151
|
Illesca P, Valenzuela R, Espinosa A, Echeverría F, Soto-Alarcón S, Ortiz M, Campos C, Vargas R, Videla LA. The metabolic dysfunction of white adipose tissue induced in mice by a high-fat diet is abrogated by co-administration of docosahexaenoic acid and hydroxytyrosol. Food Funct 2021; 11:9086-9102. [PMID: 33026007 DOI: 10.1039/d0fo01790f] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Nutritional interventions are promising tools for the prevention of obesity. The n-3 long-chain polyunsaturated fatty acid (n-3 LCPUFA) docosahexaenoic acid (DHA) modulates immune and metabolic responses while the antioxidant hydroxytyrosol (HT) prevents oxidative stress (OS) in white adipose tissue (WAT). OBJECTIVE The DHA plus HT combined protocol prevents WAT alterations induced by a high-fat diet in mice. Main related mechanisms. METHODS Male C57BL/6J mice were fed a control diet (CD; 10% fat, 20% protein, and 70% carbohydrates) or a high fat diet (HFD) (60% fat, 20% protein, and 20% carbohydrates) for 12 weeks, without and with supplementation of DHA (50 mg kg-1 day-1), HT (5 mg kg-1 day-1) or both. Measurements of WAT metabolism include morphological parameters, DHA content in phospholipids (gas chromatography), lipogenesis, OS and inflammation markers, mitochondrial activity and gene expression of transcription factors SREBP-1c, PPAR-γ, NF-κB (p65) and Nrf2 (quantitative polymerase chain reaction and enzyme-linked immunosorbent assay). RESULTS The combined DHA and HT intervention attenuated obesity development, suppressing the HFD-induced inflammatory and lipogenic signals, increasing antioxidant defenses, and maintaining the phospholipid LCPUFA n-3 content and mitochondrial function in WAT. At the systemic level, the combined intervention also improved the regulation of glucose and adipokine homeostasis. CONCLUSION The combined DHA and HT protocol appears to be an important nutritional strategy for the treatment of metabolic diseases, with abrogation of obesity-driven metabolic inflammation and recovery of a small-healthy adipocyte phenotype.
Collapse
Affiliation(s)
- Paola Illesca
- Laboratory of Studies of Metabolic Diseases Related to Nutrition, Faculty of Biochemistry, University of Litoral, Santa Fe, Argentina
| | - Rodrigo Valenzuela
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Alejandra Espinosa
- Medical Technology Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Francisca Echeverría
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Sandra Soto-Alarcón
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Macarena Ortiz
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Cristian Campos
- Medical Technology Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Romina Vargas
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
152
|
PEGylated AdipoRon derivatives improve glucose and lipid metabolism under insulinopenic and high-fat diet conditions. J Lipid Res 2021; 62:100095. [PMID: 34214600 PMCID: PMC8327158 DOI: 10.1016/j.jlr.2021.100095] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/17/2021] [Indexed: 12/19/2022] Open
Abstract
The pleiotropic actions of adiponectin in improving cell survival and metabolism have motivated the development of small-molecule therapeutic agents for treating diabetes and lipotoxicity. AdipoRon is a synthetic agonist of the adiponectin receptors, yet is limited by its poor solubility and bioavailability. In this work, we expand on the protective effects of AdipoRon in pancreatic β-cells and examine how structural modifications could affect the activity, pharmacokinetics, and bioavailability of this small molecule. We describe a series of AdipoRon analogs containing amphiphilic ethylene glycol (PEG) chains. Among these, AdipoRonPEG5 induced pleiotropic effects in mice under insulinopenic and high-fat diet (HFD) conditions. While both AdipoRon and AdipoRonPEG5 substantially attenuate palmitate-induced lipotoxicity in INS-1 cells, only AdipoRonPEG5 treatment is accompanied by a significant reduction in cytotoxic ceramides. In vivo, AdipoRonPEG5 can substantially reduce pancreatic, hepatic, and serum ceramide species, with a concomitant increase in the corresponding sphingoid bases and improves insulin sensitivity of mice under HFD feeding conditions. Furthermore, hyperglycemia in streptozotocin (STZ)-induced insulinopenic adiponectin-null mice is also attenuated upon AdipoRonPEG5 treatment. Our results suggest that AdipoRonPEG5 is more effective in reducing ceramides and dihydroceramides in the liver of HFD-fed mice than AdipoRon, consistent with its potent activity in activating ceramidase in vitro in INS-1 cells. Additionally, these results indicate that the beneficial effects of AdipoRonPEG5 can be partially attributed to improved pharmacokinetics as compared with AdipoRon, thus suggesting that further derivatization may improve affinity and tissue-specific targeting.
Collapse
|
153
|
Blaszczak AM, Jalilvand A, Hsueh WA. Adipocytes, Innate Immunity and Obesity: A Mini-Review. Front Immunol 2021; 12:650768. [PMID: 34248937 PMCID: PMC8264354 DOI: 10.3389/fimmu.2021.650768] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
The role of adipose tissue (AT) inflammation in obesity and its multiple related-complications is a rapidly expanding area of scientific interest. Within the last 30 years, the role of the adipocyte as an endocrine and immunologic cell has been progressively established. Like the macrophage, the adipocyte is capable of linking the innate and adaptive immune system through the secretion of adipokines and cytokines; exosome release of lipids, hormones, and microRNAs; and contact interaction with other immune cells. Key innate immune cells in AT include adipocytes, macrophages, neutrophils, and innate lymphoid cells type 2 (ILC2s). The role of the innate immune system in promoting adipose tissue inflammation in obesity will be highlighted in this review. T cells and B cells also play important roles in contributing to AT inflammation and are discussed in this series in the chapter on adaptive immunity.
Collapse
Affiliation(s)
- Alecia M Blaszczak
- Hsueh Laboratory, The Ohio State University Wexner Medical Center, Diabetes and Metabolism Research Center, Columbus, OH, United States
| | - Anahita Jalilvand
- Hsueh Laboratory, The Ohio State University Wexner Medical Center, Diabetes and Metabolism Research Center, Columbus, OH, United States
| | - Willa A Hsueh
- Hsueh Laboratory, The Ohio State University Wexner Medical Center, Diabetes and Metabolism Research Center, Columbus, OH, United States
| |
Collapse
|
154
|
Wiggins KB, Smith MA, Schultz-Cherry S. The Nature of Immune Responses to Influenza Vaccination in High-Risk Populations. Viruses 2021; 13:v13061109. [PMID: 34207924 PMCID: PMC8228336 DOI: 10.3390/v13061109] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
The current pandemic has brought a renewed appreciation for the critical importance of vaccines for the promotion of both individual and public health. Influenza vaccines have been our primary tool for infection control to prevent seasonal epidemics and pandemics such as the 2009 H1N1 influenza A virus pandemic. Certain high-risk populations, including the elderly, people with obesity, and individuals with comorbidities such as type 2 diabetes mellitus, are more susceptible to increased disease severity and decreased vaccine efficacy. High-risk populations have unique microenvironments and immune responses that contribute to increased vulnerability for influenza infections. This review focuses on these differences as we investigate the variations in immune responses to influenza vaccination. In order to develop better influenza vaccines, it is critical to understand how to improve responses in our ever-growing high-risk populations.
Collapse
|
155
|
PAR2 Deficiency Induces Mitochondrial ROS Generation and Dysfunctions, Leading to the Inhibition of Adipocyte Differentiation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6683033. [PMID: 34211632 PMCID: PMC8205587 DOI: 10.1155/2021/6683033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/29/2021] [Indexed: 11/18/2022]
Abstract
Protease-activated receptor 2 (PAR2) is a member of G-protein-coupled receptors and affects ligand-modulated calcium signaling. Although PAR2 signaling promotes obesity and adipose tissue inflammation in high fat- (HF-) fed conditions, its role in adipocyte differentiation under nonobesogenic conditions needs to be elucidated. Here, we used several tissues and primary-cultured adipocytes of mice lacking PAR2 to study its role in the development of adipose tissues. C57BL/6J mice with PAR2 deficiency exhibited a mild lipodystrophy-like phenotype in a chow diet-fed condition. When adipocyte differentiation was examined using primary-cultured preadipocytes, PAR2 deficiency led to a notable decrease in adipocyte differentiation and related protein expression, and PAR2 agonist treatment elevated adipocyte differentiation. Regarding the mechanism, PAR2-deficient preadipocytes exhibited impaired mitochondrial energy consumption. Further studies indicated that calcium-related signaling pathways for mitochondrial biogenesis are disrupted in the adipose tissues of PAR2-deficient mice and PAR2-deficient preadipocytes. Also, a PAR2 antagonist elevated mitochondrial reactive oxygen species and reduced the MitoTracker fluorescent signal in preadipocytes. Our studies revealed that PAR2 is important for the development of adipose tissue under basal conditions through the regulation of mitochondrial biogenesis and adipocyte differentiation.
Collapse
|
156
|
Méndez-Lara KA, Rodríguez-Millán E, Sebastián D, Blanco-Soto R, Camacho M, Nan MN, Diarte-Añazco EMG, Mato E, Lope-Piedrafita S, Roglans N, Laguna JC, Alonso N, Mauricio D, Zorzano A, Villarroya F, Villena JA, Blanco-Vaca F, Julve J. Nicotinamide Protects Against Diet-Induced Body Weight Gain, Increases Energy Expenditure, and Induces White Adipose Tissue Beiging. Mol Nutr Food Res 2021; 65:e2100111. [PMID: 33870623 DOI: 10.1002/mnfr.202100111] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/31/2021] [Indexed: 12/30/2022]
Abstract
SCOPE Interventions that boost NAD+ availability are of potential therapeutic interest for obesity treatment. The potential of nicotinamide (NAM), the amide form of vitamin B3 and a physiological precursor of nicotinamide adenine dinucleotide (NAD)+ , in preventing weight gain has not previously been studied in vivo. Other NAD+ precursors have been shown to decrease weight gain; however, their impact on adipose tissue is not addressed. METHODS AND RESULTS Two doses of NAM (high dose: 1% and low dose: 0.25%) are given by drinking water to C57BL/6J male mice, starting at the same time as the high-fat diet feeding. NAM supplementation protects against diet-induced obesity by augmenting global body energy expenditure in C57BL/6J male mice. The manipulation markedly alters adipose morphology and metabolism, particularly in inguinal (i) white adipose tissue (iWAT). An increased number of brown and beige adipocyte clusters, protein abundance of uncoupling protein 1 (UCP1), mitochondrial activity, adipose NAD+ , and phosphorylated AMP-activated protein kinase (P-AMPK) levels are observed in the iWAT of treated mice. Notably, a significant improvement in hepatic steatosis, inflammation, and glucose tolerance is also observed in NAM high-dose treated mice. CONCLUSION NAM influences whole-body energy expenditure by driving changes in the adipose phenotype. Thus, NAM is an attractive potential treatment for preventing obesity and associated complications.
Collapse
Affiliation(s)
- Karen Alejandra Méndez-Lara
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, 08041, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona (UAB), Barcelona, 08193, Spain
| | - Elisabeth Rodríguez-Millán
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, 08041, Spain
| | - David Sebastián
- Departament de Bioquímica i Biomedicina, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, 08028, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, 28028, Spain
| | - Rosi Blanco-Soto
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, 28028, Spain
| | - Mercedes Camacho
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, 08041, Spain
| | - Madalina N Nan
- Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, 08041, Spain
| | - Elena M G Diarte-Añazco
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, 08041, Spain
| | - Eugènia Mato
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, 28028, Spain
| | - Silvia Lope-Piedrafita
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, 28028, Spain
- Servei de Ressonància Magnètica Nuclear, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, 08193, Spain
| | - Núria Roglans
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, 08028, Spain
| | - Juan Carlos Laguna
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, 08028, Spain
| | - Núria Alonso
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, 28028, Spain
- Servei d'Endocrinologia, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, 08916, Spain
| | - Dídac Mauricio
- Departament de Bioquímica i Biomedicina, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, 08028, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, 28028, Spain
- Servei de Endocrinologia i Nutrició, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, 08041, Spain
| | - Antonio Zorzano
- Departament de Bioquímica i Biomedicina, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, 08028, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, 28028, Spain
| | - Francesc Villarroya
- Departament de Bioquímica i Biomedicina, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, 08028, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, CIBEROBN, Madrid, 28028, Spain
| | - Josep A Villena
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, 28028, Spain
- Laboratori de Metabolisme i Obesitat, Unitat de Diabetis i Metabolisme, Institut de Recerca del Vall d'Hebron, Barcelona, 08035, Spain
| | - Francisco Blanco-Vaca
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona (UAB), Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, 28028, Spain
- Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, 08041, Spain
| | - Josep Julve
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau i Institut d'Investigació Biomèdica de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, 08041, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona (UAB), Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, 28028, Spain
| |
Collapse
|
157
|
Ryu J, Hadley JT, Li Z, Dong F, Xu H, Xin X, Zhang Y, Chen C, Li S, Guo X, Zhao JL, Leach RJ, Abdul-Ghani MA, DeFronzo RA, Kamat A, Liu F, Dong LQ. Adiponectin Alleviates Diet-Induced Inflammation in the Liver by Suppressing MCP-1 Expression and Macrophage Infiltration. Diabetes 2021; 70:1303-1316. [PMID: 34162682 PMCID: PMC8275886 DOI: 10.2337/db20-1073] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 03/10/2021] [Indexed: 01/21/2023]
Abstract
Adiponectin is an adipokine that exerts insulin-sensitizing and anti-inflammatory roles in insulin target tissues including liver. While the insulin-sensitizing function of adiponectin has been extensively investigated, the precise mechanism by which adiponectin alleviates diet-induced hepatic inflammation remains elusive. Here, we report that hepatocyte-specific knockout (KO) of the adaptor protein APPL2 enhanced adiponectin sensitivity and prevented mice from developing high-fat diet-induced inflammation, insulin resistance, and glucose intolerance, although it caused fatty liver. The improved anti-inflammatory and insulin-sensitizing effects in the APPL2 hepatocyte-specific KO mice were largely reversed by knocking out adiponectin. Mechanistically, hepatocyte APPL2 deficiency enhances adiponectin signaling in the liver, which blocks TNF-α-stimulated MCP-1 expression via inhibiting the mTORC1 signaling pathway, leading to reduced macrophage infiltration and thus reduced inflammation in the liver. With results taken together, our study uncovers a mechanism underlying the anti-inflammatory role of adiponectin in the liver and reveals the hepatic APPL2-mTORC1-MCP-1 axis as a potential target for treating overnutrition-induced inflammation in the liver.
Collapse
Affiliation(s)
- Jiyoon Ryu
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Jason T Hadley
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Zhi Li
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Feng Dong
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Huan Xu
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Xiaoban Xin
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Ye Zhang
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Cang Chen
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Senlin Li
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Xiaoning Guo
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Jared L Zhao
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Robin J Leach
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Muhammad A Abdul-Ghani
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Ralph A DeFronzo
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Amrita Kamat
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
- Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX
| | - Feng Liu
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Lily Q Dong
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX
| |
Collapse
|
158
|
Raza S, Tewari A, Rajak S, Sinha RA. Vitamins and non-alcoholic fatty liver disease: A Molecular Insight ⋆. LIVER RESEARCH (BEIJING, CHINA) 2021; 5:62-71. [PMID: 34221537 PMCID: PMC7611112 DOI: 10.1016/j.livres.2021.03.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/23/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023]
Abstract
The incidence of non-alcoholic fatty liver disease (NAFLD) is rising rapidly across the globe. NAFLD pathogenesis is largely driven by an imbalance in hepatic energy metabolism and at present, there is no approved drug for its treatment. The liver plays a crucial role in micronutrient metabolism and deregulation of this micronutrient metabolism may contribute to the pathogenesis of NAFLD. Vitamins regulate several enzymatic processes in the liver, and derangement in vitamin metabolism is believed to play a critical role in NAFLD progression. The anti-oxidant activities of vitamin C and E have been attributed to mitigate hepatocyte injury, and alterations in the serum levels of vitamin D, vitamin B12 and folate have shown a strong correlation with NAFLD severity. This review aims to highlight the role of these vitamins, which represent promising therapeutic targets for the management of NAFLD.
Collapse
Affiliation(s)
- Sana Raza
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Archana Tewari
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Sangam Rajak
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Rohit A. Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
159
|
Álvarez-Artime A, García-Soler B, Sainz RM, Mayo JC. Emerging Roles for Browning of White Adipose Tissue in Prostate Cancer Malignant Behaviour. Int J Mol Sci 2021; 22:5560. [PMID: 34074045 PMCID: PMC8197327 DOI: 10.3390/ijms22115560] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
In addition to its well-known role as an energy repository, adipose tissue is one of the largest endocrine organs in the organism due to its ability to synthesize and release different bioactive molecules. Two main types of adipose tissue have been described, namely white adipose tissue (WAT) with a classical energy storage function, and brown adipose tissue (BAT) with thermogenic activity. The prostate, an exocrine gland present in the reproductive system of most mammals, is surrounded by periprostatic adipose tissue (PPAT) that contributes to maintaining glandular homeostasis in conjunction with other cell types of the microenvironment. In pathological conditions such as the development and progression of prostate cancer, adipose tissue plays a key role through paracrine and endocrine signaling. In this context, the role of WAT has been thoroughly studied. However, the influence of BAT on prostate tumor development and progression is unclear and has received much less attention. This review tries to bring an update on the role of different factors released by WAT which may participate in the initiation, progression and metastasis, as well as to compile the available information on BAT to discuss and open a new field of knowledge about the possible protective role of BAT in prostate cancer.
Collapse
Affiliation(s)
- Alejandro Álvarez-Artime
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario s/n, 33011 Oviedo, Spain
| | - Belén García-Soler
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
| | - Rosa María Sainz
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario s/n, 33011 Oviedo, Spain
| | - Juan Carlos Mayo
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario s/n, 33011 Oviedo, Spain
| |
Collapse
|
160
|
Ding Y, Zhang L, Yao X, Zhang H, He X, Fan Z, Song Z. Honokiol Alleviates High-Fat Diet-Induced Obesity of Mice by Inhibiting Adipogenesis and Promoting White Adipose Tissue Browning. Animals (Basel) 2021; 11:1493. [PMID: 34064117 PMCID: PMC8224378 DOI: 10.3390/ani11061493] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 12/14/2022] Open
Abstract
Honokiol (HON) is one of the main biological active components of the traditional Chinese medicine Magnolia officinalis and has many health benefits. The aim of this study was to investigate whether HON could alleviate obesity in mice by inhibiting adipogenesis and promoting the browning of white adipose tissue (WAT). C57BL/6 mice were divided into five groups and fed with a normal diet (ND), high-fat diet (HFD), or HFD supplemented with 200 (H200), 400 (H400), or 800 (H800) mg/kg BW HON for 8 weeks. The results showed that the mice fed HFD plus HON had lower body fat ratios (BFRs) and smaller adipocyte diameters in the epididymal WAT compared with those of the HFD group. With a proteomics analysis, the HON group upregulated 30 proteins and downregulated 98 proteins in the epididymal WAT of mice, and the steroid O-acyltransferase 1 (SOAT1) was screened as a key protein. The HON supplement prevented HFD-induced adipogenesis by reduced the mRNA and protein expression of SOAT1 and CCAAT/enhancer-binding protein-α (C/EBPα), suggesting that SOAT1 might play an important role in regulating adipogenesis. Moreover, HON treatment increased the expression of proteins related to the classical pathways of energy and lipid metabolism, such as AMP-activated kinase (AMPK) and acetyl-CoA carboxylase (ACC), and promoted the browning of epididymal WAT by upregulation of the protein expression of uncoupling protein 1 (UCP1) in the HFD mice. In conclusion, these results suggest that HON supplements could prevent increases in body fat for HFD mice by suppressing adipogenesis and promoting WAT browning.
Collapse
Affiliation(s)
- Yanan Ding
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (Y.D.); (L.Z.); (X.Y.); (H.Z.); (X.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha 410128, China
| | - Longlin Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (Y.D.); (L.Z.); (X.Y.); (H.Z.); (X.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha 410128, China
| | - Xiaofeng Yao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (Y.D.); (L.Z.); (X.Y.); (H.Z.); (X.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha 410128, China
| | - Haihan Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (Y.D.); (L.Z.); (X.Y.); (H.Z.); (X.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha 410128, China
| | - Xi He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (Y.D.); (L.Z.); (X.Y.); (H.Z.); (X.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha 410128, China
| | - Zhiyong Fan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (Y.D.); (L.Z.); (X.Y.); (H.Z.); (X.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha 410128, China
| | - Zehe Song
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (Y.D.); (L.Z.); (X.Y.); (H.Z.); (X.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
161
|
Dragoljevic D, Veiga CB, Michell DL, Shihata WA, Al-Sharea A, Head GA, Murphy AJ, Kraakman MJ, Lee MKS. A spontaneously hypertensive diet-induced atherosclerosis-prone mouse model of metabolic syndrome. Biomed Pharmacother 2021; 139:111668. [PMID: 34243630 DOI: 10.1016/j.biopha.2021.111668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Metabolic Syndrome (MetS) is a complex and multifactorial condition often characterised by obesity, hypertension, hyperlipidaemia, insulin resistance, glucose intolerance and fasting hyperglycaemia. Collectively, MetS can increase the risk of atherosclerotic-cardiovascular disease, which is the leading cause of death worldwide. However, no animal model currently exists to study MetS in the context of atherosclerosis. In this study we developed a pre-clinical mouse model that recapitulates the spectrum of MetS features while developing atherosclerosis. When BPHx mice were placed on a western type diet for 16 weeks, all the classical characteristics of MetS were observed. Comprehensive metabolic analyses and atherosclerotic imaging revealed BPHx mice to be obese and hypertensive, with elevated total plasma cholesterol and triglyceride levels, that accelerated atherosclerosis. Altogether, we demonstrate that the BPHx mouse has all the major components of MetS, and accelerates the development of atherosclerosis.
Collapse
Affiliation(s)
- Dragana Dragoljevic
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; The University of Melbourne, Melbourne, VIC, Australia; Monash University, Melbourne, VIC, Australia
| | - Camilla Bertuzzo Veiga
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; The University of Melbourne, Melbourne, VIC, Australia
| | | | - Waled A Shihata
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Annas Al-Sharea
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Geoffrey A Head
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; The University of Melbourne, Melbourne, VIC, Australia; Monash University, Melbourne, VIC, Australia
| | | | - Man K S Lee
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; The University of Melbourne, Melbourne, VIC, Australia; Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
162
|
Ren J, Wu NN, Wang S, Sowers JR, Zhang Y. Obesity cardiomyopathy: evidence, mechanisms, and therapeutic implications. Physiol Rev 2021; 101:1745-1807. [PMID: 33949876 PMCID: PMC8422427 DOI: 10.1152/physrev.00030.2020] [Citation(s) in RCA: 215] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The prevalence of heart failure is on the rise and imposes a major health threat, in part, due to the rapidly increased prevalence of overweight and obesity. To this point, epidemiological, clinical, and experimental evidence supports the existence of a unique disease entity termed “obesity cardiomyopathy,” which develops independent of hypertension, coronary heart disease, and other heart diseases. Our contemporary review evaluates the evidence for this pathological condition, examines putative responsible mechanisms, and discusses therapeutic options for this disorder. Clinical findings have consolidated the presence of left ventricular dysfunction in obesity. Experimental investigations have uncovered pathophysiological changes in myocardial structure and function in genetically predisposed and diet-induced obesity. Indeed, contemporary evidence consolidates a wide array of cellular and molecular mechanisms underlying the etiology of obesity cardiomyopathy including adipose tissue dysfunction, systemic inflammation, metabolic disturbances (insulin resistance, abnormal glucose transport, spillover of free fatty acids, lipotoxicity, and amino acid derangement), altered intracellular especially mitochondrial Ca2+ homeostasis, oxidative stress, autophagy/mitophagy defect, myocardial fibrosis, dampened coronary flow reserve, coronary microvascular disease (microangiopathy), and endothelial impairment. Given the important role of obesity in the increased risk of heart failure, especially that with preserved systolic function and the recent rises in COVID-19-associated cardiovascular mortality, this review should provide compelling evidence for the presence of obesity cardiomyopathy, independent of various comorbid conditions, underlying mechanisms, and offer new insights into potential therapeutic approaches (pharmacological and lifestyle modification) for the clinical management of obesity cardiomyopathy.
Collapse
Affiliation(s)
- Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China.,Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Ne N Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Shuyi Wang
- School of Medicine, Shanghai University, Shanghai, China.,University of Wyoming College of Health Sciences, Laramie, Wyoming
| | - James R Sowers
- Dalton Cardiovascular Research Center, Diabetes and Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
163
|
Porro S, Genchi VA, Cignarelli A, Natalicchio A, Laviola L, Giorgino F, Perrini S. Dysmetabolic adipose tissue in obesity: morphological and functional characteristics of adipose stem cells and mature adipocytes in healthy and unhealthy obese subjects. J Endocrinol Invest 2021; 44:921-941. [PMID: 33145726 DOI: 10.1007/s40618-020-01446-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
The way by which subcutaneous adipose tissue (SAT) expands and undergoes remodeling by storing excess lipids through expansion of adipocytes (hypertrophy) or recruitment of new precursor cells (hyperplasia) impacts the risk of developing cardiometabolic and respiratory diseases. In unhealthy obese subjects, insulin resistance, type 2 diabetes, hypertension, and obstructive sleep apnoea are typically associated with pathologic SAT remodeling characterized by adipocyte hypertrophy, as well as chronic inflammation, hypoxia, increased visceral adipose tissue (VAT), and fatty liver. In contrast, metabolically healthy obese individuals are generally associated with SAT development characterized by the presence of smaller and numerous mature adipocytes, and a lower degree of VAT inflammation and ectopic fat accumulation. The remodeling of SAT and VAT is under genetic regulation and influenced by inherent depot-specific differences of adipose tissue-derived stem cells (ASCs). ASCs have multiple functions such as cell renewal, adipogenic capacity, and angiogenic properties, and secrete a variety of bioactive molecules involved in vascular and extracellular matrix remodeling. Understanding the mechanisms regulating the proliferative and adipogenic capacity of ASCs from SAT and VAT in response to excess calorie intake has become a focus of interest over recent decades. Here, we summarize current knowledge about the biological mechanisms able to foster or impair the recruitment and adipogenic differentiation of ASCs during SAT and VAT development, which regulate body fat distribution and favorable or unfavorable metabolic responses.
Collapse
Affiliation(s)
- S Porro
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - V A Genchi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Cignarelli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Natalicchio
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - L Laviola
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - F Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy.
| | - S Perrini
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
164
|
Winn NC, Cottam MA, Wasserman DH, Hasty AH. Exercise and Adipose Tissue Immunity: Outrunning Inflammation. Obesity (Silver Spring) 2021; 29:790-801. [PMID: 33899336 DOI: 10.1002/oby.23147] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/02/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023]
Abstract
Chronic inflammation is considered a precipitating factor and possibly an underlying cause of many noncommunicable diseases, including cardiovascular disease, metabolic diseases, and some cancers. Obesity, which manifests in more than 650 million people worldwide, is the most common chronic inflammatory condition, with visceral adiposity thought to be the major inflammatory hub that links obesity and chronic disease. Adipose tissue (AT) inflammation is triggered or heightened in large part by (1) accelerated immune cell recruitment, (2) reshaping of the AT stromal-immuno landscape (e.g., immune cells, endothelial cells, fibroblasts, adipocyte progenitors), and (3) perturbed AT immune cell function. Exercise, along with diet management, is a cornerstone in promoting weight loss and preventing weight regain. This review focuses on evidence that increased physical activity reduces AT inflammation caused by hypercaloric diets or genetic obesity. The precise cell types and mechanisms responsible for the therapeutic effects of exercise on AT inflammation remain poorly understood. This review summarizes what is known about obesity-induced AT inflammation and immunomodulation and highlights mechanisms by which aerobic exercise combats inflammation by remodeling the AT immune landscape. Furthermore, key areas are highlighted that require future exploration and novel discoveries into the burgeoning field of how the biology of exercise affects AT immunity.
Collapse
Affiliation(s)
- Nathan C Winn
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Matthew A Cottam
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, Tennessee, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
165
|
Jacquelot N, Belz GT, Seillet C. Neuroimmune Interactions and Rhythmic Regulation of Innate Lymphoid Cells. Front Neurosci 2021; 15:657081. [PMID: 33994930 PMCID: PMC8116950 DOI: 10.3389/fnins.2021.657081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/29/2021] [Indexed: 01/21/2023] Open
Abstract
The Earth’s rotation around its axis, is one of the parameters that never changed since life emerged. Therefore, most of the organisms from the cyanobacteria to humans have conserved natural oscillations to regulate their physiology. These daily oscillations define the circadian rhythms that set the biological clock for almost all physiological processes of an organism. They allow the organisms to anticipate and respond behaviorally and physiologically to changes imposed by the day/night cycle. As other physiological systems, the immune system is also regulated by circadian rhythms and while diurnal variation in host immune responses to lethal infection have been observed for many decades, the underlying mechanisms that affect immune function and health have only just started to emerge. These oscillations are generated by the central clock in our brain, but neuroendocrine signals allow the synchronization of the clocks in peripheral tissues. In this review, we discuss how the neuroimmune interactions create a rhythmic activity of the innate lymphoid cells. We highlight how the disruption of these rhythmic regulations of immune cells can disturb homeostasis and lead to the development of chronic inflammation in murine models.
Collapse
Affiliation(s)
- Nicolas Jacquelot
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Cyril Seillet
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
166
|
Li N, Zhao S, Zhang Z, Zhu Y, Gliniak CM, Vishvanath L, An YA, Wang MY, Deng Y, Zhu Q, Shan B, Sherwood A, Onodera T, Oz OK, Gordillo R, Gupta RK, Liu M, Horvath TL, Dixit VD, Scherer PE. Adiponectin preserves metabolic fitness during aging. eLife 2021; 10:65108. [PMID: 33904399 PMCID: PMC8099426 DOI: 10.7554/elife.65108] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
Adiponectin is essential for the regulation of tissue substrate utilization and systemic insulin sensitivity. Clinical studies have suggested a positive association of circulating adiponectin with healthspan and lifespan. However, the direct effects of adiponectin on promoting healthspan and lifespan remain unexplored. Here, we are using an adiponectin null mouse and a transgenic adiponectin overexpression model. We directly assessed the effects of circulating adiponectin on the aging process and found that adiponectin null mice display exacerbated age-related glucose and lipid metabolism disorders. Moreover, adiponectin null mice have a significantly shortened lifespan on both chow and high-fat diet. In contrast, a transgenic mouse model with elevated circulating adiponectin levels has a dramatically improved systemic insulin sensitivity, reduced age-related tissue inflammation and fibrosis, and a prolonged healthspan and median lifespan. These results support a role of adiponectin as an essential regulator for healthspan and lifespan.
Collapse
Affiliation(s)
- Na Li
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States.,Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Zhuzhen Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Yi Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Christy M Gliniak
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Lavanya Vishvanath
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Yu A An
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - May-Yun Wang
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Yingfeng Deng
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Qingzhang Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Bo Shan
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Amber Sherwood
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Toshiharu Onodera
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Orhan K Oz
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Tamas L Horvath
- Department of Comparative Medicine and Immunobiology, Yale School of Medicine, New Haven, United States
| | - Vishwa Deep Dixit
- Department of Comparative Medicine and Immunobiology, Yale School of Medicine, New Haven, United States.,Yale Center for Research on Aging, Yale School of Medicine, New Haven, United States
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States.,Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
167
|
Ribnicky D, Kim SB, Poulev A, Wang Y, Boudreau A, Raskin I, Bisson J, Ray GJ, Chen SN, Richard A, Stephens JM, Pauli GF. Prenylated Coumaric Acids from Artemisia scoparia Beneficially Modulate Adipogenesis. JOURNAL OF NATURAL PRODUCTS 2021; 84:1078-1086. [PMID: 33830759 PMCID: PMC8132292 DOI: 10.1021/acs.jnatprod.0c01149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Two new diprenylated coumaric acid isomers (1a and 1b) and two known congeners, capillartemisin A (2) and B (3), were isolated from Artemisia scoparia as bioactive markers using bioactivity-guided HPLC fractionation. Their structures were determined by spectroscopic means, including 1D and 2D NMR methods and LC-MS, with their purity assessed by 1D 1H pure shift qNMR spectroscopic analysis. The bioactivity of compounds was evaluated by enhanced accumulation of lipids, as measured using Oil Red O staining, and by increased expression of several adipocyte marker genes, including adiponectin in 3T3-L1 adipocytes relative to untreated negative controls. Compared to the plant's 80% EtOH extract, these purified compounds showed significant but still weaker inhibition of TNFα-induced lipolysis in 3T3-L1 adipocytes. This suggests that additional bioactive substances are responsible for the multiple metabolically favorable effects on adipocytes observed with Artemisia scoparia extract.
Collapse
Affiliation(s)
- David Ribnicky
- Corresponding Authors Tel: +1 312 355 1949 Fax: +1 312 413 5894 (David Ribnicky): (Guido Pauli):
| | - Seon Beom Kim
- Corresponding Authors Tel: +1 312 355 1949 Fax: +1 312 413 5894 (David Ribnicky): (Guido Pauli):
| | - Alexander Poulev
- Department of Plant Biology, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Yang Wang
- Department of Plant Biology, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Anik Boudreau
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808, United States
| | - Ilya Raskin
- Department of Plant Biology, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Jonathan Bisson
- Center for Natural Product Technologies, Pharmacognosy Institute, and Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Illinois 60612, United States
| | - G. Joseph Ray
- Pharmacognosy Institute and Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Illinois 60612, United States
| | - Shao-Nong Chen
- Center for Natural Product Technologies, Pharmacognosy Institute, and Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Illinois 60612, United States
| | - Allison Richard
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808, United States
| | - Jacqueline M. Stephens
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808, United States
| | - Guido F. Pauli
- Corresponding Authors Tel: +1 312 355 1949 Fax: +1 312 413 5894 (David Ribnicky): (Guido Pauli):
| |
Collapse
|
168
|
Zhang M, Ceyhan Y, Kaftanovskaya EM, Vasquez JL, Vacher J, Knop FK, Nathanson L, Agoulnik AI, Ittmann MM, Agoulnik IU. INPP4B protects from metabolic syndrome and associated disorders. Commun Biol 2021; 4:416. [PMID: 33772116 PMCID: PMC7998001 DOI: 10.1038/s42003-021-01940-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/03/2021] [Indexed: 02/01/2023] Open
Abstract
A high fat diet and obesity have been linked to the development of metabolic dysfunction and the promotion of multiple cancers. The causative cellular signals are multifactorial and not yet completely understood. In this report, we show that Inositol Polyphosphate-4-Phosphatase Type II B (INPP4B) signaling protects mice from diet-induced metabolic dysfunction. INPP4B suppresses AKT and PKC signaling in the liver thereby improving insulin sensitivity. INPP4B loss results in the proteolytic cleavage and activation of a key regulator in de novo lipogenesis and lipid storage, SREBP1. In mice fed with the high fat diet, SREBP1 increases expression and activity of PPARG and other lipogenic pathways, leading to obesity and non-alcoholic fatty liver disease (NAFLD). Inpp4b-/- male mice have reduced energy expenditure and respiratory exchange ratio leading to increased adiposity and insulin resistance. When treated with high fat diet, Inpp4b-/- males develop type II diabetes and inflammation of adipose tissue and prostate. In turn, inflammation drives the development of high-grade prostatic intraepithelial neoplasia (PIN). Thus, INPP4B plays a crucial role in maintenance of overall metabolic health and protects from prostate neoplasms associated with metabolic dysfunction.
Collapse
Affiliation(s)
- Manqi Zhang
- Department of Medicine, Duke University, Durham, NC, USA
| | - Yasemin Ceyhan
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Elena M Kaftanovskaya
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Judy L Vasquez
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Jean Vacher
- Department of Medicine, Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, QC, Canada
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Lubov Nathanson
- Institute for Neuro Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Alexander I Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL, USA
| | - Michael M Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX, USA
| | - Irina U Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
- Biomolecular Sciences Institute, Florida International University, Miami, FL, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
169
|
Verschoor PJ, Greig FH, Rochford JJ, Levate G, Delibegovic M, Thompson D, Leeson-Payne A, Dekeryte R, Banks R, Ramos JW, Nixon GF. Phosphoprotein enriched in astrocytes (PEA)-15 is a novel regulator of adipose tissue expansion. Sci Rep 2021; 11:6949. [PMID: 33772049 PMCID: PMC7997924 DOI: 10.1038/s41598-021-86250-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/26/2021] [Indexed: 12/17/2022] Open
Abstract
Excessive expansion of adipose tissue in obesity typically leads to overflow and accumulation of lipids in other tissues, causing fatty liver disease and atherosclerosis. The intracellular protein, phosphoprotein enriched in astrocytes (PEA)-15 has been linked to metabolic disease but its role in lipid storage has not been examined. To delineate the role of PEA-15 in adipose tissue, we placed PEA-15−/− mice on a high fat diet. These mice developed increased body weight and greater white adipose tissue expansion compared to high fat diet-fed wild type mice. This was due to increased adipocyte cell size in PEA-15−/− mice consistent with greater lipid storage capacity. Surprisingly, PEA-15−/− mice exhibited improvements in whole body insulin sensitivity, lower hepatic weight and decreased serum triglycerides indicating a protective phenotype. To determine effects on atherosclerosis, PEA-15−/− mice were crossed with the ApoE−/− mice on a high fat diet. Strikingly, these mice were protected from atherosclerosis and had less hepatic lipid accumulation despite increased adiposity. Therefore, we reveal for the first time that PEA-15 plays a novel role in regulating the expansion of adipose tissue. Decreasing PEA-15 expression increases the sequestering of lipids in adipose tissue, protecting other tissues in obesity, thereby improving metabolic health.
Collapse
Affiliation(s)
- Pola J Verschoor
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Fiona H Greig
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Justin J Rochford
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.,Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Giovanni Levate
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Mirela Delibegovic
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Dawn Thompson
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | | | - Ruta Dekeryte
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Ruth Banks
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Joe W Ramos
- University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, USA
| | - Graeme F Nixon
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
170
|
Adipose stem cells in obesity: challenges and opportunities. Biosci Rep 2021; 40:225001. [PMID: 32452515 PMCID: PMC7284323 DOI: 10.1042/bsr20194076] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/08/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue, the storage of excessive energy in the body, secretes various proteins called adipokines, which connect the body’s nutritional status to the regulation of energy balance. Obesity triggers alterations of quantity and quality of various types of cells that reside in adipose tissue, including adipose stem cells (ASCs; referred to as adipose-derived stem/stromal cells in vitro). These alterations in the functionalities and properties of ASCs impair adipose tissue remodeling and adipose tissue function, which induces low-grade systemic inflammation, progressive insulin resistance, and other metabolic disorders. In contrast, the ability of ASCs to recruit new adipocytes when faced with caloric excess leads to healthy adipose tissue expansion, associated with lower amounts of inflammation, fibrosis, and insulin resistance. This review focuses on recent advances in our understanding of the identity of ASCs and their roles in adipose tissue development, homeostasis, expansion, and thermogenesis, and how these roles go awry in obesity. A better understanding of the biology of ASCs and their adipogenesis may lead to novel therapeutic targets for obesity and metabolic disease.
Collapse
|
171
|
Maeda R, Kami D, Shikuma A, Suzuki Y, Taya T, Matoba S, Gojo S. RNA decay in processing bodies is indispensable for adipogenesis. Cell Death Dis 2021; 12:285. [PMID: 33731683 PMCID: PMC7969960 DOI: 10.1038/s41419-021-03537-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 12/19/2022]
Abstract
The RNA decay pathway plays key regulatory roles in cell identities and differentiation processes. Although adipogenesis is transcriptionally and epigenetically regulated and has been thoroughly investigated, how RNA metabolism that contributes to the stability of phenotype-shaping transcriptomes participates in differentiation remains elusive. In this study, we investigated Ddx6, an essential component of processing bodies (PBs) that executes RNA decay and translational repression in the cytoplasm and participates in the cellular transition of reprogramming. Upon adipogenic induction, Ddx6 dynamically accumulated to form PBs with a binding partner, 4E-T, at the early phase prior to emergence of intracellular lipid droplets. In contrast, preadipocytes with Ddx6 knockout (KO) or 4E-T knockdown (KD) failed to generate PBs, resulting in significant suppression of adipogenesis. Transcription factors related to preadipocytes and negative regulators of adipogenesis that were not expressed under adipogenic stimulation were maintained in Ddx6-KO and 4E-T-KD preadipocytes under adipogenic induction. Elimination of Dlk1, a major negative regulator of adipogenesis, in 3T3L1 Ddx6-KO cells did not restore adipogenic differentiation capacity to any extent. Similar to murine cells, human primary mesenchymal stem cells, which can differentiate into adipocytes upon stimulation with adipogenic cocktails, required DDX6 to maturate into adipocytes. Therefore, RNA decay of the entire parental transcriptome, rather than removal of a strong negative regulator, could be indispensable for adipogenesis.
Collapse
Affiliation(s)
- Ryotaro Maeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Kami
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akira Shikuma
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yosuke Suzuki
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshihiko Taya
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Gojo
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| |
Collapse
|
172
|
Gómez-Zorita S, Queralt M, Vicente MA, González M, Portillo MP. Metabolically healthy obesity and metabolically obese normal weight: a review. J Physiol Biochem 2021; 77:175-189. [PMID: 33704694 DOI: 10.1007/s13105-020-00781-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023]
Abstract
Despite the general relationship between obesity and its co-morbidities, there are both obese individuals who scarcely present the associated pathologies (metabolically healthy obese; MHO) and individuals who present obesity alterations despite having normal weight (metabolically obese normal weight; MONW). It is still difficult to define metabolically MHO and MONW individuals because different classifications have been used in the studies reported. Indeed, different inclusion criteria have been used to discriminate between metabolically healthy and metabolically unhealthy subjects. Due to this and other reasons, such as differences in ethnicity, genetics, and lifestyle of the populations, data concerning the prevalence of MHO and MONW are very variable. The main determinants of MHO are type of growth (hypertrophy or hyperplasia), anatomical location, inflammation of adipose tissue, ectopic fat accumulation, genetic factors, and lifestyles factors. In the case of MONW, the main determinants are genetic background and lifestyle factors. With regard to treatment, it is not clear whether MHO subjects would benefit from traditional lifestyle interventions, based on diet energy restriction and increased physical activity. For MONW subjects, there is still no specialized treatment, and the therapies are the same as those used in obese subjects.
Collapse
Affiliation(s)
- Saioa Gómez-Zorita
- Nutrition and Obesity Group. Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, Spain. .,BIOARABA Health Research Institute, Vitoria, Spain. .,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Vitoria, Spain.
| | - Maite Queralt
- Nutrition and Obesity Group. Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, Spain
| | - Maria Angeles Vicente
- BIOARABA Health Research Institute, Vitoria, Spain.,Alava University Hospital (Osakidetza), Vitoria, Spain
| | - Marcela González
- Nutrition and Food Science Department, Faculty of Biochemistry and Biological Sciences, National University of Litoral and National Scientific and Technical Research Council (CONICET), 3000, Santa Fe, Argentina
| | - María P Portillo
- Nutrition and Obesity Group. Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, Spain.,BIOARABA Health Research Institute, Vitoria, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Vitoria, Spain
| |
Collapse
|
173
|
Asc-1 regulates white versus beige adipocyte fate in a subcutaneous stromal cell population. Nat Commun 2021; 12:1588. [PMID: 33707431 PMCID: PMC7952576 DOI: 10.1038/s41467-021-21826-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 02/10/2021] [Indexed: 01/08/2023] Open
Abstract
Adipose tissue expansion, as seen in obesity, is often metabolically detrimental causing insulin resistance and the metabolic syndrome. However, white adipose tissue expansion at early ages is essential to establish a functional metabolism. To understand the differences between adolescent and adult adipose tissue expansion, we studied the cellular composition of the stromal vascular fraction of subcutaneous adipose tissue of two and eight weeks old mice using single cell RNA sequencing. We identified a subset of adolescent preadipocytes expressing the mature white adipocyte marker Asc-1 that showed a low ability to differentiate into beige adipocytes compared to Asc-1 negative cells in vitro. Loss of Asc-1 in subcutaneous preadipocytes resulted in spontaneous differentiation of beige adipocytes in vitro and in vivo. Mechanistically, this was mediated by a function of the amino acid transporter ASC-1 specifically in proliferating preadipocytes involving the intracellular accumulation of the ASC-1 cargo D-serine. Adipose tissue is composed of a variety of cell types, including adipocyte precursor populations, that contribute to adipose tissue function upon differentiation. Here, using scRNA-sequencing of adolescent and adult mouse subcutaneous adipose tissue, the authors identify an Asc-1 positive preadipocyte population that is enriched in adolescent subcutaneous fat and demonstrate that loss of Asc-1 triggers spontaneous beige adipocyte differentiation.
Collapse
|
174
|
The Potential Role of Exosomes in Child and Adolescent Obesity. CHILDREN-BASEL 2021; 8:children8030196. [PMID: 33800718 PMCID: PMC7999028 DOI: 10.3390/children8030196] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/17/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023]
Abstract
Child and adolescent obesity constitute one of the greatest contemporary public health menaces. The enduring disproportion between calorie intake and energy consumption, determined by a complex interaction of genetic, epigenetic, and environmental factors, finally leads to the development of overweight and obesity. Child and adolescent overweight/obesity promotes smoldering systemic inflammation (“para-inflammation”) and increases the likelihood of later metabolic and cardiovascular complications, including metabolic syndrome and its components, which progressively deteriorate during adulthood. Exosomes are endosome-derived extracellular vesicles that are secreted by a variety of cells, are naturally taken-up by target cells, and may be involved in many physiological and pathological processes. Over the last decade, intensive research has been conducted regarding the special role of exosomes and the non-coding (nc) RNAs they contain (primarily micro (mi) RNAs, long (l) non-coding RNAs, messenger (m) RNAs and other molecules) in inter-cellular communications. Through their action as communication mediators, exosomes may contribute to the pathogenesis of obesity and associated disorders. There is increasing evidence that exosomal miRNAs and lncRNAs are involved in pivotal processes of adipocyte biology and that, possibly, play important roles in gene regulation linked to human obesity. This review aims to improve our understanding of the roles of exosomes and their cargo in the development of obesity and related metabolic and inflammatory disorders. We examined their potential roles in adipose tissue physiology and reviewed the scarce data regarding the altered patterns of circulating miRNAs and lncRNAs observed in obese children and adolescents, compared them to the equivalent, more abundant existing findings of adult studies, and speculated on their proposed mechanisms of action. Exosomal miRNAs and lncRNAs could be applied as cardiometabolic risk biomarkers, useful in the early diagnosis and prevention of obesity. Furthermore, the targeting of crucial circulating exosomal cargo to tissues involved in the pathogenesis and maintenance of obesity could provide a novel therapeutic approach to this devastating and management-resistant pandemic.
Collapse
|
175
|
Candi E, Campanelli M, Sica G, Schinzari F, Rovella V, Di Daniele N, Melino J, Tesauro M. Differences in the vascular and metabolic profiles between metabolically healthy and unhealthy obesity. ENDOCRINE AND METABOLIC SCIENCE 2021; 2:100077. [DOI: 10.1016/j.endmts.2020.100077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
176
|
Al-Mrabeh A. β-Cell Dysfunction, Hepatic Lipid Metabolism, and Cardiovascular Health in Type 2 Diabetes: New Directions of Research and Novel Therapeutic Strategies. Biomedicines 2021; 9:226. [PMID: 33672162 PMCID: PMC7927138 DOI: 10.3390/biomedicines9020226] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/09/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) remains a major problem for people with type 2 diabetes mellitus (T2DM), and dyslipidemia is one of the main drivers for both metabolic diseases. In this review, the major pathophysiological and molecular mechanisms of β-cell dysfunction and recovery in T2DM are discussed in the context of abnormal hepatic lipid metabolism and cardiovascular health. (i) In normal health, continuous exposure of the pancreas to nutrient stimulus increases the demand on β-cells. In the long term, this will not only stress β-cells and decrease their insulin secretory capacity, but also will blunt the cellular response to insulin. (ii) At the pre-diabetes stage, β-cells compensate for insulin resistance through hypersecretion of insulin. This increases the metabolic burden on the stressed β-cells and changes hepatic lipoprotein metabolism and adipose tissue function. (iii) If this lipotoxic hyperinsulinemic environment is not removed, β-cells start to lose function, and CVD risk rises due to lower lipoprotein clearance. (iv) Once developed, T2DM can be reversed by weight loss, a process described recently as remission. However, the precise mechanism(s) by which calorie restriction causes normalization of lipoprotein metabolism and restores β-cell function are not fully established. Understanding the pathophysiological and molecular basis of β-cell failure and recovery during remission is critical to reduce β-cell burden and loss of function. The aim of this review is to highlight the link between lipoprotein export and lipid-driven β-cell dysfunction in T2DM and how this is related to cardiovascular health. A second aim is to understand the mechanisms of β-cell recovery after weight loss, and to explore new areas of research for developing more targeted future therapies to prevent T2DM and the associated CVD events.
Collapse
Affiliation(s)
- Ahmad Al-Mrabeh
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Magnetic Resonance Centre, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
177
|
Rajesh Y, Sarkar D. Association of Adipose Tissue and Adipokines with Development of Obesity-Induced Liver Cancer. Int J Mol Sci 2021; 22:ijms22042163. [PMID: 33671547 PMCID: PMC7926723 DOI: 10.3390/ijms22042163] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/20/2022] Open
Abstract
Obesity is rapidly dispersing all around the world and is closely associated with a high risk of metabolic diseases such as insulin resistance, dyslipidemia, and nonalcoholic fatty liver disease (NAFLD), leading to carcinogenesis, especially hepatocellular carcinoma (HCC). It results from an imbalance between food intake and energy expenditure, leading to an excessive accumulation of adipose tissue (AT). Adipocytes play a substantial role in the tumor microenvironment through the secretion of several adipokines, affecting cancer progression, metastasis, and chemoresistance via diverse signaling pathways. AT is considered an endocrine organ owing to its ability to secrete adipokines, such as leptin, adiponectin, resistin, and a plethora of inflammatory cytokines, which modulate insulin sensitivity and trigger chronic low-grade inflammation in different organs. Even though the precise mechanisms are still unfolding, it is now established that the dysregulated secretion of adipokines by AT contributes to the development of obesity-related metabolic disorders. This review focuses on several obesity-associated adipokines and their impact on obesity-related metabolic diseases, subsequent metabolic complications, and progression to HCC, as well as their role as potential therapeutic targets. The field is rapidly developing, and further research is still required to fully understand the underlying mechanisms for the metabolic actions of adipokines and their role in obesity-associated HCC.
Collapse
Affiliation(s)
- Yetirajam Rajesh
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Devanand Sarkar
- Massey Cancer Center, Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine (VIMM), Virginia Commonwealth University, Richmond, VA 23298, USA
- Correspondence: ; Tel.: +1-804-827-2339
| |
Collapse
|
178
|
Perpétuo L, Voisin PM, Amado F, Hirtz C, Vitorino R. Ghrelin and adipokines: An overview of their physiological role, antimicrobial activity and impact on cardiovascular conditions. VITAMINS AND HORMONES 2021; 115:477-509. [PMID: 33706959 DOI: 10.1016/bs.vh.2020.12.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The human body has many different hormones that interact with each other and with other factors such as proteins, cell receptors and metabolites. There is still a limited understanding of some of the underlying biological mechanisms of some hormones. In the past decades, science and technology have made major advancements in regard to innovation and knowledge in fields such as medicine. However, some conditions are complex and have many variables that their full picture is still unclear, even though some of these conditions have an alarming rate of incidence and serious health consequences. Conditions such as type 2 diabetes, obesity, nonalcoholic liver disease (NAFLD), cancer in its different forms and even mental conditions, such as Alzheimer's disease, are some of the most common diseases in the 21st century. These conditions are relevant not only because of their high incidence on the general population, but also because of their severity. In this chapter, we present an overview of cardiovascular (CV) diseases. According to the World Health Organization (WHO), cardiovascular diseases, such as coronary artery disease (CAD), heart attack, cardiomyopathy and heart failure (among others), are the number one cause of death worldwide. In 2016, it was estimated that 17.9 million people died from CV diseases, representing more than 30% of all global deaths. Approximately 95% of people who died from CV diseases were so-called "premature deaths" because were referenced to individuals under the age of 70 years old. In this chapter we described some of the hormones that may have an impact on CV diseases, including ghrelin, a peptide that is mostly produced in the stomach, known to induce hunger. Ghrelin is linked to an increase in body fat, i.e., adipose tissue in animals. For this reason, we also included the adipokines leptin, adiponectin and resistin. The main objectives of this chapter are to present the state of the art knowledge concerning the mechanisms of each hormone relevant to CV diseases; to compile data and results that further elucidate the relevance of these peptides for several physiological events, conditions and diseases; and to discuss the metabolic impact of each hormone. We established connections between multiple peptides and the underlying condition/disease with tools such as STRING, referring to research using databases, such as UniProt, DisGeNET and Proteomics DB. Fig. 1 shows a network that summarizes the information presented in this chapter, which serves as a visual representation.
Collapse
Affiliation(s)
- Luís Perpétuo
- iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | | | - Francisco Amado
- LAQV-REQUIMTE, Departamento de Química, Universidade de Aveiro, Aveiro, Portugal
| | - Christophe Hirtz
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Rui Vitorino
- iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal; UnIC, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina da Universidade do Porto, Porto, Portugal; LAQV-REQUIMTE, Departamento de Química, Universidade de Aveiro, Aveiro, Portugal.
| |
Collapse
|
179
|
Hydes T, Alam U, Cuthbertson DJ. The Impact of Macronutrient Intake on Non-alcoholic Fatty Liver Disease (NAFLD): Too Much Fat, Too Much Carbohydrate, or Just Too Many Calories? Front Nutr 2021; 8:640557. [PMID: 33665203 PMCID: PMC7921724 DOI: 10.3389/fnut.2021.640557] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a growing epidemic, in parallel with the obesity crisis, rapidly becoming one of the commonest causes of chronic liver disease worldwide. Diet and physical activity are important determinants of liver fat accumulation related to insulin resistance, dysfunctional adipose tissue, and secondary impaired lipid storage and/or increased lipolysis. While it is evident that a hypercaloric diet (an overconsumption of calories) promotes liver fat accumulation, it is also clear that the macronutrient composition can modulate this risk. A number of other baseline factors modify the overfeeding response, which may be genetic or environmental. Although it is difficult to disentangle the effects of excess calories vs. specifically the individual effects of excessive carbohydrates and/or fats, isocaloric, and hypercaloric dietary intervention studies have been implemented to provide insight into the effects of different macronutrients, sub-types and their relative balance, on the regulation of liver fat. What has emerged is that different types of fat and carbohydrates differentially influence liver fat accumulation, even when diets are isocaloric. Furthermore, distinct molecular and metabolic pathways mediate the effects of carbohydrates and fat intake on hepatic steatosis. Fat accumulation appears to act through impairments in lipid storage and/or increased lipolysis, whereas carbohydrate consumption has been shown to promote liver fat accumulation through de novo lipogenesis. Effects differ dependent upon carbohydrate and fat type. Saturated fat and fructose induce the greatest increase in intrahepatic triglycerides (IHTG), insulin resistance, and harmful ceramides compared with unsaturated fats, which have been found to be protective. Decreased intake of saturated fats and avoidance of added sugars are therefore the two most important dietary interventions that can lead to a reduction in IHTG and potentially the associated risk of developing type 2 diabetes. A healthy and balanced diet and regular physical activity must remain the cornerstones of effective lifestyle intervention to prevent the development and progression of NAFLD. Considering the sub-type of each macronutrient, in addition to the quantity, are critical determinants of liver health.
Collapse
Affiliation(s)
- Theresa Hydes
- Department of Metabolic and Cardiovascular Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.,Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Uazman Alam
- Department of Metabolic and Cardiovascular Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.,Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Daniel J Cuthbertson
- Department of Metabolic and Cardiovascular Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.,Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
180
|
Passos GR, Ghezzi AC, Antunes E, de Oliveira MG, Mónica FZ. The Role of Periprostatic Adipose Tissue on Prostate Function in Vascular-Related Disorders. Front Pharmacol 2021; 12:626155. [PMID: 33643052 PMCID: PMC7908035 DOI: 10.3389/fphar.2021.626155] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
The lower urinary tract symptoms (LUTS) secondary to benign prostatic hyperplasia (BPH) are highly prevalent worldwide. Clinical and experimental data suggest that the incidence of LUTS-BPH is higher in patients with vascular-related disorders such as in pelvic ischemia, obesity and diabetes as well as in the ageing population. Obesity is an important risk factor that predisposes to glucose intolerance, insulin resistance, dyslipidemia, type 2 diabetes mellitus and cardiovascular disorders. Prospective studies showed that obese men are more likely to develop LUTS-BPH than non-obese men. Yet, men with greater waist circumferences were also at a greater risk of increased prostate volume and prostate-specific antigen than men with lower waist circumference. BPH is characterized by an enlarged prostate and increased smooth muscle tone, thus causing urinary symptoms. Data from experimental studies showed a significant increase in prostate and epididymal adipose tissue weight of obese mice when compared with lean mice. Adipose tissues that are in direct contact with specific organs have gained attention due to their potential paracrine role. The prostate gland is surrounded by periprostatic adipose tissue (PPAT), which is believed to play a paracrine role by releasing growth factors, pro-inflammatory, pro-oxidant, contractile and anti-contractile substances that interfere in prostate reactivity and growth. Therefore, this review is divided into two main parts, one focusing on the role of adipokines in the context of obesity that can lead to LUTS/BPH and the second part focusing on the mediators released from PPAT and the possible pathways that may interfere in the prostate microenvironment.
Collapse
|
181
|
Lourido F, Quenti D, Salgado-Canales D, Tobar N. Domeless receptor loss in fat body tissue reverts insulin resistance induced by a high-sugar diet in Drosophila melanogaster. Sci Rep 2021; 11:3263. [PMID: 33547367 PMCID: PMC7864986 DOI: 10.1038/s41598-021-82944-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Insulin resistance is a hallmark of type 2 diabetes resulting from the confluence of several factors, including genetic susceptibility, inflammation, and diet. Under this pathophysiological condition, the dysfunction of the adipose tissue triggered by the excess caloric supply promotes the loss of sensitivity to insulin at the local and peripheral level, a process in which different signaling pathways are involved that are part of the metabolic response to the diet. Besides, the dysregulation of insulin signaling is strongly associated with inflammatory processes in which the JAK/STAT pathway plays a central role. To better understand the role of JAK/STAT signaling in the development of insulin resistance, we used a simple organism, Drosophila melanogaster, as a type 2 diabetes model generated by the consumption of a high-sugar diet. In this model, we studied the effects of inhibiting the expression of the JAK/STAT pathway receptor Domeless, in fat body, on adipose metabolism and glycemic control. Our results show that the Domeless receptor loss in fat body cells reverses both hyperglycemia and the increase in the expression of the insulin resistance marker Nlaz, observed in larvae fed a high sugar diet. This effect is consistent with a significant reduction in Dilp2 mRNA expression and an increase in body weight compared to wild-type flies fed high sugar diets. Additionally, the loss of Domeless reduced the accumulation of triglycerides in the fat body cells of larvae fed HSD and also significantly increased the lifespan of adult flies. Taken together, our results show that the loss of Domeless in the fat body reverses at least in part the dysmetabolism induced by a high sugar diet in a Drosophila type 2 diabetes model.
Collapse
Affiliation(s)
- Fernanda Lourido
- Cellular Biology Laboratory, Institute of Nutrition and Food Technology (INTA), University of Chile, Av. El Líbano, 5524, Macul, Santiago, Chile
| | - Daniela Quenti
- Cellular Biology Laboratory, Institute of Nutrition and Food Technology (INTA), University of Chile, Av. El Líbano, 5524, Macul, Santiago, Chile
| | - Daniela Salgado-Canales
- Cellular Biology Laboratory, Institute of Nutrition and Food Technology (INTA), University of Chile, Av. El Líbano, 5524, Macul, Santiago, Chile
| | - Nicolás Tobar
- Cellular Biology Laboratory, Institute of Nutrition and Food Technology (INTA), University of Chile, Av. El Líbano, 5524, Macul, Santiago, Chile.
| |
Collapse
|
182
|
Galanin I, Nicu C, Tower JI. Facial Fat Fitness: A New Paradigm to Understand Facial Aging and Aesthetics. Aesthetic Plast Surg 2021; 45:151-163. [PMID: 32914326 DOI: 10.1007/s00266-020-01933-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/16/2020] [Indexed: 12/22/2022]
Abstract
Traditionally, facial adipose tissue has been perceived and treated as a homogenous volume-occupying subcutaneous depot. However, recent research from across disciplines is converging to reveal a far more anatomically organized and functionally dynamic role of facial adipose tissue. In this narrative review, we will discuss new insights into adipocyte function and facial adipose anatomy that have far-reaching implications for the practice of aesthetic facial plastic surgery. These concepts are synthesized into a "facial fat fitness" model which can be used to explain clinical observations in facial aging and aesthetic surgery. Fat fitness relates to the quality of facial adipose tissue, as opposed to quantity, and describes whether adipose tissue is in a predominantly healthy hyperplastic or unhealthy hypertrophic state. Fat fitness is modulated by lifestyle factors, and may be impacted positively or negatively by facial aesthetic treatments. LEVEL OF EVIDENCE IV: This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
| | - Carina Nicu
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jacob I Tower
- Department of Otolaryngology-Head and Neck Surgery, Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, 130 East 77th Street, 10th Floor, New York, NY, USA.
| |
Collapse
|
183
|
Wang L, Luo Y, Luo L, Wu D, Ding X, Zheng H, Wu H, Liu B, Yang X, Silva F, Wang C, Zhang X, Zheng X, Chen J, Brigman J, Mandell M, Zhou Z, Liu F, Yang XO, Liu M. Adiponectin restrains ILC2 activation by AMPK-mediated feedback inhibition of IL-33 signaling. J Exp Med 2021; 218:e20191054. [PMID: 33104171 PMCID: PMC7590510 DOI: 10.1084/jem.20191054] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/20/2019] [Accepted: 09/01/2020] [Indexed: 12/11/2022] Open
Abstract
ILC2s are present in adipose tissue and play a critical role in regulating adipose thermogenesis. However, the mechanisms underlying the activation of adipose-resident ILC2s remain poorly defined. Here, we show that IL-33, a potent ILC2 activator, stimulates phosphorylation of AMPK at Thr172 via TAK1 in primary ILC2s, which provides a feedback mechanism to inhibit IL-33-induced NF-κB activation and IL-13 production. Treating ILC2s with adiponectin or an adiponectin receptor agonist (AdipoRon) activated AMPK and decreased IL-33-NF-κB signaling. AdipoRon also suppressed cold-induced thermogenic gene expression and energy expenditure in vivo. In contrast, adiponectin deficiency increased the ILC2 fraction and activation, leading to up-regulated thermogenic gene expression in adipose tissue of cold-exposed mice. ILC2 deficiency or blocking ILC2 function by neutralization of the IL-33 receptor with anti-ST2 diminished the suppressive effect of adiponectin on cold-induced adipose thermogenesis and energy expenditure. Taken together, our study reveals that adiponectin is a negative regulator of ILC2 function in adipose tissue via AMPK-mediated negative regulation of IL-33 signaling.
Collapse
Affiliation(s)
- Lu Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Luo
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Central South University, Changsha, Hunan, China
| | - Liping Luo
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Dandan Wu
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM
- Department of Microbiology and Molecular Genetics, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Xiaofeng Ding
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Handong Zheng
- Department of Microbiology and Molecular Genetics, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Haisha Wu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bilian Liu
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Central South University, Changsha, Hunan, China
| | - Xin Yang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Floyd Silva
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Chunqing Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Xianyun Zheng
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Jindong Chen
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jonathan Brigman
- Department of Neuroscience, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Michael Mandell
- Department of Microbiology and Molecular Genetics, University of New Mexico Health Sciences Center, Albuquerque, NM
- Autophagy, Inflammation and Metabolism Center for Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Zhiguang Zhou
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Central South University, Changsha, Hunan, China
| | - Feng Liu
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX
| | - Xuexian O. Yang
- Department of Microbiology and Molecular Genetics, University of New Mexico Health Sciences Center, Albuquerque, NM
- Autophagy, Inflammation and Metabolism Center for Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM
- Autophagy, Inflammation and Metabolism Center for Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM
| |
Collapse
|
184
|
Chen YJ, Lin CW, Peng YJ, Huang CW, Chien YS, Huang TH, Liao PX, Yang WY, Wang MH, Mersmann HJ, Wu SC, Chuang TY, Lin YY, Kuo WH, Ding ST. Overexpression of Adiponectin Receptor 1 Inhibits Brown and Beige Adipose Tissue Activity in Mice. Int J Mol Sci 2021; 22:ijms22020906. [PMID: 33477525 PMCID: PMC7831094 DOI: 10.3390/ijms22020906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 01/05/2023] Open
Abstract
Adult humans and mice possess significant classical brown adipose tissues (BAT) and, upon cold-induction, acquire brown-like adipocytes in certain depots of white adipose tissues (WAT), known as beige adipose tissues or WAT browning/beiging. Activating thermogenic classical BAT or WAT beiging to generate heat limits diet-induced obesity or type-2 diabetes in mice. Adiponectin is a beneficial adipokine resisting diabetes, and causing “healthy obese” by increasing WAT expansion to limit lipotoxicity in other metabolic tissues during high-fat feeding. However, the role of its receptors, especially adiponectin receptor 1 (AdipoR1), on cold-induced thermogenesis in vivo in BAT and in WAT beiging is still elusive. Here, we established a cold-induction procedure in transgenic mice over-expressing AdipoR1 and applied a live 3-D [18F] fluorodeoxyglucose-PET/CT (18F-FDG PET/CT) scanning to measure BAT activity by determining glucose uptake in cold-acclimated transgenic mice. Results showed that cold-acclimated mice over-expressing AdipoR1 had diminished cold-induced glucose uptake, enlarged adipocyte size in BAT and in browned WAT, and reduced surface BAT/body temperature in vivo. Furthermore, decreased gene expression, related to thermogenic Ucp1, BAT-specific markers, BAT-enriched mitochondrial markers, lipolysis and fatty acid oxidation, and increased expression of whitening genes in BAT or in browned subcutaneous inguinal WAT of AdipoR1 mice are congruent with results of PET/CT scanning and surface body temperature in vivo. Moreover, differentiated brown-like beige adipocytes isolated from pre-adipocytes in subcutaneous WAT of transgenic AdipoR1 mice also had similar effects of lowered expression of thermogenic Ucp1, BAT selective markers, and BAT mitochondrial markers. Therefore, this study combines in vitro and in vivo results with live 3-D scanning and reveals one of the many facets of the adiponectin receptors in regulating energy homeostasis, especially in the involvement of cold-induced thermogenesis.
Collapse
MESH Headings
- Adipocytes, Beige/metabolism
- Adipose Tissue, Beige/diagnostic imaging
- Adipose Tissue, Beige/metabolism
- Adipose Tissue, Brown/diagnostic imaging
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/diagnostic imaging
- Adipose Tissue, White/metabolism
- Animals
- Energy Metabolism/genetics
- Gene Expression Regulation, Developmental/genetics
- Mice
- Mice, Transgenic/genetics
- Mice, Transgenic/metabolism
- Mitochondria/genetics
- Obesity/genetics
- Obesity/metabolism
- Obesity/pathology
- Positron-Emission Tomography
- Receptors, Adiponectin/genetics
- Thermogenesis/genetics
- Uncoupling Protein 1/genetics
Collapse
Affiliation(s)
- Yu-Jen Chen
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan; (C.-W.L.); (S.-C.W.)
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
- Correspondence: (Y.-J.C.); (Y.-Y.L.); (W.-H.K.); (S.-T.D.); Tel.: +886-2-3366-4175 (S.-T.D.)
| | - Chiao-Wei Lin
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan; (C.-W.L.); (S.-C.W.)
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
| | - Yu-Ju Peng
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
| | - Chao-Wei Huang
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
| | - Yi-Shan Chien
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
| | - Tzu-Hsuan Huang
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
| | - Pei-Xin Liao
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
| | - Wen-Yuan Yang
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
| | - Mei-Hui Wang
- Institute of Nuclear Energy Research, Taoyuan 325, Taiwan;
| | - Harry J. Mersmann
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
| | - Shinn-Chih Wu
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan; (C.-W.L.); (S.-C.W.)
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
| | - Tai-Yuan Chuang
- Department of Athletics, National Taiwan University, Taipei 10617, Taiwan;
| | - Yuan-Yu Lin
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
- Correspondence: (Y.-J.C.); (Y.-Y.L.); (W.-H.K.); (S.-T.D.); Tel.: +886-2-3366-4175 (S.-T.D.)
| | - Wen-Hung Kuo
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 10617, Taiwan
- Correspondence: (Y.-J.C.); (Y.-Y.L.); (W.-H.K.); (S.-T.D.); Tel.: +886-2-3366-4175 (S.-T.D.)
| | - Shih-Torng Ding
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan; (C.-W.L.); (S.-C.W.)
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan; (Y.-J.P.); (C.-W.H.); (Y.-S.C.); (T.-H.H.); (P.-X.L.); (W.-Y.Y.); (H.J.M.)
- Correspondence: (Y.-J.C.); (Y.-Y.L.); (W.-H.K.); (S.-T.D.); Tel.: +886-2-3366-4175 (S.-T.D.)
| |
Collapse
|
185
|
Derous D, Sahu J, Douglas A, Lusseau D, Wenzel M. Comparative genomics of cetartiodactyla: energy metabolism underpins the transition to an aquatic lifestyle. CONSERVATION PHYSIOLOGY 2021; 9:coaa136. [PMID: 33505701 PMCID: PMC7816800 DOI: 10.1093/conphys/coaa136] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/09/2020] [Accepted: 12/19/2020] [Indexed: 05/28/2023]
Abstract
Foraging disruption caused by human activities is emerging as a key issue in cetacean conservation because it can affect nutrient levels and the amount of energy available to individuals to invest into reproduction. Our ability to predict how anthropogenic stressors affect these ecological processes and ultimately population trajectory depends crucially on our understanding of the complex physiological mechanisms that detect nutrient availability and regulate energy metabolism, foraging behavior and life-history decisions. These physiological mechanisms are likely to differ considerably from terrestrial mammalian model systems. Here, we examine nucleotide substitution rates in cetacean and other artiodactyl genomes to identify signatures of selection in genes associated with nutrient sensing pathways. We also estimated the likely physiological consequences of adaptive amino acid substitutions for pathway functions. Our results highlight that genes involved in the insulin, mTOR and NF-ĸB pathways are subject to significant positive selection in cetaceans compared to terrestrial artiodactyla. These genes may have been positively selected to enable cetaceans to adapt to a glucose-poor diet, to overcome deleterious effects caused by hypoxia during diving (e.g. oxidative stress and inflammation) and to modify fat-depot signaling functions in a manner different to terrestrial mammals. We thus show that adaptation in cetaceans to an aquatic lifestyle significantly affected functions in nutrient sensing pathways. The use of fat stores as a condition index in cetaceans may be confounded by the multiple and critical roles fat has in regulating cetacean metabolism, foraging behavior and diving physiology.
Collapse
Affiliation(s)
- Davina Derous
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Jagajjit Sahu
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Alex Douglas
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - David Lusseau
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
- National Institute of Aquatic Resources, Technical University of Denmark, Kgs. Lyngby, Copenhagen, Denmark
| | - Marius Wenzel
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
- Centre for Genome Enabled Biology and Medicine, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
186
|
Abstract
The landmark discoveries of leptin and adiponectin firmly established adipose tissue as a sophisticated and highly active endocrine organ, opening a new era of investigating adipose-mediated tissue crosstalk. Both obesity-associated hyperleptinemia and hypoadiponectinemia are important biomarkers to predict cardiovascular outcomes, suggesting a crucial role for adiponectin and leptin in obesity-associated cardiovascular disorders. Normal physiological levels of adiponectin and leptin are indeed essential to maintain proper cardiovascular function. Insufficient adiponectin and leptin signaling results in cardiovascular dysfunction. However, a paradox of high levels of both leptin and adiponectin is emerging in the pathogenesis of cardiovascular disorders. Here, we (1) summarize the recent progress in the field of adiponectin and leptin and its association with cardiovascular disorders, (2) further discuss the underlying mechanisms for this new paradox of leptin and adiponectin action, and (3) explore the possible application of partial leptin reduction, in addition to increasing the adiponectin/leptin ratio as a means to prevent or reverse cardiovascular disorders.
Collapse
Affiliation(s)
- Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine (S.Z., C.M.K., P.E.S.), The University of Texas Southwestern Medical Center, Dallas
| | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine (S.Z., C.M.K., P.E.S.), The University of Texas Southwestern Medical Center, Dallas
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine (S.Z., C.M.K., P.E.S.), The University of Texas Southwestern Medical Center, Dallas.,Department of Cell Biology (P.E.S.), The University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
187
|
Komici K, Dello Iacono A, De Luca A, Perrotta F, Bencivenga L, Rengo G, Rocca A, Guerra G. Adiponectin and Sarcopenia: A Systematic Review With Meta-Analysis. Front Endocrinol (Lausanne) 2021; 12:576619. [PMID: 33935962 PMCID: PMC8082154 DOI: 10.3389/fendo.2021.576619] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Sarcopenia is a progressive loss of skeletal muscle mass whose pathophysiology has been proposed to possibly involve mechanisms of altered inflammatory status and endocrine function. Adiponectin has been shown to modulate inflammatory status and muscle metabolism. However, the possible association between adiponectin levels and sarcopenia is poorly understood. In order to fill this gap, in the present manuscript we aimed to summarize the current evidence with a systematic review and a meta-analysis of studies reporting serum adiponectin levels in patients with sarcopenia compared to non-sarcopenic controls. METHODS An electronic search through Medline/PubMed, Cochrane Library, and Science Direct was performed till March 1, 2020. From the included papers, meta-analysis of cross-sectional studies comparing serum levels of adiponectin between patients with sarcopenia and controls was performed. RESULTS Out of 1,370 initial studies, seven studies were meta-analyzed. Sarcopenic participants had significantly higher levels of adiponectin Hedges' g with 95% confidence interval (CI): 1.20 (0.19-2.22), p = 0.02 than controls. Subgroup analysis, performed in Asian population and focused on identification of the condition based on AWGS criteria, reported higher adiponectin levels in sarcopenic population (2.1 (0.17-4.03), p = 0.03 and I2 = 98.98%. Meta-regression analysis revealed female gender to significantly influence the results as demonstrated by beta = 0.14 (95% CI (0.010-0.280), p = 0.040). CONCLUSIONS Our meta-analysis found evidence that sarcopenia is associated with higher adiponectin levels. However, caution is warranted on the interpretation of these findings, and future longitudinal research is required to disentangle and better understand the topic.
Collapse
Affiliation(s)
- Klara Komici
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
- *Correspondence: Klara Komici,
| | - Antonio Dello Iacono
- School of Health and Life Sciences, University of the West of Scotland, Hamilton, United Kingdom
| | - Antonio De Luca
- Section of Human Anatomy, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Fabio Perrotta
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
- Istituti Clinici Scientifici Maugeri SpA Società Benefit (ICS Maugeri SpA SB), Telese Terme, Italy
| | - Aldo Rocca
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| |
Collapse
|
188
|
da Silva Fernandes J, Schuelter-Trevisol F, Cancelier ACL, Gonçalves e Silva HC, de Sousa DG, Atkinson RL, Trevisol DJ. Adenovirus 36 prevalence and association with human obesity: a systematic review. Int J Obes (Lond) 2021; 45:1342-1356. [PMID: 33753885 PMCID: PMC7983349 DOI: 10.1038/s41366-021-00805-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/01/2021] [Accepted: 03/04/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Obesity has numerous etiologies and includes biological factors. Studies have demonstrated that the human adenovirus subtype 36 (Adv36) is an adipogenic agent and causes metabolic alterations. Study results on the prevalence of Adv36 and clinical effects in humans vary substantially. This was a systematic review to summarize the studies on the prevalence of Adv36 infection and its association with human obesity. METHODS A systematic literature review was conducted using the preferred reporting items for systematic reviews and meta-analysis (PRISMA). Observational or experimental studies found in the Medline, Embase, LILACS, Science Direct and SciELO databases that presented results on the prevalence of Adv36 in humans were included. RESULTS Thirty-seven studies were screened. A total of 10,300 adults aged 18-70 years and 4585 children and adolescents aged 3-18 years were assessed. The average prevalence of Adv36 among adults was 22.9%, ranging from 5.5% to 49.8%. Among children and adolescents, the average prevalence of Adv36 was 28.9%, ranging from 7.5% to 73.9%. There was a positive statistical relationship between Adv36 and weight gain, obesity, or metabolic changes in 31 studies. However, in four studies there was no association with obesity, and in one, no association was described. One of the studies showed an inverse correlation, i.e., Adv36 was a protective factor against obesity. CONCLUSION Strong evidence suggested a positive association between viral infection and obesity. However, due to the multi-causality of obesity and heterogeneity of studies, diagnostic tests should be standardized and easily accessible by the population to estimate the overall prevalence of Adv36 infection and its association with obesity.
Collapse
Affiliation(s)
- Jaime da Silva Fernandes
- grid.412297.b0000 0001 0648 9933Postgraduate Program in Health Sciences at University of Southern Santa Catarina (UNISUL), Tubarão, Brazil ,grid.412297.b0000 0001 0648 9933Medical School at University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Fabiana Schuelter-Trevisol
- grid.412297.b0000 0001 0648 9933Postgraduate Program in Health Sciences at University of Southern Santa Catarina (UNISUL), Tubarão, Brazil ,grid.412297.b0000 0001 0648 9933Medical School at University of Southern Santa Catarina (UNISUL), Tubarão, Brazil ,Clinical Research Center of Hospital Nossa Senhora da Conceição, Tubarão, Santa Catarina Brazil
| | - Ana Carolina Lobor Cancelier
- grid.412297.b0000 0001 0648 9933Postgraduate Program in Health Sciences at University of Southern Santa Catarina (UNISUL), Tubarão, Brazil ,grid.412297.b0000 0001 0648 9933Medical School at University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Helena Caetano Gonçalves e Silva
- grid.412297.b0000 0001 0648 9933Postgraduate Program in Health Sciences at University of Southern Santa Catarina (UNISUL), Tubarão, Brazil ,grid.412297.b0000 0001 0648 9933Medical School at University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Daiana Gomes de Sousa
- grid.412297.b0000 0001 0648 9933Medical School at University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Richard L. Atkinson
- grid.224260.00000 0004 0458 8737Virginia Commonwealth University, School of Medicine, Richmond, VA USA
| | - Daisson José Trevisol
- grid.412297.b0000 0001 0648 9933Postgraduate Program in Health Sciences at University of Southern Santa Catarina (UNISUL), Tubarão, Brazil ,grid.412297.b0000 0001 0648 9933Medical School at University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| |
Collapse
|
189
|
Siemienowicz KJ, Coukan F, Franks S, Rae MT, Duncan WC. Aberrant subcutaneous adipogenesis precedes adult metabolic dysfunction in an ovine model of polycystic ovary syndrome (PCOS). Mol Cell Endocrinol 2021; 519:111042. [PMID: 33010309 DOI: 10.1016/j.mce.2020.111042] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/13/2022]
Abstract
Polycystic ovary syndrome (PCOS) affects over 10% of women. Insulin resistance, elevated free fatty acids (FFAs) and increased adiposity are key factors contributing to metabolic dysfunction in PCOS. We hypothesised that aberrant adipogenesis during adolescence, and downstream metabolic perturbations, contributes to the metabolic phenotype of adult PCOS. We used prenatally androgenised (PA) sheep as a clinically realistic model of PCOS. During adolescence, but not during fetal or early life of PA sheep, adipogenesis was decreased in subcutaneous adipose tissue (SAT) accompanied by decreased leptin, adiponectin, and increased FFAs. In adulthood, PA sheep developed adipocyte hypertrophy in SAT paralleled by increased expression of inflammatory markers, elevated FFAs and increased expression of genes linked to fat accumulation in visceral adipose tissue. This study provides better understanding into the pathophysiology of PCOS from puberty to adulthood and identifies opportunity for early clinical intervention to normalise adipogenesis and ameliorate the metabolic phenotype.
Collapse
Affiliation(s)
- Katarzyna J Siemienowicz
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, EH16 4TJ, UK; School of Applied Sciences, Edinburgh Napier University, Edinburgh, EH11 4BN, UK.
| | - Flavien Coukan
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, EH11 4BN, UK
| | - Stephen Franks
- Institute of Reproductive and Developmental Biology, Imperial College, London, UK
| | - Mick T Rae
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, EH11 4BN, UK
| | - W Colin Duncan
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| |
Collapse
|
190
|
Kawai T, Autieri MV, Scalia R. Adipose tissue inflammation and metabolic dysfunction in obesity. Am J Physiol Cell Physiol 2020; 320:C375-C391. [PMID: 33356944 DOI: 10.1152/ajpcell.00379.2020] [Citation(s) in RCA: 907] [Impact Index Per Article: 181.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Several lines of preclinical and clinical research have confirmed that chronic low-grade inflammation of adipose tissue is mechanistically linked to metabolic disease and organ tissue complications in the overweight and obese organism. Despite this widely confirmed paradigm, numerous open questions and knowledge gaps remain to be investigated. This is mainly due to the intricately intertwined cross-talk of various pro- and anti-inflammatory signaling cascades involved in the immune response of expanding adipose depots, particularly the visceral adipose tissue. Adipose tissue inflammation is initiated and sustained over time by dysfunctional adipocytes that secrete inflammatory adipokines and by infiltration of bone marrow-derived immune cells that signal via production of cytokines and chemokines. Despite its low-grade nature, adipose tissue inflammation negatively impacts remote organ function, a phenomenon that is considered causative of the complications of obesity. The aim of this review is to broadly present an overview of adipose tissue inflammation by highlighting the most recent reports in the scientific literature and summarizing our overall understanding of the field. We also discuss key endogenous anti-inflammatory mediators and analyze their mechanistic role(s) in the pathogenesis and treatment of adipose tissue inflammation. In doing so, we hope to stimulate studies to uncover novel physiological, cellular, and molecular targets for the treatment of obesity.
Collapse
Affiliation(s)
- Tatsuo Kawai
- The Cardiovascular Research Center and The Limole Center for Integrated Lymphatic Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Michael V Autieri
- The Cardiovascular Research Center and The Limole Center for Integrated Lymphatic Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Rosario Scalia
- The Cardiovascular Research Center and The Limole Center for Integrated Lymphatic Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
191
|
Mechanisms by which adiponectin reverses high fat diet-induced insulin resistance in mice. Proc Natl Acad Sci U S A 2020; 117:32584-32593. [PMID: 33293421 PMCID: PMC7768680 DOI: 10.1073/pnas.1922169117] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adiponectin has emerged as a potential therapy for type 2 diabetes mellitus, but the molecular mechanism by which adiponectin reverses insulin resistance remains unclear. Two weeks of globular adiponectin (gAcrp30) treatment reduced fasting plasma glucose, triglyceride (TAG), and insulin concentrations and reversed whole-body insulin resistance, which could be attributed to both improved insulin-mediated suppression of endogenous glucose production and increased insulin-stimulated glucose uptake in muscle and adipose tissues. These improvements in liver and muscle sensitivity were associated with ∼50% reductions in liver and muscle TAG and plasma membrane (PM)-associated diacylglycerol (DAG) content and occurred independent of reductions in total ceramide content. Reductions of PM DAG content in liver and skeletal muscle were associated with reduced PKCε translocation in liver and reduced PKCθ and PKCε translocation in skeletal muscle resulting in increased insulin-stimulated insulin receptor tyrosine1162 phosphorylation, IRS-1/IRS-2-associated PI3-kinase activity, and Akt-serine phosphorylation. Both gAcrp30 and full-length adiponectin (Acrp30) treatment increased eNOS/AMPK activation in muscle and muscle fatty acid oxidation. gAcrp30 and Acrp30 infusions also increased TAG uptake in epididymal white adipose tissue (eWAT), which could be attributed to increased lipoprotein lipase (LPL) activity. These data suggest that adiponectin and adiponectin-related molecules reverse lipid-induced liver and muscle insulin resistance by reducing ectopic lipid storage in these organs, resulting in decreased plasma membrane sn-1,2-DAG-induced nPKC activity and increased insulin signaling. Adiponectin mediates these effects by both promoting the storage of TAG in eWAT likely through stimulation of LPL as well as by stimulation of AMPK in muscle resulting in increased muscle fat oxidation.
Collapse
|
192
|
Dankel SN, Grytten E, Bjune JI, Nielsen HJ, Dietrich A, Blüher M, Sagen JV, Mellgren G. COL6A3 expression in adipose tissue cells is associated with levels of the homeobox transcription factor PRRX1. Sci Rep 2020; 10:20164. [PMID: 33214660 PMCID: PMC7678848 DOI: 10.1038/s41598-020-77406-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/21/2020] [Indexed: 01/28/2023] Open
Abstract
Fibrillar collagen COL6α3 in adipose tissue has been associated with obesity, inflammation, insulin resistance and cancer. We here aimed to identify novel transcriptional regulators of COL6A3 expression. Based on a transcriptome dataset of adipose tissue, we identified strong correlations for 56 genes with COL6A3 mRNA, including targets of TGF-β/SMAD signaling. Among the identified candidates, the homeobox transcription factor PRRX1 showed a particularly striking co-expression with COL6A3, validated across several different cohorts, including patients with extreme obesity, insulin sensitive and resistant obesity (subcutaneous and omental), after profound fat loss (subcutaneous), and lean controls (subcutaneous). In human and mouse adipose cells, PRRX1 knockdown reduced COL6A3 mRNA and PRRX1 overexpression transactivated a reporter construct with the endogenous human COL6A3 promoter. Stable PRRX1 overexpression in 3T3-L1 cells induced Col6a3 mRNA threefold specifically after adipogenic induction, whereas TGF-β1 treatment upregulated Col6a3 mRNA also in the preadipocyte state. Interestingly, pro-inflammatory stimulus (i.e., TNF-α treatment) decreased PRRX1-mediated Col6a3 transactivation and mRNA expression, supporting a role for this mechanism in the regulation of adipose tissue inflammation. In conclusion, we identified the homeobox factor PRRX1 as a novel transcriptional regulator associated with COL6A3 expression, providing new insight into the regulatory mechanisms of altered adipose tissue function in obesity and insulin resistance.
Collapse
Affiliation(s)
- Simon N Dankel
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway. .,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway.
| | - Elise Grytten
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway.,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Jan-Inge Bjune
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway.,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | | | - Arne Dietrich
- Department of Surgery, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Jørn V Sagen
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway.,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway. .,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
193
|
BMP7 overexpression in adipose tissue induces white adipogenesis and improves insulin sensitivity in ob/ob mice. Int J Obes (Lond) 2020; 45:449-460. [PMID: 33110143 DOI: 10.1038/s41366-020-00700-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 08/26/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND/OBJECTIVES During obesity, hypertrophic enlargement of white adipose tissue (WAT) promotes ectopic lipid deposition and development of insulin resistance. In contrast, WAT hyperplasia is associated with preservation of insulin sensitivity. The complex network of factors that regulates white adipogenesis is not fully understood. Bone morphogenic protein 7 (BMP7) can induce brown adipogenesis, but its role on white adipogenesis remains to be elucidated. Here, we assessed BMP7-mediated effects on white adipogenesis in ob/ob mice. METHODS BMP7 was overexpressed in either WAT or liver of ob/ob mice using adeno-associated viral (AAV) vectors. Analysis of gene expression, histological and morphometric alterations, and metabolites and hormones concentrations were carried out. RESULTS Overexpression of BMP7 in adipocytes of subcutaneous and visceral WAT increased fat mass, the proportion of small-size adipocytes and the expression of adipogenic and mature adipocyte genes, suggesting induction of adipogenesis irrespective of fat depot. These changes were associated with reduced hepatic steatosis and improved insulin sensitivity. In contrast, liver-specific overproduction of BMP7 did not promote WAT hyperplasia despite BMP7 circulating levels were similar to those achieved after genetic engineering of WAT. CONCLUSIONS This study unravels a new autocrine/paracrine role of BMP7 on white adipogenesis and highlights that BMP7 may modulate WAT plasticity and increase insulin sensitivity.
Collapse
|
194
|
Adipocyte lipolysis: from molecular mechanisms of regulation to disease and therapeutics. Biochem J 2020; 477:985-1008. [PMID: 32168372 DOI: 10.1042/bcj20190468] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/19/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022]
Abstract
Fatty acids (FAs) are stored safely in the form of triacylglycerol (TAG) in lipid droplet (LD) organelles by professional storage cells called adipocytes. These lipids are mobilized during adipocyte lipolysis, the fundamental process of hydrolyzing TAG to FAs for internal or systemic energy use. Our understanding of adipocyte lipolysis has greatly increased over the past 50 years from a basic enzymatic process to a dynamic regulatory one, involving the assembly and disassembly of protein complexes on the surface of LDs. These dynamic interactions are regulated by hormonal signals such as catecholamines and insulin which have opposing effects on lipolysis. Upon stimulation, patatin-like phospholipase domain containing 2 (PNPLA2)/adipocyte triglyceride lipase (ATGL), the rate limiting enzyme for TAG hydrolysis, is activated by the interaction with its co-activator, alpha/beta hydrolase domain-containing protein 5 (ABHD5), which is normally bound to perilipin 1 (PLIN1). Recently identified negative regulators of lipolysis include G0/G1 switch gene 2 (G0S2) and PNPLA3 which interact with PNPLA2 and ABHD5, respectively. This review focuses on the dynamic protein-protein interactions involved in lipolysis and discusses some of the emerging concepts in the control of lipolysis that include allosteric regulation and protein turnover. Furthermore, recent research demonstrates that many of the proteins involved in adipocyte lipolysis are multifunctional enzymes and that lipolysis can mediate homeostatic metabolic signals at both the cellular and whole-body level to promote inter-organ communication. Finally, adipocyte lipolysis is involved in various diseases such as cancer, type 2 diabetes and fatty liver disease, and targeting adipocyte lipolysis is of therapeutic interest.
Collapse
|
195
|
Flavonoids in adipose tissue inflammation and atherosclerosis: one arrow, two targets. Clin Sci (Lond) 2020; 134:1403-1432. [PMID: 32556180 DOI: 10.1042/cs20200356] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023]
Abstract
Flavonoids are polyphenolic compounds naturally occurring in fruits and vegetables, in addition to beverages such as tea and coffee. Flavonoids are emerging as potent therapeutic agents for cardiovascular as well as metabolic diseases. Several studies corroborated an inverse relationship between flavonoid consumption and cardiovascular disease (CVD) or adipose tissue inflammation (ATI). Flavonoids exert their anti-atherogenic effects by increasing nitric oxide (NO), reducing reactive oxygen species (ROS), and decreasing pro-inflammatory cytokines. In addition, flavonoids alleviate ATI by decreasing triglyceride and cholesterol levels, as well as by attenuating inflammatory mediators. Furthermore, flavonoids inhibit synthesis of fatty acids and promote their oxidation. In this review, we discuss the effect of the main classes of flavonoids, namely flavones, flavonols, flavanols, flavanones, anthocyanins, and isoflavones, on atherosclerosis and ATI. In addition, we dissect the underlying molecular and cellular mechanisms of action for these flavonoids. We conclude by supporting the potential benefit for flavonoids in the management or treatment of CVD; yet, we call for more robust clinical studies for safety and pharmacokinetic values.
Collapse
|
196
|
Field BC, Gordillo R, Scherer PE. The Role of Ceramides in Diabetes and Cardiovascular Disease Regulation of Ceramides by Adipokines. Front Endocrinol (Lausanne) 2020; 11:569250. [PMID: 33133017 PMCID: PMC7564167 DOI: 10.3389/fendo.2020.569250] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolic dysfunction is intertwined with the pathophysiology of both diabetes and cardiovascular disease. Recently, one particular lipid class has been shown to influence the development and sustainment of these diseases: ceramides. As a subtype of sphingolipids, these species are particularly central to many sphingolipid pathways. Increased levels of ceramides are known to correlate with impaired cardiovascular and metabolic health. Furthermore, the interaction between ceramides and adipokines, most notably adiponectin and leptin, appears to play a role in the pathophysiology of these conditions. Adiponectin appears to counteract the detrimental effects of elevated ceramides, largely through activation of the ceramidase activity of its receptors. Elevated ceramides appear to worsen leptin resistance, which is an important phenomenon in the pathophysiology of obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Bianca C. Field
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
197
|
Ullah H, De Filippis A, Khan H, Xiao J, Daglia M. An overview of the health benefits of Prunus species with special reference to metabolic syndrome risk factors. Food Chem Toxicol 2020; 144:111574. [PMID: 32679287 DOI: 10.1016/j.fct.2020.111574] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 02/08/2023]
Abstract
Metabolic syndrome is a cluster of pathologies and conditions such as obesity, hyperglycemia, hyperlipidemia and hypertension representing a serious health concern in many countries due to its high rate of mortality and morbidity. Insulin resistance is known to play a central role in the development of metabolic syndrome and several risk factors, including visceral obesity, oxidative stress and chronic inflammation, could trigger insulin resistance. Different strategies are currently in practice to manage metabolic syndrome. Along with dietary components, botanicals contain secondary metabolites, which may play a pivotal role in the maintenance of health by combating chronic disorders. Genus Prunus is classified under family Rosaceae and consists of 400-430 species. This genus contains some important species of fruits and ornamental plants. Prunus species contain important micronutrients such as vitamins and minerals and their consumption could maintain health by nourishing the body with essential and non-essential compounds. Besides nutritional components, they also contain bioactive compounds such as polyphenols, which make them potential alternative therapeutic agents for a number of chronic disorders including dysregulated metabolic conditions. The present review is designed to highlight the evidence-based effects of Prunus species against metabolic syndrome risk factors.
Collapse
Affiliation(s)
- Hammad Ullah
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Anna De Filippis
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, 23200, Pakistan
| | - Jianbo Xiao
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, 212013, China
| | - Maria Daglia
- Department of Pharmacy, University of Naples Federico II, Naples, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
198
|
Sellin J, Fülle JB, Thiele C, Bauer R, Bülow MH. Free fatty acid determination as a tool for modeling metabolic diseases in Drosophila. JOURNAL OF INSECT PHYSIOLOGY 2020; 126:104090. [PMID: 32730782 DOI: 10.1016/j.jinsphys.2020.104090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 06/10/2020] [Accepted: 07/15/2020] [Indexed: 06/11/2023]
Abstract
Free or non-esterified fatty acids are the product of lipolysis of storage fat, i.e. triacylglyceroles. When the amount of fat exceeds the capacity of lipid-storing organs, free fatty acids affect and damage other non-lipid-storing organs. This process is termed lipotoxicity. Within a cell, free fatty acids can damage mitochondria, and lipotoxicity-induced mitochondrial damage has been associated recently with Peroxisomal Biogenesis Disorders. Drosophila melanogaster has a rising popularity as a model organism for metabolic diseases, but an optimized assay for measuring free fatty acids in Drosophila tissue samples is missing. Here we present a detailed protocol highlighting technical requirements and pitfalls to determine free fatty acids in samples of Drosophila tissue. The colorimetric assay allows the reproducible and cost-efficient measurement of free fatty acids in a 96 well plate format. We used our assay to determine changes in free fatty acid levels in different developmental stages and feeding conditions, and found that larvae and adults have different patterns of free fatty acid formation during starvation. Our assay is a valuable tool in the modeling of metabolic diseases with Drosophila melanogaster.
Collapse
Affiliation(s)
- Julia Sellin
- University of Bonn, Life & Medical Sciences Institute (LIMES), Molecular Developmental Biology, Carl-Troll-Straße 31, 53115 Bonn, Germany.
| | - Judith B Fülle
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK; Skin Research Institute of Singapore, A*STAR, 8A Biomedical Grove, Immunos #06-06, Singapore, Singapore
| | - Christoph Thiele
- University of Bonn, Life & Medical Sciences Institute (LIMES), Biochemistry & Cell Biology of Lipids, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Reinhard Bauer
- University of Bonn, Life & Medical Sciences Institute (LIMES), Molecular Developmental Biology, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Margret H Bülow
- University of Bonn, Life & Medical Sciences Institute (LIMES), Molecular Developmental Biology, Carl-Troll-Straße 31, 53115 Bonn, Germany.
| |
Collapse
|
199
|
Shabalala SC, Dludla PV, Mabasa L, Kappo AP, Basson AK, Pheiffer C, Johnson R. The effect of adiponectin in the pathogenesis of non-alcoholic fatty liver disease (NAFLD) and the potential role of polyphenols in the modulation of adiponectin signaling. Biomed Pharmacother 2020; 131:110785. [PMID: 33152943 DOI: 10.1016/j.biopha.2020.110785] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 02/08/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases worldwide, as it affects up to 30 % of adults in Western countries. Moreover, NAFLD is also considered an independent risk factor for cardiovascular diseases. Insulin resistance and inflammation have been identified as key factors in the pathophysiology of NAFLD. Although the mechanisms associated with the development of NAFLD remain to be fully elucidated, a complex interaction between adipokines and cytokines appear to play a crucial role in the development of this condition. Adiponectin is the most common adipokine known to be inversely linked with insulin resistance, lipid accumulation, inflammation and NAFLD. Consequently, the focus has been on the use of new therapies that may enhance hepatic expression of adiponectin downstream targets or increase the serum levels of adiponectin in the treatment NAFLD. While currently used therapies show limited efficacy in this aspect, accumulating evidence suggest that various dietary polyphenols may stimulate adiponectin levels, offering potential protection against the development of insulin resistance, inflammation and NAFLD as well as associated conditions of metabolic syndrome. As such, this review provides a better understanding of the role polyphenols play in modulating adiponectin signaling to protect against NAFLD. A brief discussion on the regulation of adiponectin during disease pathophysiology is also covered to underscore the potential protective effects of polyphenols against NAFLD. Some of the prominent polyphenols described in the manuscript include aspalathin, berberine, catechins, chlorogenic acid, curcumin, genistein, piperine, quercetin, and resveratrol.
Collapse
Affiliation(s)
- Samukelisiwe C Shabalala
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa; Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, KwaDlangezwa, 3886, South Africa
| | - Phiwayinkosi V Dludla
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa; Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, 60131, Italy
| | - Lawrence Mabasa
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa
| | - Abidemi P Kappo
- Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park, 2006, South Africa
| | - Albertus K Basson
- Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, KwaDlangezwa, 3886, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa; Department of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, 7505, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa; Department of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, 7505, South Africa.
| |
Collapse
|
200
|
Zhang T, Qin X, Cao Y, Zhang J, Zhao J. Sea buckthorn ( Hippophae rhamnoides L.) oil enhances proliferation, adipocytes differentiation and insulin sensitivity in 3T3-L1 cells. Food Sci Biotechnol 2020; 29:1511-1518. [PMID: 33088600 DOI: 10.1007/s10068-020-00817-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 08/15/2020] [Accepted: 08/24/2020] [Indexed: 12/23/2022] Open
Abstract
The objective of this study is to investigate the effects of sea buckthorn oil (SBO) on proliferation, adipogenic differentiation and insulin sensitivity of 3T3-L1 cells. Results showed that SBO increased cell proliferation ability, accompanied by up-regulated proliferating cell nuclear antigen content (p < 0.05) and p38 activity (p < 0.05). SBO also promoted adipogenesis and enhanced adipogenic transcriptional factors expression. Mitochondrial biogenesis related gene expressions were elevated in SBO treated cells (p < 0.05). Of note, SBO also increased glucose uptake and glucose transporter 4 abundance (p < 0.05). Cells treated with SBO exhibited greater phosphorylated insulin receptor substrate 1 (p < 0.05), phosphorylated-Akt (p < 0.05) and phosphorylated AMP-activated protein kinase (p < 0.01) contents. When taken together, these results suggest that SBO promotes 3T3-L1 cells proliferation, adipogenesis and insulin sensitivity.
Collapse
Affiliation(s)
- Ting Zhang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801 Shanxi People's Republic of China
| | - Xuze Qin
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801 Shanxi People's Republic of China
| | - Yuxin Cao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801 Shanxi People's Republic of China
| | - Jianxin Zhang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801 Shanxi People's Republic of China
| | - Junxing Zhao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801 Shanxi People's Republic of China
| |
Collapse
|