula
- Glucose Transport and Protein Trafficking, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Maria Björkqvist
- Brain Disease Biomarker Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
38
|
High-fat diet consumption by male rat offspring of obese mothers exacerbates adipose tissue hypertrophy and metabolic alterations in adult life. Br J Nutr 2022:1-10. [PMID: 36412162 DOI: 10.1017/s0007114522003737] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Obese mothers' offspring develop obesity and metabolic alterations in adulthood. Poor postnatal dietary patterns also contribute to obesity and its comorbidities. We aimed to determine whether in obese mothers' offspring an adverse postnatal environment, such as high-fat diet (HFD) consumption (second hit) exacerbates body fat accumulation, metabolic alterations and adipocyte size distribution. Female Wistar rats ate chow (C-5 %-fat) or HFD (maternal obesity (MO)-25 %-fat) from weaning until the end of lactation. Male offspring were weaned on either control (C/C and MO/C, maternal diet/offspring diet) or HFD (C/HF and MO/HF) diet. At 110 postnatal days, offspring were killed. Fat depots were excised to estimate adiposity index (AI). Serum glucose, triglyceride, leptin, insulin, insulin resistance index (HOMA-IR), corticosterone and dehydroepiandrosterone (DHEA) were determined. Adipocyte size distribution was evaluated in retroperitoneal fat. Body weight was similar in C/C and MO/C but higher in C/HF and MO/HF. AI, leptin, insulin and HOMA-IR were higher in MO/C and C/HF v. C/C but lower than MO/HF. Glucose increased in MO/HF v. MO/C. C/HF and MO/C had higher triglyceride and corticosterone than C/C, but lower corticosterone than MO/HF. DHEA and the DHEA/corticosterone ratio were lower in C/HF and MO/C v. C/C, but higher than MO/HF. Small adipocyte proportion decreased while large adipocyte proportions increased in MO/C and C/HF v. C/C and exacerbated in MO/HF v. C/HF. Postnatal consumption of a HFD by the offspring of obese mothers exacerbates body fat accumulation as well as the decrease of small and the increase of large adipocytes, which leads to larger metabolic abnormalities.
Collapse
|
39
|
Lin Z, Lin X, Lai Y, Han C, Fan X, Tang J, Mo S, Su J, Liang S, Shang J, Lv X, Guo S, Pang R, Zhou J, Zhang T, Zhang F. Ponatinib modulates the metabolic profile of obese mice by inhibiting adipose tissue macrophage inflammation. Front Pharmacol 2022; 13:1040999. [DOI: 10.3389/fphar.2022.1040999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
Obesity-induced metabolic syndrome is a rapidly growing conundrum, reaching epidemic proportions globally. Chronic inflammation in obese adipose tissue plays a key role in metabolic syndrome with a series of local and systemic effects such as inflammatory cell infiltration and inflammatory cytokine secretion. Adipose tissue macrophages (ATM), as one of the main regulators in this process, are particularly crucial for pharmacological studies on obesity-related metabolic syndrome. Ponatinib, a multi-targeted tyrosine kinase inhibitor originally used to treat leukemia, has recently been found to improve dyslipidemia and atherosclerosis, suggesting that it may have profound effect on metabolic syndrome, although the mechanisms underlying have not yet been revealed. Here we discovered that ponatinib significantly improved insulin sensitivity in leptin deficient obese mice. In addition to that, ponatinib treatment remarkably ameliorated high fat diet-induced hyperlipidemia and inhibited ectopic lipid deposition in the liver. Interestingly, although ponatinib did not reduce but increase the weight of white adipose tissue (WAT), it remarkably suppressed the inflammatory response in WAT and preserved its function. Mechanistically, we showed that ponatinib had no direct effect on hepatocyte or adipocyte but attenuated free fatty acid (FFA) induced macrophage transformation from pro-inflammatory to anti-inflammatory phenotype. Moreover, adipocytes co-cultured with FFA-treated macrophages exhibited insulin resistance, while pre-treat these macrophages with ponatinib can ameliorate this process. These results suggested that the beneficial effects of ponatinib on metabolic disorders are achieved by inhibiting the inflammatory phenotypic transformation of ATMs, thereby maintaining the physiological function of adipose tissue under excessive obesity. The data here not only revealed the novel therapeutic function of ponatinib, but also provided a theoretical basis for the application of multi-target tyrosine kinase inhibitors in metabolic diseases.
Collapse
|
40
|
Thomaz FM, de Jesus Simão J, da Silva VS, Machado MMF, Oyama LM, Ribeiro EB, Alonso Vale MIC, Telles MM. Ginkgo biloba Extract Stimulates Adipogenesis in 3T3-L1 Preadipocytes. Pharmaceuticals (Basel) 2022; 15:ph15101294. [PMID: 36297406 PMCID: PMC9610090 DOI: 10.3390/ph15101294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/08/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Smaller adipocytes are related to the reversal of metabolic disorders, suggesting that molecules that can act in the adipogenesis pathway are of great interest. The objective of this study was to investigate the effect of Ginkgo biloba extract (GbE) in modulating the differentiation in preadipocytes. 3T3-L1 preadipocytes were differentiated for 7 days into adipocytes without (control group) and with GbE at 1.0 mg/mL. Lipid content and gene expression were analyzed on day 7 (D7) by Oil Red O staining and PCR Array Gene Expression. Western blotting analysis of the key adipogenesis markers was evaluated during the differentiation process at days 3 (D3), 5 (D5), and 7 (D7). GbE increased lipid content and raised the gene expression of the main adipogenesis markers. Key proteins of the differentiation process were modulated by GbE, since C/EBPβ levels were decreased, while C/EBPα levels were increased at D7. Regarding the mature adipocytes’ markers, GbE enhanced the levels of both FABP4 at D5, and perilipin at D3 and D5. In summary, the present findings showed that GbE modulated the adipogenesis pathway suggesting that the treatment could accelerate the preadipocyte maturation, stimulating the expression of mature adipocyte proteins earlier than expected.
Collapse
Affiliation(s)
- Fernanda Malanconi Thomaz
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
| | - Jussara de Jesus Simão
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
| | - Viviane Simões da Silva
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
| | - Meira Maria Forcelini Machado
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
| | - Lila Missae Oyama
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo—UNIFESP, São Paulo 04023-062, Brazil
| | - Eliane Beraldi Ribeiro
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo—UNIFESP, São Paulo 04023-062, Brazil
| | - Maria Isabel Cardoso Alonso Vale
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
- Correspondence:
| | - Monica Marques Telles
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo—UNIFESP, São Paulo 04023-062, Brazil
| |
Collapse
|
41
|
Cechinel LR, Batabyal RA, Freishtat RJ, Zohn IE. Parental obesity-induced changes in developmental programming. Front Cell Dev Biol 2022; 10:918080. [PMID: 36274855 PMCID: PMC9585252 DOI: 10.3389/fcell.2022.918080] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Many studies support the link between parental obesity and the predisposition to develop adult-onset metabolic syndromes that include obesity, high blood pressure, dyslipidemia, insulin resistance, and diabetes in the offspring. As the prevalence of obesity increases in persons of childbearing age, so does metabolic syndrome in their descendants. Understanding how parental obesity alters metabolic programs in the progeny, predisposing them to adult-onset metabolic syndrome, is key to breaking this cycle. This review explores the basis for altered metabolism of offspring exposed to overnutrition by focusing on critical developmental processes influenced by parental obesity. We draw from human and animal model studies, highlighting the adaptations in metabolism that occur during normal pregnancy that become maladaptive with obesity. We describe essential phases of development impacted by parental obesity that contribute to long-term alterations in metabolism in the offspring. These encompass gamete formation, placentation, adipogenesis, pancreas development, and development of brain appetite control circuits. Parental obesity alters the developmental programming of these organs in part by inducing epigenetic changes with long-term consequences on metabolism. While exposure to parental obesity during any of these phases is sufficient to alter long-term metabolism, offspring often experience multiple exposures throughout their development. These insults accumulate to increase further the susceptibility of the offspring to the obesogenic environments of modern society.
Collapse
|
42
|
Steckelings UM, Widdop RE, Sturrock ED, Lubbe L, Hussain T, Kaschina E, Unger T, Hallberg A, Carey RM, Sumners C. The Angiotensin AT 2 Receptor: From a Binding Site to a Novel Therapeutic Target. Pharmacol Rev 2022; 74:1051-1135. [PMID: 36180112 PMCID: PMC9553111 DOI: 10.1124/pharmrev.120.000281] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/19/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
Discovered more than 30 years ago, the angiotensin AT2 receptor (AT2R) has evolved from a binding site with unknown function to a firmly established major effector within the protective arm of the renin-angiotensin system (RAS) and a target for new drugs in development. The AT2R represents an endogenous protective mechanism that can be manipulated in the majority of preclinical models to alleviate lung, renal, cardiovascular, metabolic, cutaneous, and neural diseases as well as cancer. This article is a comprehensive review summarizing our current knowledge of the AT2R, from its discovery to its position within the RAS and its overall functions. This is followed by an in-depth look at the characteristics of the AT2R, including its structure, intracellular signaling, homo- and heterodimerization, and expression. AT2R-selective ligands, from endogenous peptides to synthetic peptides and nonpeptide molecules that are used as research tools, are discussed. Finally, we summarize the known physiological roles of the AT2R and its abundant protective effects in multiple experimental disease models and expound on AT2R ligands that are undergoing development for clinical use. The present review highlights the controversial aspects and gaps in our knowledge of this receptor and illuminates future perspectives for AT2R research. SIGNIFICANCE STATEMENT: The angiotensin AT2 receptor (AT2R) is now regarded as a fully functional and important component of the renin-angiotensin system, with the potential of exerting protective actions in a variety of diseases. This review provides an in-depth view of the AT2R, which has progressed from being an enigma to becoming a therapeutic target.
Collapse
Affiliation(s)
- U Muscha Steckelings
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert E Widdop
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Edward D Sturrock
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Lizelle Lubbe
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Tahir Hussain
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Elena Kaschina
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Thomas Unger
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Anders Hallberg
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert M Carey
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Colin Sumners
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| |
Collapse
|
43
|
Fryklund C, Neuhaus M, Morén B, Borreguero-Muñoz A, Lundmark R, Stenkula KG. Expansion of the Inguinal Adipose Tissue Depot Correlates With Systemic Insulin Resistance in C57BL/6J Mice. Front Cell Dev Biol 2022; 10:942374. [PMID: 36158197 PMCID: PMC9489915 DOI: 10.3389/fcell.2022.942374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
To accommodate surplus energy, the adipose tissue expands by increasing adipocyte size (hypertrophy) and number (hyperplasia). The presence of hypertrophic adipocytes is a key characteristic of adipose tissue dysfunction. High-fat diet (HFD) fed C57BL/6J mice are a commonly used model to study obesity and obesity-related complications. In the present study, we have characterized adipose plasticity, at both the cellular and tissue level, by examining the temporal development of systemic insulin resistance and adiposity in response to HFD-feeding for 4, 8, and 12 weeks (4w, 8w, and 12w). Within the same time frame, we examined systemic metabolic flexibility and adipose plasticity when switching from HFD- to chow-diet during the last 2 weeks of diet intervention (referred to as the reverse (REV) group: 4wREV (2w HFD+2w chow), 8wREV (6w HFD+2w chow), 12wREV (10w HFD+2w chow)). In response to HFD-feeding over time, the 12w group had impaired systemic insulin sensitivity compared to both the 4w and 8w groups, accompanied by an increase in hypertrophic inguinal adipocytes and liver triglycerides. After reversing from HFD- to chow-feeding, most parameters were completely restored to chow control levels for 4wREV and 8wREV groups. In contrast, the 12wREV group had a significantly increased number of hypertrophic adipocytes, liver triglycerides accumulation, and impaired systemic insulin sensitivity compared to chow-fed mice. Further, image analysis at the single-cell level revealed a cell-size dependent organization of actin filaments for all feeding conditions. Indeed, the impaired adipocyte size plasticity in the 12wREV group was accompanied by increased actin filamentation and reduced insulin-stimulated glucose uptake compared with chow-fed mice. In summary, these results demonstrate that the C57BL/6J HFD-feeding model has a large capacity to restore adipocyte cell size and systemic insulin sensitivity, and that a metabolic tipping point occurs between 8 and 12w of HFD-feeding where this plasticity deteriorates. We believe these findings provide substantial understanding of C57BL/6J mice as an obesity model, and that an increased pool of hypertrophic ING adipocytes could contribute to aggravated insulin resistance.
Collapse
Affiliation(s)
- Claes Fryklund
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- *Correspondence: Claes Fryklund,
| | - Mathis Neuhaus
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Björn Morén
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Integrative Medical Biology, Umeå University, Umeå, Sweden
| | | | | | - Karin G. Stenkula
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
44
|
Gupta A, Balakrishnan B, Karki S, Slayton M, Jash S, Banerjee S, Grahn THM, Jambunathan S, Disney S, Hussein H, Kong D, Lowell BB, Natarajan P, Reddy UK, Gokce N, Sharma VM, Puri V. Human CIDEC transgene improves lipid metabolism and protects against high-fat diet-induced glucose intolerance in mice. J Biol Chem 2022; 298:102347. [PMID: 35963433 PMCID: PMC9472082 DOI: 10.1016/j.jbc.2022.102347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/08/2022] [Accepted: 07/20/2022] [Indexed: 11/12/2022] Open
Abstract
Cell death–inducing DNA fragmentation factor-like effector C (CIDEC) expression in adipose tissue positively correlates with insulin sensitivity in obese humans. Further, E186X, a single-nucleotide CIDEC variant is associated with lipodystrophy, hypertriglyceridemia, and insulin resistance. To establish the unknown mechanistic link between CIDEC and maintenance of systemic glucose homeostasis, we generated transgenic mouse models expressing CIDEC (Ad-CIDECtg) and CIDEC E186X variant (Ad-CIDECmut) transgene specifically in the adipose tissue. We found that Ad-CIDECtg but not Ad-CIDECmut mice were protected against high-fat diet-induced glucose intolerance. Furthermore, we revealed the role of CIDEC in lipid metabolism using transcriptomics and lipidomics. Serum triglycerides, cholesterol, and low-density lipoproteins were lower in high-fat diet-fed Ad-CIDECtg mice compared to their littermate controls. Mechanistically, we demonstrated that CIDEC regulates the enzymatic activity of adipose triglyceride lipase via interacting with its activator, CGI-58, to reduce free fatty acid release and lipotoxicity. In addition, we confirmed that CIDEC is indeed a vital regulator of lipolysis in adipose tissue of obese humans, and treatment with recombinant CIDEC decreased triglyceride breakdown in visceral human adipose tissue. Our study unravels a central pathway whereby adipocyte-specific CIDEC plays a pivotal role in regulating adipose lipid metabolism and whole-body glucose homeostasis. In summary, our findings identify human CIDEC as a potential ‘drug’ or a ‘druggable’ target to reverse obesity-induced lipotoxicity and glucose intolerance.
Collapse
Affiliation(s)
- Abhishek Gupta
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Bijinu Balakrishnan
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Shakun Karki
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Mark Slayton
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Sukanta Jash
- Alpert Medical school of Brown University, Brown University, RI, USA
| | - Sayani Banerjee
- Alpert Medical school of Brown University, Brown University, RI, USA
| | - Tan Hooi Min Grahn
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University Hospital, Lund, Sweden
| | | | - Sarah Disney
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Hebaallaha Hussein
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Dong Kong
- Division of Endocrinology, Department of Pediatrics, F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | | | - Umesh K Reddy
- Department of Biology, West Virginia State University, Institute, WV, USA
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Vishva M Sharma
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| | - Vishwajeet Puri
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| |
Collapse
|
45
|
Stenkula KG, Klemendz LE, Fryklund C, Wierup N, Alsalim W, Landin-Olsson M, Trinh L, Månsson S, Bennet L. Postprandial triglyceride levels rather than fat distribution may reflect early signs of disturbed fat metabolism in Iraqi immigrants. Lipids Health Dis 2022; 21:68. [PMID: 35927727 PMCID: PMC9351238 DOI: 10.1186/s12944-022-01679-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/19/2022] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Previous studies have shown that at a similar body mass index, Middle Eastern immigrants are more insulin resistant and at higher risk for type 2 diabetes (T2D) than native Europeans. Insulin resistance is strongly associated with disturbed fat metabolism and cardiovascular disease (CVD). However, fat metabolism is poorly investigated comparing Middle Eastern and European ethnicities. METHODS This observational study included 26 Iraqi and 16 Swedish-born men without T2D or clinical risk factors for CVD. An oral fat tolerance test (OFTT) was performed, where plasma triglycerides (p-TG) were measured for 6 h. mRNA expression and adipocyte size were measured in subcutaneous adipose tissue biopsies collected prior to OFTT, and magnetic resonance imaging was conducted to assess body fat distribution. RESULTS The median p-TG accumulation was higher and the clearance slower among Iraqis than Swedes. None of the groups reached their fasting p-TG (Iraqis 1.55 mmol/l; Swedes 0.95 mmol/l) after 6 h (Iraqis p-TG 3.10 mmol/l; Swedes p-TG 1.50 mmol/l). Adipocyte size, mRNA expression, and fat accumulation in the liver, muscle and abdomen were similar in both groups. CONCLUSION Postprandial p-TG levels rather than fat distribution may reflect early signs of disturbed fat metabolism in Iraqi immigrants without CVD risk factors.
Collapse
Affiliation(s)
- Karin G Stenkula
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden.
| | | | - Claes Fryklund
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Nils Wierup
- Lund University Diabetes Centre, Malmö, Sweden
| | - Wathik Alsalim
- Lund University, Skåne University Hospital Lund, Lund, Sweden
| | | | - Lena Trinh
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Sven Månsson
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Louise Bennet
- Lund University, Skåne University Hospital Lund, Lund, Sweden
| |
Collapse
|
46
|
White U, Beyl RA, Ravussin E. A higher proportion of small adipocytes is associated with increased visceral and ectopic lipid accumulation during weight gain in response to overfeeding in men. Int J Obes (Lond) 2022; 46:1560-1563. [PMID: 35599261 PMCID: PMC9348546 DOI: 10.1038/s41366-022-01150-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Adipose tissue (AT) expansion occurs by hypertrophy (increase in size) and hyperplasia (increase in number) of adipocytes. The AT expandability hypothesis postulates that impaired subcutaneous AT expansion leads to ectopic fat accretion, contributing to impaired metabolic health. The role of adipogenesis as a contributing factor is debatable. SUBJECTS/METHODS In the present analysis, we assess changes in adipocyte size distribution in relation to changes in ectopic fat accretion in response to 8-weeks of overfeeding in 22 men (28 ± 5.4 years; BMI 25.5 ± 2.3 kg/m2) who were fed 40% over their baseline energy requirements. RESULTS Participants gained 6.7 ± 2.1 kg. The percentage of small adipocytes (p = 0.03) and the peak diameter of large adipocytes (p = 0.01) increased after overfeeding. At baseline, the percentage of small adipocytes was positively correlated with % body fat (p = 0.03), SAT mass (p = 0.01), VAT mass (p = 0.02), VAT:TAT (p = 0.05), and IHL (p = 0.09; trend). The relative (percent) change in small adipocytes was positively associated with the increase in whole-body fat (p = 0.001), VAT mass (p = 0.0003), VAT:TAT (p = 0.01), and IHL (p = 0.007) in response to overfeeding. CONCLUSIONS These findings, surprisingly, indicate that during substantial weight gain, an increase in small adipocytes (suggesting hyperplastic expansion) is associated with impaired (not improved) metabolic health outcomes, specifically visceral and ectopic fat accumulation. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov Identifier- NCT01672632.
Collapse
Affiliation(s)
- Ursula White
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| | - Robbie A Beyl
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| |
Collapse
|
47
|
Cofano F, Perna GD, Bongiovanni D, Roscigno V, Baldassarre BM, Petrone S, Tartara F, Garbossa D, Bozzaro M. Obesity and Spine Surgery: A Qualitative Review About Outcomes and Complications. Is It Time for New Perspectives on Future Researches? Global Spine J 2022; 12:1214-1230. [PMID: 34128419 PMCID: PMC9210241 DOI: 10.1177/21925682211022313] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
STUDY DESIGN Literature review. OBJECTIVES An increasing number of obese patients requires operative care for degenerative spinal disorders. The aim of this review is to analyze the available evidence regarding the role of obesity on outcomes after spine surgery. Peri-operative complications and clinical results are evaluated for both cervical and lumbar surgery. Furthermore, the contribution of MIS techniques for lumbar surgery to play a role in reducing risks has been analyzed. METHODS Only articles published in English in the last 10 years were reviewed. Inclusion criteria of the references were based on the scope of this review, according to PRISMA guidelines. Moreover, only paper analyzing obesity-related complications in spine surgery have been selected and thoroughly reviewed. Each article was classified according to its rating of evidence using the Sacket Grading System. RESULTS A total number of 1636 articles were found, but only 130 of them were considered to be relevant after thorough evaluation and according to PRISMA checklist. The majority of the included papers were classified according to the Sacket Grading System as Level 2 (Retrospective Studies). CONCLUSION Evidence suggest that obese patients could benefit from spine surgery and outcomes be satisfactory. A higher rate of peri-operative complications is reported among obese patients, especially in posterior approaches. The use of MIS techniques plays a key role in order to reduce surgical risks. Further studies should evaluate the role of multidisciplinary counseling between spine surgeons, nutritionists and bariatric surgeons, in order to plan proper weight loss before elective spine surgery.
Collapse
Affiliation(s)
- Fabio Cofano
- Spine Surgery Unit, Humanitas Gradenigo Hospital, Turin, Italy,Department of Neuroscience “Rita Levi Montalcini,” Unit of Neurosurgery, University of Turin, Turin, Italy
| | - Giuseppe Di Perna
- Department of Neuroscience “Rita Levi Montalcini,” Unit of Neurosurgery, University of Turin, Turin, Italy
| | - Daria Bongiovanni
- Division of Endocrinology, Andrology and Metabolism, Humanitas Gradenigo Hospital, Turin, Italy
| | - Vittoria Roscigno
- Division of Endocrinology, Andrology and Metabolism, Humanitas Gradenigo Hospital, Turin, Italy
| | - Bianca Maria Baldassarre
- Department of Neuroscience “Rita Levi Montalcini,” Unit of Neurosurgery, University of Turin, Turin, Italy
| | - Salvatore Petrone
- Department of Neuroscience “Rita Levi Montalcini,” Unit of Neurosurgery, University of Turin, Turin, Italy,Salvatore Petrone, Department of Neuroscience “Rita Levi Montalcini,” Unit of Neurosurgery, University of Turin, Via Cherasco 15, Turin 10126, Italy.
| | - Fulvio Tartara
- Spine Surgery Unit, Humanitas Gradenigo Hospital, Turin, Italy
| | - Diego Garbossa
- Department of Neuroscience “Rita Levi Montalcini,” Unit of Neurosurgery, University of Turin, Turin, Italy
| | - Marco Bozzaro
- Spine Surgery Unit, Humanitas Gradenigo Hospital, Turin, Italy
| |
Collapse
|
48
|
Kotowska D, Neuhaus M, Heyman-Lindén L, Morén B, Li S, Kryvokhyzha D, Berger K, Stenkula KG. Short-term lingonberry feeding is associated with decreased insulin levels and altered adipose tissue function in high-fat diet fed C57BL/6J mice. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
49
|
Tiedemann K, Muthu ML, Reinhardt DP, Komarova SV. Male Marfan mice are predisposed to high fat diet induced obesity, diabetes, and fatty liver. Am J Physiol Cell Physiol 2022; 323:C354-C366. [PMID: 35759435 DOI: 10.1152/ajpcell.00062.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Gene mutations in the extracellular matrix protein fibrillin-1 cause connective tissue disorders including Marfan syndrome (MFS) with clinical symptoms in the cardiovascular, skeletal, and ocular systems. MFS patients also exhibit alterations in adipose tissues, which in some individuals leads to lipodystrophy, whereas in others to obesity. We have recently demonstrated that fibrillin-1 regulates adipose tissue homeostasis. Here, we examined how fibrillin-1 abnormality affects metabolic adaptation to different diets. We used two MFS mouse models: Hypomorph Fbn1mgR/mgR mice and Fbn1C1041G/+ mice with a fibrillin-1 missense mutation. When Fbn1mgR/mgR mice were fed with high fat diet (HFD) for 12 weeks, male mice were heavier than littermate controls (LC), whereas female mice gained less weight compared to LC. Female Fbn1C1041G/+ mice on a HFD for 24 weeks were similarly protected from weight gain. Male Fbn1C1041G/+ mice on HFD demonstrated higher insulin levels, insulin intolerance, circulating levels of cholesterol and high-density lipoproteins. Moreover, male HFD-fed Fbn1C1041G/+ mice showed a higher liver weight and a fatty liver phenotype, which was reduced to LC levels after orchiectomy. Phosphorylation of protein kinase-like endoplasmic reticulum kinase (PERK) as well as the expression of sterol regulatory element-binding protein 1 (Srebp1) in livers of HFD-fed male Fbn1C1041G/+ mice were elevated. In conclusion, the data demonstrate that male mice of both MFS models are susceptible to HFD-induced obesity and diabetes. Moreover, male Fbn1C1041G/+ mice develop a fatty liver phenotype, likely mediated by a baseline increased endoplasmic reticulum stress. In contrast, female MFS mice were protected from the consequence of HFD.
Collapse
Affiliation(s)
- Kerstin Tiedemann
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, Canada.,Shriners Hospital for Children - Canada, Montréal, Canada
| | - Muthu L Muthu
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Dieter P Reinhardt
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, Canada.,Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Svetlana V Komarova
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, Canada.,Shriners Hospital for Children - Canada, Montréal, Canada
| |
Collapse
|
50
|
Ruiz-Malagón AJ, Rodríguez-Sojo MJ, Hidalgo-García L, Molina-Tijeras JA, García F, Pischel I, Romero M, Duarte J, Diez-Echave P, Rodríguez-Cabezas ME, Rodríguez-Nogales A, Gálvez J. The Antioxidant Activity of Thymus serpyllum Extract Protects against the Inflammatory State and Modulates Gut Dysbiosis in Diet-Induced Obesity in Mice. Antioxidants (Basel) 2022; 11:antiox11061073. [PMID: 35739969 PMCID: PMC9219752 DOI: 10.3390/antiox11061073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 02/01/2023] Open
Abstract
Nowadays, there is an increasing interest in alternative therapies in the treatment of metabolic syndrome that combine efficacy and safety profiles. Therefore, this study aimed to evaluate the effect of an extract of Thymus serpyllum, containing rosmarinic acid, on high-fat diet (HFD)-induced obesity mice, highlighting the impact of its antioxidant activity on the inflammatory status and gut dysbiosis. The extract was administered daily (50, 100 and 150 mg/kg) in HFD-fed mice. The treatment reduced body weight gain, glucose and lipid metabolic profiles. Moreover, the extract ameliorated the inflammatory status, with the c-Jun N-terminal kinases (JUNK) pathway being involved, and showed a significant antioxidant effect by the reduction of radical scavenging activity and the mitigation of lipid peroxidation. Moreover, the extract was able to modulate the altered gut microbiota, restoring microbial richness and diversity, and augmenting the counts of short-chain fatty acid producing bacteria, which have been associated with the maintenance of gut permeability and weight regulation. In conclusion, the antioxidant activity of Thymus serpyllum extract displayed a positive impact on obesity and its metabolic alterations, also reducing systemic inflammation. These effects may be mediated by modulation of the gut microbiota.
Collapse
Affiliation(s)
- Antonio Jesús Ruiz-Malagón
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (A.J.R.-M.); (M.J.R.-S.); (L.H.-G.); (J.A.M.-T.); (M.R.); (J.D.); (A.R.-N.); (J.G.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
| | - María Jesús Rodríguez-Sojo
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (A.J.R.-M.); (M.J.R.-S.); (L.H.-G.); (J.A.M.-T.); (M.R.); (J.D.); (A.R.-N.); (J.G.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
| | - Laura Hidalgo-García
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (A.J.R.-M.); (M.J.R.-S.); (L.H.-G.); (J.A.M.-T.); (M.R.); (J.D.); (A.R.-N.); (J.G.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
| | - José Alberto Molina-Tijeras
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (A.J.R.-M.); (M.J.R.-S.); (L.H.-G.); (J.A.M.-T.); (M.R.); (J.D.); (A.R.-N.); (J.G.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
| | - Federico García
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Servicio Microbiología, Hospital Universitario Clínico San Cecilio, 18100 Granada, Spain
| | - Ivo Pischel
- Centre for Pharmacognosy and Phytotherapy, UCL School of Pharmacy, University of London, London WC1N 1AX, UK;
| | - Miguel Romero
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (A.J.R.-M.); (M.J.R.-S.); (L.H.-G.); (J.A.M.-T.); (M.R.); (J.D.); (A.R.-N.); (J.G.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Juan Duarte
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (A.J.R.-M.); (M.J.R.-S.); (L.H.-G.); (J.A.M.-T.); (M.R.); (J.D.); (A.R.-N.); (J.G.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Patricia Diez-Echave
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (A.J.R.-M.); (M.J.R.-S.); (L.H.-G.); (J.A.M.-T.); (M.R.); (J.D.); (A.R.-N.); (J.G.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Correspondence: (P.D.-E.); (M.E.R.-C.); Tel.: +34-958241519 (M.E.R.-C.)
| | - María Elena Rodríguez-Cabezas
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (A.J.R.-M.); (M.J.R.-S.); (L.H.-G.); (J.A.M.-T.); (M.R.); (J.D.); (A.R.-N.); (J.G.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Correspondence: (P.D.-E.); (M.E.R.-C.); Tel.: +34-958241519 (M.E.R.-C.)
| | - Alba Rodríguez-Nogales
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (A.J.R.-M.); (M.J.R.-S.); (L.H.-G.); (J.A.M.-T.); (M.R.); (J.D.); (A.R.-N.); (J.G.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
| | - Julio Gálvez
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (A.J.R.-M.); (M.J.R.-S.); (L.H.-G.); (J.A.M.-T.); (M.R.); (J.D.); (A.R.-N.); (J.G.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|