1
|
Jornayvaz FR, Gariani K, Somm E, Jaquet V, Bouzakri K, Szanto I. NADPH oxidases in healthy white adipose tissue and in obesity: function, regulation, and clinical implications. Obesity (Silver Spring) 2024; 32:1799-1811. [PMID: 39315402 DOI: 10.1002/oby.24113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/19/2024] [Accepted: 06/11/2024] [Indexed: 09/25/2024]
Abstract
Reactive oxygen species, when produced in a controlled manner, are physiological modulators of healthy white adipose tissue (WAT) expansion and metabolic function. By contrast, unbridled production of oxidants is associated with pathological WAT expansion and the establishment of metabolic dysfunctions, most notably insulin resistance and type 2 diabetes mellitus. NADPH oxidases (NOXs) produce oxidants in an orderly fashion and are present in adipocytes and in other diverse WAT-constituent cell types. Recent studies have established several links between aberrant NOX-derived oxidant production, adiposity, and metabolic homeostasis. The objective of this review is to highlight the physiological roles attributed to diverse NOX isoforms in healthy WAT and summarize current knowledge of the metabolic consequences related to perturbations in their adequate oxidant production. We detail WAT-related alterations in preclinical investigations conducted in NOX-deficient murine models. In addition, we review clinical studies that have employed NOX inhibitors and currently available data related to human NOX mutations in metabolic disturbances. Future investigations aimed at understanding the integration of NOX-derived oxidants in the regulation of the WAT cellular redox network are essential for designing successful redox-related precision therapies to curb obesity and attenuate obesity-associated metabolic pathologies.
Collapse
Affiliation(s)
- François R Jornayvaz
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Karim Gariani
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Emmanuel Somm
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Vincent Jaquet
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
- RE.A.D.S. Unit (Readers, Assay Development and Screening Unit), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Karim Bouzakri
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Ildiko Szanto
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
2
|
Hong C, Li X, Zhang K, Huang Q, Li B, Xin H, Hu B, Meng F, Zhu X, Tang D, Hu C, Tao C, Li J, Cao Y, Wang H, Deng B, Wang S. Novel perspectives on autophagy-oxidative stress-inflammation axis in the orchestration of adipogenesis. Front Endocrinol (Lausanne) 2024; 15:1404697. [PMID: 38982993 PMCID: PMC11232368 DOI: 10.3389/fendo.2024.1404697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/30/2024] [Indexed: 07/11/2024] Open
Abstract
Adipose tissue, an indispensable organ, fulfils the pivotal role of energy storage and metabolism and is instrumental in maintaining the dynamic equilibrium of energy and health of the organism. Adipocyte hypertrophy and adipocyte hyperplasia (adipogenesis) are the two primary mechanisms of fat deposition. Mature adipocytes are obtained by differentiating mesenchymal stem cells into preadipocytes and redifferentiation. However, the mechanisms orchestrating adipogenesis remain unclear. Autophagy, an alternative cell death pathway that sustains intracellular energy homeostasis through the degradation of cellular components, is implicated in regulating adipogenesis. Furthermore, adipose tissue functions as an endocrine organ, producing various cytokines, and certain inflammatory factors, in turn, modulate autophagy and adipogenesis. Additionally, autophagy influences intracellular redox homeostasis by regulating reactive oxygen species, which play pivotal roles in adipogenesis. There is a growing interest in exploring the involvement of autophagy, inflammation, and oxidative stress in adipogenesis. The present manuscript reviews the impact of autophagy, oxidative stress, and inflammation on the regulation of adipogenesis and, for the first time, discusses their interactions during adipogenesis. An integrated analysis of the role of autophagy, inflammation and oxidative stress will contribute to elucidating the mechanisms of adipogenesis and expediting the exploration of molecular targets for treating obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Chun Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Xinming Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Kunli Zhang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Qiuyan Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Baohong Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Haiyun Xin
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Bin Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Fanming Meng
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Xiangxing Zhu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Dongsheng Tang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Chuanhuo Hu
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Guangxi University, Nanning, China
| | - Chenyu Tao
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, Hebei, China
| | - Jianhao Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Yang Cao
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Hai Wang
- Chinese Academy of Sciences (CAS) Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health- Hong Kong University (GIBH-HKU) Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Bo Deng
- Division of Nephrology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sutian Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| |
Collapse
|
3
|
Herb M. NADPH Oxidase 3: Beyond the Inner Ear. Antioxidants (Basel) 2024; 13:219. [PMID: 38397817 PMCID: PMC10886416 DOI: 10.3390/antiox13020219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Reactive oxygen species (ROS) were formerly known as mere byproducts of metabolism with damaging effects on cellular structures. The discovery and description of NADPH oxidases (Nox) as a whole enzyme family that only produce this harmful group of molecules was surprising. After intensive research, seven Nox isoforms were discovered, described and extensively studied. Among them, the NADPH oxidase 3 is the perhaps most underrated Nox isoform, since it was firstly discovered in the inner ear. This stigma of Nox3 as "being only expressed in the inner ear" was also used by me several times. Therefore, the question arose whether this sentence is still valid or even usable. To this end, this review solely focuses on Nox3 and summarizes its discovery, the structural components, the activating and regulating factors, the expression in cells, tissues and organs, as well as the beneficial and detrimental effects of Nox3-mediated ROS production on body functions. Furthermore, the involvement of Nox3-derived ROS in diseases progression and, accordingly, as a potential target for disease treatment, will be discussed.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50935 Cologne, Germany;
- German Centre for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| |
Collapse
|
4
|
Engin A. Reappraisal of Adipose Tissue Inflammation in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:297-327. [PMID: 39287856 DOI: 10.1007/978-3-031-63657-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Chronic low-grade inflammation is a central component in the pathogenesis of obesity-related expansion of adipose tissue and complications in other metabolic tissues. Five different signaling pathways are defined as dominant determinants of adipose tissue inflammation: These are increased circulating endotoxin due to dysregulation in the microbiota-gut-brain axis, systemic oxidative stress, macrophage accumulation, and adipocyte death. Finally, the nucleotide-binding and oligomerization domain (NOD) leucine-rich repeat family pyrin domain-containing 3 (NLRP3) inflammasome pathway is noted to be a key regulator of metabolic inflammation. The NLRP3 inflammasome and associated metabolic inflammation play an important role in the relationships among fatty acids and obesity. Several highly active molecules, including primarily leptin, resistin, adiponectin, visfatin, and classical cytokines, are abundantly released from adipocytes. The most important cytokines that are released by inflammatory cells infiltrating obese adipose tissue are tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), monocyte chemoattractant protein 1 (MCP-1) (CCL-2), and IL-1. All these molecules mentioned above act on immune cells, causing local and then general inflammation. Three metabolic pathways are noteworthy in the development of adipose tissue inflammation: toll-like receptor 4 (TLR4)/phosphatidylinositol-3'-kinase (PI3K)/Protein kinase B (Akt) signaling pathway, endoplasmic reticulum (ER) stress-derived unfolded protein response (UPR), and inhibitor of nuclear factor kappa-B kinase beta (IKKβ)-nuclear factor kappa B (NF-κB) pathway. In fact, adipose tissue inflammation is an adaptive response that contributes to a visceral depot barrier that effectively filters gut-derived endotoxin. Excessive fatty acid release worsens adipose tissue inflammation and contributes to insulin resistance. However, suppression of adipose inflammation in obesity with anti-inflammatory drugs is not a rational solution and paradoxically promotes insulin resistance, despite beneficial effects on weight gain. Inflammatory pathways in adipocytes are indeed indispensable for maintaining systemic insulin sensitivity. Cannabinoid type 1 receptor (CB1R) is important in obesity-induced pro-inflammatory response; however, blockade of CB1R, contrary to anti-inflammatory drugs, breaks the links between insulin resistance and adipose tissue inflammation. Obesity, however, could be decreased by improving leptin signaling, white adipose tissue browning, gut microbiota interactions, and alleviating inflammation. Furthermore, capsaicin synthesized by chilies is thought to be a new and promising therapeutic option in obesity, as it prevents metabolic endotoxemia and systemic chronic low-grade inflammation caused by high-fat diet.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
5
|
Nakhlband A, Garjani A, Saeedi N, Omidi Y, Ghaffari S, Barar J, Eskandani M. Atherosclerosis preventive effects of marrubiin against (TNF-α)-induced oxidative stress and apoptosis. J Cardiovasc Thorac Res 2023; 15:174-180. [PMID: 38028719 PMCID: PMC10590460 DOI: 10.34172/jcvtr.2023.31704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 08/04/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Atherosclerosis is a complicated cascade of inflammatory processes, oxidative stress, and apoptosis, making it the most prevalent cardiovascular disease. The onset and progression of cardiovascular diseases are greatly influenced by oxidative stress. Targeting oxidative stress is an effective strategy for treating such diseases. Marrubiin is a bioactive furan labdane diterpenoid acts as a strong antioxidant to protect against oxidative damage. This study aimed to investigate the protective effects of marrubiin against oxidative stress and apoptosis in a cellular model of the vascular system. Methods Human umbilical vein endothelial cells were treated with varying concentration of marrubiin and its IC50 value was determined. The antioxidant potential of marrubiin was assessed by measuring the intracellular level of glutathione (GSH) using a colorimetric technique. Since apoptosis plays a significant role in the plaque rupture, the study also evaluated the protective effects of marrubiin on the expression of key genes involved in apoptotic pathways. Results Cells treated with marrubiin showed increased GSH levels compared to cell therapy control cells, indicating marrubiin's ability to counteract the effects of TNF-α's on GSH levels. Furthermore real-time PCR analysis demonstrated that marrubiin upregulated Bcl-xl while downregulating caspase3 and Nox4 in treated cells. These findings suggest that marrubiin protects against apoptosis and oxidative stress. Conclusion Based on our findings, marrubiin is recommended as a preventive/therapeutic treatment for diseases caused by elevated intracellular reactive oxygen species levels in cardiovascular diseases.
Collapse
Affiliation(s)
- Ailar Nakhlband
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Garjani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazli Saeedi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Armstrong HC, Russell DJF, Moss SEW, Pomeroy P, Bennett KA. Fitness correlates of blubber oxidative stress and cellular defences in grey seals (Halichoerus grypus): support for the life-history-oxidative stress theory from an animal model of simultaneous lactation and fasting. Cell Stress Chaperones 2023; 28:551-566. [PMID: 36933172 PMCID: PMC10469160 DOI: 10.1007/s12192-023-01332-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 03/19/2023] Open
Abstract
Life-history-oxidative stress theory predicts that elevated energy costs during reproduction reduce allocation to defences and increase cellular stress, with fitness consequences, particularly when resources are limited. As capital breeders, grey seals are a natural system in which to test this theory. We investigated oxidative damage (malondialdehyde (MDA) concentration) and cellular defences (relative mRNA abundance of heat shock proteins (Hsps) and redox enzymes (REs)) in blubber of wild female grey seals during the lactation fast (n = 17) and summer foraging (n = 13). Transcript abundance of Hsc70 increased, and Nox4, a pro-oxidant enzyme, decreased throughout lactation. Foraging females had higher mRNA abundance of some Hsps and lower RE transcript abundance and MDA concentrations, suggesting they experienced lower oxidative stress than lactating mothers, which diverted resources into pup rearing at the expense of blubber tissue damage. Lactation duration and maternal mass loss rate were both positively related to pup weaning mass. Pups whose mothers had higher blubber glutathione-S-transferase (GST) expression at early lactation gained mass more slowly. Higher glutathione peroxidase (GPx) and lower catalase (CAT) were associated with longer lactation but reduced maternal transfer efficiency and lower pup weaning mass. Cellular stress, and the ability to mount effective cellular defences, could proscribe lactation strategy in grey seal mothers and thus affect pup survival probability. These data support the life-history-oxidative stress hypothesis in a capital breeding mammal and suggest lactation is a period of heightened vulnerability to environmental factors that exacerbate cellular stress. Fitness consequences of stress may thus be accentuated during periods of rapid environmental change.
Collapse
Affiliation(s)
- Holly C Armstrong
- Marine Biology and Ecology Research Centre, Plymouth University, Drake Circus, Plymouth, PL4 8AA, UK.
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, KY16 9JP, UK.
| | - Debbie J F Russell
- Sea Mammal Research Unit, Scottish Oceans Institute, University of St Andrews, St Andrews, KY16 8LB, UK
| | - Simon E W Moss
- Sea Mammal Research Unit, Scottish Oceans Institute, University of St Andrews, St Andrews, KY16 8LB, UK
| | - Paddy Pomeroy
- Sea Mammal Research Unit, Scottish Oceans Institute, University of St Andrews, St Andrews, KY16 8LB, UK
| | - Kimberley A Bennett
- Division of Health Science, School of Applied Sciences, Abertay University, Dundee, DD1 1HG, UK
| |
Collapse
|
7
|
Hua H, Wu M, Wu T, Ji Y, Jin L, Du Y, Zhang Y, Huang S, Zhang A, Ding G, Liu Q, Jia Z. Reduction of NADPH oxidase 4 in adipocytes contributes to the anti-obesity effect of dihydroartemisinin. Heliyon 2023; 9:e14028. [PMID: 36915539 PMCID: PMC10006843 DOI: 10.1016/j.heliyon.2023.e14028] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 02/27/2023] Open
Abstract
Artemisinin derivatives have been found to have anti-obesity effects recently, but the mechanism is still controversial. Herein, long-term DHA treatment in obese mice significantly reduced the body weight and improved glucose metabolism. However, short-term DHA treatment did not affect glucose metabolism in obese mice, suggesting that the improved glucose metabolism in mice with DHA treatment could be secondary to body weight reduction. Consistent with previous reports, we observed that DHA inhibited the differentiation of adipocytes. Mechanistically, DHA significantly reduced the expression of NADPH oxidase 4 (NOX4) in white adipose tissue (WAT) of mice and differentiated adipocytes, and using NOX4 siRNA or the NOX4 inhibitor GKT137831 significantly attenuated adipocyte differentiation. Over-expression of NOX4 partially reversed the inhibition effect of DHA on adipogenic differentiation of preadipocytes. In addition, targeted proteomics analysis showed that DHA improved the abnormality of metabolic pathways. In conclusion, DHA significantly reduced fat mass and improved glucose metabolism in obese mice, possibly by inhibiting NOX4 expression to suppress adipocyte differentiation and lipid accumulation in adipocytes.
Collapse
Affiliation(s)
- Hu Hua
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Mengqiu Wu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Tong Wu
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Ji
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Lv Jin
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Du
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Guixia Ding
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qianqi Liu
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
miR-199a Downregulation as a Driver of the NOX4/HIF-1α/VEGF-A Pathway in Thyroid and Orbital Adipose Tissues from Graves′ Patients. Int J Mol Sci 2021; 23:ijms23010153. [PMID: 35008579 PMCID: PMC8745087 DOI: 10.3390/ijms23010153] [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: 11/02/2021] [Revised: 12/14/2021] [Accepted: 12/22/2021] [Indexed: 12/20/2022] Open
Abstract
Graves’ disease (GD) is an autoimmune thyroiditis often associated with Graves’ orbitopathy (GO). GD thyroid and GO orbital fat share high oxidative stress (OS) and hypervascularization. We investigated the metabolic pathways leading to OS and angiogenesis, aiming to further decipher the link between local and systemic GD manifestations. Plasma and thyroid samples were obtained from patients operated on for multinodular goiters (controls) or GD. Orbital fats were from GO or control patients. The NADPH-oxidase-4 (NOX4)/HIF-1α/VEGF-A signaling pathway was investigated by Western blotting and immunostaining. miR-199a family expression was evaluated following quantitative real-time PCR and/or in situ hybridization. In GD thyroids and GO orbital fats, NOX4 was upregulated and correlated with HIF-1α stabilization and VEGF-A overexpression. The biotin assay identified NOX4, HIF-1α and VEGF-A as direct targets of miR-199a-5p in cultured thyrocytes. Interestingly, GD thyroids, GD plasmas and GO orbital fats showed a downregulation of miR-199a-3p/-5p. Our results also highlighted an activation of STAT-3 signaling in GD thyroids and GO orbital fats, a transcription factor known to negatively regulate miR-199a expression. We identified NOX4/HIF-1α/VEGF-A as critical actors in GD and GO. STAT-3-dependent regulation of miR-199a is proposed as a common driver leading to these events in GD thyroids and GO orbital fats.
Collapse
|
9
|
Gire D, Acharya J, Malik S, Inamdar S, Ghaskadbi S. Molecular mechanism of anti-adipogenic effect of vitexin in differentiating hMSCs. Phytother Res 2021; 35:6462-6471. [PMID: 34612537 DOI: 10.1002/ptr.7300] [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: 05/12/2021] [Revised: 08/28/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022]
Abstract
In this study, we evaluated a detailed molecular mechanism of anti-adipogenic activity of vitexin, apigenin flavone glucoside, present in germinated fenugreek seeds, in differentiating human mesenchymal stem cells (hMSCs). The lipid content of differentiated adipocytes was estimated by ORO staining. Effect on mitotic clonal expansion was checked by cell cycle analysis. Expression of early and terminal adipocyte differentiation markers, anti- and pro-adipogenic transcription factors and signalling intermediates regulating them was evaluated at RNA and protein level. We found vitexin to be non-cytotoxic up to 20 μM at which intracellular lipid accumulation was significantly decreased. Cell cycle analysis suggested that vitexin does not affect mitotic clonal expansion. Expression of early and late differentiation markers, such as CEBPα, CEBPβ, PPARγ, FABP4, perilipin, adiponectin and Glut4 was significantly reduced in the presence of vitexin. Expression of KLF4 and KLF15, positive regulators of PPARγ, was decreased, whereas that of negative regulators, namely KLF2, GATA2, miR20a, miR27a, miR27b, miR128, miR130a, miR130b, miR182 and miR548 increased with vitexin treatment. This effect was mediated by the activation of the AMP-activated protein kinase (AMPK) pathway via the activation of LepR and additionally by inhibiting ROS. Thus, our results showed that vitexin regulates the expression of PPARγ and inhibits adipogenesis of hMSCs at an early stage of differentiation.
Collapse
Affiliation(s)
- Dhananjay Gire
- Department of Zoology, Savitribai Phule Pune University, Pune, India
| | - Jhankar Acharya
- Department of Zoology, Savitribai Phule Pune University, Pune, India
| | - Sajad Malik
- Department of Zoology, Savitribai Phule Pune University, Pune, India
| | - Shrirang Inamdar
- Department of Zoology, Savitribai Phule Pune University, Pune, India
| | - Saroj Ghaskadbi
- Department of Zoology, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
10
|
Ahn CB, Je JY. Anti-adipogenic peptides from ark shell protein hydrolysate: Purification, identification and anti-adipogenic effect. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
11
|
Contribution of Adipose Tissue Oxidative Stress to Obesity-Associated Diabetes Risk and Ethnic Differences: Focus on Women of African Ancestry. Antioxidants (Basel) 2021; 10:antiox10040622. [PMID: 33921645 PMCID: PMC8073769 DOI: 10.3390/antiox10040622] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 12/14/2022] Open
Abstract
Adipose tissue (AT) storage capacity is central in the maintenance of whole-body homeostasis, especially in obesity states. However, sustained nutrients overflow may dysregulate this function resulting in adipocytes hypertrophy, AT hypoxia, inflammation and oxidative stress. Systemic inflammation may also contribute to the disruption of AT redox equilibrium. AT and systemic oxidative stress have been involved in the development of obesity-associated insulin resistance (IR) and type 2 diabetes (T2D) through several mechanisms. Interestingly, fat accumulation, body fat distribution and the degree of how adiposity translates into cardio-metabolic diseases differ between ethnicities. Populations of African ancestry have a higher prevalence of obesity and higher T2D risk than populations of European ancestry, mainly driven by higher rates among African women. Considering the reported ethnic-specific differences in AT distribution and function and higher levels of systemic oxidative stress markers, oxidative stress is a potential contributor to the higher susceptibility for metabolic diseases in African women. This review summarizes existing evidence supporting this hypothesis while acknowledging a lack of data on AT oxidative stress in relation to IR in Africans, and the potential influence of other ethnicity-related modulators (e.g., genetic-environment interplay, socioeconomic factors) for consideration in future studies with different ethnicities.
Collapse
|
12
|
John CM, Arockiasamy S. 3,5-Dimethoxy-4-benzoic acid (syringic acid) a natural phenolic acid reduces reactive oxygen species in differentiated 3T3-L1 adipocytes. In Vitro Cell Dev Biol Anim 2021; 57:386-394. [PMID: 33772407 DOI: 10.1007/s11626-021-00549-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/12/2021] [Indexed: 01/11/2023]
Abstract
Preadipocytes under nutrient excess mature to lipid-laden adipocytes that are hotspots for generation of reactive oxygen species (ROS) imbalance and oxidative stress. Syringic acid (SA), a natural phenolic acid, was evaluated for its in vitro antioxidant and ROS modulation during in matured 3T3-L1 adipocytes. Following 10 d, the SA-treated adipocytes were evaluated for the levels of glutathione (GSH) and antioxidant enzymes such as superoxide dismutase (SOD) and catalase (CAT). The levels of peroxides in mature adipocytes were estimated using dichlorofluorescein (DCF) cleavage fluorescence. The level of NADPH oxidase 4 (NOX4) expression was also investigated following 10-d differentiation period. SA significantly improved the levels of GSH, SOD, and CAT in matured adipocytes. Reduction in ROS production levels was also witnessed by decrease in DCF cleavage. SA showed concentration-dependent inhibition of NOX4 by day 7 of adipogenesis when compared with differentiated and undifferentiated cells. Moreover, SA exhibited effective antioxidant and anti-radical scavenging activity. These results suggest that SA in addition to inhibiting adipogenesis can strongly reduce ROS stress in mature adipocytes by upregulating levels of intracellular antioxidants and decreasing levels of NOX4 in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Cordelia Mano John
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Sumathy Arockiasamy
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India.
| |
Collapse
|
13
|
Nani A, Murtaza B, Sayed Khan A, Khan NA, Hichami A. Antioxidant and Anti-Inflammatory Potential of Polyphenols Contained in Mediterranean Diet in Obesity: Molecular Mechanisms. Molecules 2021; 26:985. [PMID: 33673390 PMCID: PMC7918790 DOI: 10.3390/molecules26040985] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 01/02/2023] Open
Abstract
Nutrition transition can be defined as shifts in food habits, and it is characterized by high-fat (chiefly saturated animal fat), hypercaloric and salty food consumption at the expense of dietary fibers, minerals and vitamins. Western dietary patterns serve as a model for studying the impact of nutrition transition on civilization diseases, such as obesity, which is commonly associated with oxidative stress and inflammation. In fact, reactive oxygen species (ROS) overproduction can be associated with nuclear factor-κB (NF-κB)-mediated inflammation in obesity. NF-κB regulates gene expression of several oxidant-responsive adipokines including tumor necrosis factor-α (TNF-α). Moreover, AMP-activated protein kinase (AMPK), which plays a pivotal role in energy homeostasis and in modulation of metabolic inflammation, can be downregulated by IκB kinase (IKK)-dependent TNF-α activation. On the other hand, adherence to a Mediterranean-style diet is highly encouraged because of its healthy dietary pattern, which includes antioxidant nutraceuticals such as polyphenols. Indeed, hydroxycinnamic derivatives, quercetin, resveratrol, oleuropein and hydroxytyrosol, which are well known for their antioxidant and anti-inflammatory activities, exert anti-obesity proprieties. In this review, we highlight the impact of the most common polyphenols from Mediterranean foods on molecular mechanisms that mediate obesity-related oxidative stress and inflammation. Hence, we discuss the effects of these polyphenols on a number of signaling pathways. We note that Mediterranean diet (MedDiet) dietary polyphenols can de-regulate nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) and NF-κB-mediated oxidative stress, and metabolic inflammation. MedDiet polyphenols are also effective in upregulating downstream effectors of several proteins, chiefly AMPK.
Collapse
Affiliation(s)
- Abdelhafid Nani
- Laboratory of Saharan Natural Resources, African University Ahmed Draia, Adrar 01000, Algeria
- Physiologie de la Nutrition & Toxicologie, U1231 INSERM/Université de Bourgogne-Franche Comté (UBFC)/Agro-Sup, 21000 Dijon, France; (B.M.); (A.S.K.); (N.A.K.)
| | - Babar Murtaza
- Physiologie de la Nutrition & Toxicologie, U1231 INSERM/Université de Bourgogne-Franche Comté (UBFC)/Agro-Sup, 21000 Dijon, France; (B.M.); (A.S.K.); (N.A.K.)
| | - Amira Sayed Khan
- Physiologie de la Nutrition & Toxicologie, U1231 INSERM/Université de Bourgogne-Franche Comté (UBFC)/Agro-Sup, 21000 Dijon, France; (B.M.); (A.S.K.); (N.A.K.)
| | - Naim Akhtar Khan
- Physiologie de la Nutrition & Toxicologie, U1231 INSERM/Université de Bourgogne-Franche Comté (UBFC)/Agro-Sup, 21000 Dijon, France; (B.M.); (A.S.K.); (N.A.K.)
| | - Aziz Hichami
- Physiologie de la Nutrition & Toxicologie, U1231 INSERM/Université de Bourgogne-Franche Comté (UBFC)/Agro-Sup, 21000 Dijon, France; (B.M.); (A.S.K.); (N.A.K.)
| |
Collapse
|
14
|
Sharma A, Singh S, Ahmad S, Gulzar F, Schertzer JD, Tamrakar AK. NOD1 activation induces oxidative stress via NOX1/4 in adipocytes. Free Radic Biol Med 2021; 162:118-128. [PMID: 33279617 DOI: 10.1016/j.freeradbiomed.2020.11.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/11/2020] [Accepted: 11/26/2020] [Indexed: 12/20/2022]
Abstract
Activation of innate immune components promotes cell autonomous inflammation in adipocytes. Oxidative stress links pattern recognition receptor-mediated detection of inflammatory ligands and the immune response. Reactive oxygen species (ROS) may mediate the effect of nucleotide-binding oligomerization domain protein-1 (NOD1) activation on inflammation in adipocytes. Here, we define the potential role of NADPH oxidase (NOX)-derived ROS in NOD1-mediated inflammatory response in adipocytes. Differentiated 3T3-L1 adipocytes were treated with NOD1 activating ligand D-gamma-Glu-meso-diaminopimelic acid (iE-DAP) to evaluate the oxidative stress and contribution of NOX as source of intracellular ROS. NOD1 activation potently induced ROS generation in 3T3-L1 adipocytes. Of the NOX family members, expression of NOX1 and NOX4 was increased upon NOD1 activation, in a PKCδ-dependent manner. siRNA-mediated down-regulation of NOX1 or NOX4 inhibited NOD1-mediated ROS production and increased the expression of antioxidant defense enzyme catalase and superoxide dismutase (SOD). siRNA-mediated lowering of NOX1 or NOX4 also suppressed NOD1-mediated activation of JNK1/2 and NF-κB, and consequent activation of inflammatory response in 3T3-L1 adipocytes. In summary, our findings demonstrate that NOD1 activation provokes oxidative stress in adipocytes via NOX1/4 and that oxidative stress, at least in part, contributes to induction of inflammatory response. Defining the source of ROS after immune response engagement may lead to new therapeutic strategies for adipose tissue inflammation.
Collapse
Affiliation(s)
- Aditya Sharma
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Sushmita Singh
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shadab Ahmad
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Farah Gulzar
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences and Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main St. W., Hamilton, ON, L8N 3Z5, Canada
| | - Akhilesh K Tamrakar
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
15
|
The Crosstalk of Adipose-Derived Stem Cells (ADSC), Oxidative Stress, and Inflammation in Protective and Adaptive Responses. Int J Mol Sci 2020; 21:ijms21239262. [PMID: 33291664 PMCID: PMC7730805 DOI: 10.3390/ijms21239262] [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: 11/02/2020] [Revised: 12/01/2020] [Accepted: 12/01/2020] [Indexed: 02/08/2023] Open
Abstract
The potential use of stem cell-based therapies for the repair and regeneration of various tissues and organs is a major goal in repair medicine. Stem cells are classified by their potential to differentiate into functional cells. Compared with other sources, adipose-derived stem cells (ADSCs) have the advantage of being abundant and easy to obtain. ADSCs are considered to be tools for replacing, repairing, and regenerating dead or damaged cells. The capacity of ADSCs to maintain their properties depends on the balance of complex signals in their microenvironment. Their properties and the associated outcomes are in part regulated by reactive oxygen species, which mediate the oxidation-reduction state of cells as a secondary messenger. ADSC therapy has demonstrated beneficial effects, suggesting that secreted factors may provide protection. There is evidence that ADSCs secrete a number of cytokines, growth factors, and antioxidant factors into their microenvironment, thus regulating intracellular signaling pathways in neighboring cells. In this review, we introduce the roles of ADSCs in the protection of cells by modulating inflammation and immunity, and we develop their potential therapeutic properties.
Collapse
|
16
|
Kompella P, Vasquez KM. Obesity and cancer: A mechanistic overview of metabolic changes in obesity that impact genetic instability. Mol Carcinog 2019; 58:1531-1550. [PMID: 31168912 PMCID: PMC6692207 DOI: 10.1002/mc.23048] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/16/2022]
Abstract
Obesity, defined as a state of positive energy balance with a body mass index exceeding 30 kg/m2 in adults and 95th percentile in children, is an increasing global concern. Approximately one-third of the world's population is overweight or obese, and in the United States alone, obesity affects one in six children. Meta-analysis studies suggest that obesity increases the likelihood of developing several types of cancer, and with poorer outcomes, especially in children. The contribution of obesity to cancer risk requires a better understanding of the association between obesity-induced metabolic changes and its impact on genomic instability, which is a major driving force of tumorigenesis. In this review, we discuss how molecular changes during adipose tissue dysregulation can result in oxidative stress and subsequent DNA damage. This represents one of the many critical steps connecting obesity and cancer since oxidative DNA lesions can result in cancer-associated genetic instability. In addition, the by-products of the oxidative degradation of lipids (e.g., malondialdehyde, 4-hydroxynonenal, and acrolein), and gut microbiota-mediated secondary bile acid metabolites (e.g., deoxycholic acid and lithocholic acid), can function as genotoxic agents and tumor promoters. We also discuss how obesity can impact DNA repair efficiency, potentially contributing to cancer initiation and progression. Finally, we outline obesity-related epigenetic changes and identify the gaps in knowledge to be addressed for the development of better therapeutic strategies for the prevention and treatment of obesity-related cancers.
Collapse
Affiliation(s)
- Pallavi Kompella
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, 1400 Barbara Jordan Boulevard, Austin, TX 78723, USA
| | - Karen M. Vasquez
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, 1400 Barbara Jordan Boulevard, Austin, TX 78723, USA
| |
Collapse
|
17
|
Awadallah S, Hasan H, Attlee A, Raigangar V, Unnikannan H, Madkour M, Abraham MS, Rashid LM. Waist circumference is a major determinant of oxidative stress in subjects with and without metabolic syndrome. Diabetes Metab Syndr 2019; 13:2541-2547. [PMID: 31405674 DOI: 10.1016/j.dsx.2019.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 07/08/2019] [Indexed: 12/30/2022]
Abstract
AIM Oxidative stress (OS) plays a major role in pathogenic mechanisms associated with metabolic syndrome (Mets) yet the main component of Mets contributing most to OS is not well elucidated. Hence, the aim of this study was to investigate the oxidative-antioxidative status in Mets subjects and to determine the main predicting component of OS. METHODS Anthropometric measures, fasting blood glucose, lipid profile, antioxidant enzymes [catalase, superoxide dismutase (SOD) and glutathione peroxidase (GPx)], reduced glutathione (GSH), malondialdehyde (MDA) and protein carbonyl were assessed in 172 adult UAE residents. International Diabetes Federation criteria were used for Mets diagnosis. Mets Scores (0-5) were calculated and assigned per subject based on number of components. RESULTS Of all participants, 22.1% had Mets and 49.4% had large waist circumference (WC). Significant lower levels of catalase, SOD, GPx and GSH, and higher levels of MDA and protein carbonyl were observed in subjects with Mets. In addition, catalase, SOD, GPx, and GSH correlated negatively, while MDA and protein carbonyl correlated positively with almost all Mets components. Similar trend of correlations was noticed with Mets Scores. When adjusted for age and gender, linear regression analysis revealed that subjects with large WC demonstrated significantly lower levels of antioxidative enzymes and GSH, and higher levels of MDA and protein carbonyl. Consequently, WC emerged as the best predictor of OS. CONCLUSIONS The degree of OS is dependent on the Mets Scores, and WC contributes independently to increased OS among adults in UAE.
Collapse
Affiliation(s)
- Samir Awadallah
- College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
| | - Hayder Hasan
- College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Amita Attlee
- Nutrition and Health Department, College of Food and Agriculture, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Veena Raigangar
- College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Hema Unnikannan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohamed Madkour
- College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mini Sara Abraham
- College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Latifa M Rashid
- Head of Nutrition Section, Ministry of Health and Prevention, United Arab Emirates
| |
Collapse
|
18
|
H 2O 2 Metabolism in Normal Thyroid Cells and in Thyroid Tumorigenesis: Focus on NADPH Oxidases. Antioxidants (Basel) 2019; 8:antiox8050126. [PMID: 31083324 PMCID: PMC6563055 DOI: 10.3390/antiox8050126] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/18/2019] [Accepted: 04/24/2019] [Indexed: 12/23/2022] Open
Abstract
Thyroid hormone synthesis requires adequate hydrogen peroxide (H2O2) production that is utilized as an oxidative agent during the synthesis of thyroxin (T4) and triiodothyronine (T3). Thyroid H2O2 is generated by a member of the family of NADPH oxidase enzymes (NOX-es), termed dual oxidase 2 (DUOX2). NOX/DUOX enzymes produce reactive oxygen species (ROS) as their unique enzymatic activity in a timely and spatially regulated manner and therefore, are important regulators of diverse physiological processes. By contrast, dysfunctional NOX/DUOX-derived ROS production is associated with pathological conditions. Inappropriate DUOX2-generated H2O2 production results in thyroid hypofunction in rodent models. Recent studies also indicate that ROS improperly released by NOX4, another member of the NOX family, are involved in thyroid carcinogenesis. This review focuses on the current knowledge concerning the redox regulation of thyroid hormonogenesis and cancer development with a specific emphasis on the NOX and DUOX enzymes in these processes.
Collapse
|
19
|
Yuan Y, Xu F, Cao Y, Xu L, Yu C, Yang F, Zhang P, Wang L, Shen G, Wang J, Xu Y. Iron Accumulation Leads to Bone Loss by Inducing Mesenchymal Stem Cell Apoptosis Through the Activation of Caspase3. Biol Trace Elem Res 2019; 187:434-441. [PMID: 29948914 DOI: 10.1007/s12011-018-1388-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/14/2018] [Indexed: 12/18/2022]
Abstract
Osteoporosis (OP) is a disease associated with bone loss and microstructure degradation. Recent studies have shown that iron accumulation may be a risk factor for OP. Bone marrow mesenchymal stem cells (MSCs) are multipotent cells and precursors to osteoblasts. MSCs play an important role in OP. Therefore, we evaluated the correlation between MSCs and OP in an environment of iron accumulation. Serum P1NP was decreased in iron accumulation mice. Micro-CT revealed that iron accumulation decreased bone mineral density and spatial structural parameters. Iron accumulation inhibited MSC quantity in bone marrow. However, the iron chelator deferoxamine (DFO) rescued the suppression. Iron accumulation also changed the MSC cell cycle. Iron elevated MSC cell ROS level and NOX4 protein expression. MSC apoptosis was increased, and more caspase3 was cleaved after iron intervention. Our data suggests that iron accumulation inhibits MSC quantity and induces MSC apoptosis. Bone loss from iron accumulation may correlate with the inhibition of MSCs.
Collapse
Affiliation(s)
- Ye Yuan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Fei Xu
- Hematology Center of Cyrus Tang Medical Institute, Soochow University School of Medicine, Suzhou, 215123, China
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yan Cao
- Hematology Center of Cyrus Tang Medical Institute, Soochow University School of Medicine, Suzhou, 215123, China
| | - Li Xu
- Hematology Center of Cyrus Tang Medical Institute, Soochow University School of Medicine, Suzhou, 215123, China
| | - Chen Yu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Fan Yang
- Osteoporosis Institute of Soochow University, Suzhou, 215004, China
| | - Peng Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Liang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Guangsi Shen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Jianrong Wang
- Hematology Center of Cyrus Tang Medical Institute, Soochow University School of Medicine, Suzhou, 215123, China
| | - Youjia Xu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, 215004, China.
- Osteoporosis Institute of Soochow University, Suzhou, 215004, China.
| |
Collapse
|
20
|
Rharass T, Lucas S. High Glucose Level Impairs Human Mature Bone Marrow Adipocyte Function Through Increased ROS Production. Front Endocrinol (Lausanne) 2019; 10:607. [PMID: 31551934 PMCID: PMC6746912 DOI: 10.3389/fendo.2019.00607] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022] Open
Abstract
Bone marrow adipocytes (BMAds) accumulate in aging, menopause, and metabolic diseases such as Type 2 diabetes. These osteoporotic conditions are associated with oxidative stress and hyperglycemia which are both considered as critical factors underlying bone fragility. Glucose excess and reactive oxygen species (ROS) are known to favor adipogenesis over osteoblastogenesis. In this study, we investigated whether high glucose exposure could determine dysfunction of mature BMAds, specifically through ROS production. The effects of low (LG, 5 mM) or high glucose (HG, 25 mM) concentrations were examined using human bone mesenchymal stromal cells (hBMSCs) in the time course of differentiation, and, up to 21 days once adipocytes were mature. HG did not alter the adipocyte differentiation process of hBMSCs. Yet, after 21 days under HG exposure, PPARG, CEBPA, and adiponectin mRNA expressions were decreased. These alterations were also observed following adipogenic inducer withdrawal as well as in adipocytes fully differentiated in LG then cultured in HG for the last 11 days. Without inducers, HG condition also led to decreased leptin mRNA level. Importantly, intracellular and extracellular ROS concentrations measured using Amplex Red were significantly raised by 50% under HG exposure. This rise was observed once adipocytes ended differentiation and was reproduced within the different cell culture settings without any cytotoxicity. Among genes involved in ROS metabolism, the mRNA level of the H2O2 generating enzyme NOX4 was found upregulated in the presence of HG. Following cell separation, mature BMAds were shown to overproduce ROS and to display the gene alterations in contrast to non-lipid-laden cells. Finally, a non-lethal treatment with a pro-oxidant agent under LG condition reduces the mRNA levels of PPARG, adiponectin, and leptin as the HG condition does in the absence of inducers, and amplifies the effect of glucose excess on gene expression. HG concentration drives mature BMAds toward altered expression of the main adipokines and transcriptional factors. These perturbations are associated with a rise in ROS generation likely mediated through enhanced expression of NOX4. Mature BMAds are thus responsive to changes in glucose and ROS concentrations, which is relevant regarding with their phenotype and function in age- or metabolic disease-related osteoporosis.
Collapse
|
21
|
Issa N, Lachance G, Bellmann K, Laplante M, Stadler K, Marette A. Cytokines promote lipolysis in 3T3-L1 adipocytes through induction of NADPH oxidase 3 expression and superoxide production. J Lipid Res 2018; 59:2321-2328. [PMID: 30317185 DOI: 10.1194/jlr.m086504] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 10/12/2018] [Indexed: 01/14/2023] Open
Abstract
NADPH oxidase (NOX) enzymes are one of the major superoxide-generating systems in cells. NOX-generated superoxide has been suggested to promote insulin resistance in the liver. However, the role of NOX enzymes in mediating metabolic dysfunction in other insulin target tissues remains unclear. Here, we show that NOX3 expression is induced in differentiated 3T3-L1 adipocytes upon treatment with proinflammatory cytokines. Superoxide production increased concurrently with NOX3 protein expression in cytokine-treated adipocytes, which was inhibited by the NOX inhibitor diphenyleneiodonium (DPI). Treatment of adipocytes with cytokines increased lipolysis and decreased PPARγ activity. Interestingly, treatment with DPI blunted lipolysis activation by cytokines but failed to restore PPARγ activity. siRNA-mediated NOX3 downregulation also prevented cytokine-induced superoxide generation and lipolysis. In line with increasing lipolysis, cytokines increased the phosphorylation of hormone-sensitive lipase (HSL), which was reversed by treatment with DPI and silencing of NOX3 expression. We conclude that NOX3 is a cytokine-inducible superoxide-generating enzyme in adipocytes, which promotes lipolysis through increasing phosphorylation of HSL. This suggests a key role for NOX3-mediated superoxide production in the increased adipocyte lipolysis in inflammatory settings.
Collapse
Affiliation(s)
- Nahla Issa
- Department of Medicine, Cardiology Axis of the Quebec Heart and Lung Research Institute, Laval University, Quebec, QC, Canada
| | - Gabriel Lachance
- Department of Medicine, Cardiology Axis of the Quebec Heart and Lung Research Institute, Laval University, Quebec, QC, Canada
| | - Kerstin Bellmann
- Department of Medicine, Cardiology Axis of the Quebec Heart and Lung Research Institute, Laval University, Quebec, QC, Canada
| | - Mathieu Laplante
- Department of Medicine, Cardiology Axis of the Quebec Heart and Lung Research Institute, Laval University, Quebec, QC, Canada.,Cancer Research Center (CRC), Laval University, Quebec, QC, Canada
| | - Krisztian Stadler
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - André Marette
- Department of Medicine, Cardiology Axis of the Quebec Heart and Lung Research Institute, Laval University, Quebec, QC, Canada
| |
Collapse
|
22
|
Sadie-Van Gijsen H. Adipocyte biology: It is time to upgrade to a new model. J Cell Physiol 2018; 234:2399-2425. [PMID: 30192004 DOI: 10.1002/jcp.27266] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/25/2018] [Indexed: 12/15/2022]
Abstract
Globally, the obesity pandemic is profoundly affecting quality of life and economic productivity, but efforts to address this, especially on a pharmacological level, have generally proven unsuccessful to date, serving as a stark demonstration that our understanding of adipocyte biology and pathophysiology is incomplete. To deliver better insight into adipocyte function and obesity, we need improved adipocyte models with a high degree of fidelity in representing the in vivo state and with a diverse range of experimental applications. Adipocyte cell lines, especially 3T3-L1 cells, have been used extensively over many years, but these are limited in terms of relevance and versatility. In this review, I propose that primary adipose-derived stromal/stem cells (ASCs) present a superior model with which to study adipocyte biology ex vivo. In particular, ASCs afford us the opportunity to study adipocytes from different, functionally distinct, adipose depots and to investigate, by means of in vivo/ex vivo studies, the effects of many different physiological and pathophysiological factors, such as age, body weight, hormonal status, diet and nutraceuticals, as well as disease and pharmacological treatments, on the biology of adipocytes and their precursors. This study will give an overview of the characteristics of ASCs and published studies utilizing ASCs, to highlight the areas where our knowledge is lacking. More comprehensive studies in primary ASCs will contribute to an improved understanding of adipose tissue, in healthy and dysfunctional states, which will enhance our efforts to more successfully manage and treat obesity.
Collapse
Affiliation(s)
- Hanél Sadie-Van Gijsen
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa.,Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa
| |
Collapse
|
23
|
Moonen L, D'Haese PC, Vervaet BA. Epithelial Cell Cycle Behaviour in the Injured Kidney. Int J Mol Sci 2018; 19:E2038. [PMID: 30011818 PMCID: PMC6073451 DOI: 10.3390/ijms19072038] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 07/10/2018] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI), commonly caused by ischemia-reperfusion injury, has far-reaching health consequences. Despite the significant regenerative capacity of proximal tubular epithelium cells (PTCs), repair frequently fails, leading to the development of chronic kidney disease (CKD). In the last decade, it has been repeatedly demonstrated that dysregulation of the cell cycle can cause injured kidneys to progress to CKD. More precisely, severe AKI causes PTCs to arrest in the G1/S or G2/M phase of the cell cycle, leading to maladaptive repair and a fibrotic outcome. The mechanisms causing these arrests are far from known. The arrest might, at least partially, be attributed to DNA damage since activation of the DNA-damage response pathway leads to cell cycle arrest. Alternatively, cytokine signalling via nuclear factor kappa beta (NF-κβ) and p38-mitogen-activated protein kinase (p38-MAPK) pathways, and reactive oxygen species (ROS) can play a role independent of DNA damage. In addition, only a handful of cell cycle regulators (e.g., p53, p21) have been thoroughly studied during renal repair. Still, why and how PTCs decide to arrest their cell cycle and how this arrest can efficiently be overcome remain open and challenging questions. In this review we will discuss the evidence for cell cycle involvement during AKI and development of CKD together with putative therapeutic approaches.
Collapse
Affiliation(s)
- Lies Moonen
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Benjamin A Vervaet
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| |
Collapse
|
24
|
Kawamorita Y, Shiraishi T, Tamura Y, Kumagai T, Shibata S, Fujigaki Y, Hosoyamada M, Nakagawa T, Uchida S. Renoprotective effect of topiroxostat via antioxidant activity in puromycin aminonucleoside nephrosis rats. Physiol Rep 2018; 5:5/15/e13358. [PMID: 28774949 PMCID: PMC5555887 DOI: 10.14814/phy2.13358] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/19/2017] [Accepted: 06/25/2017] [Indexed: 02/07/2023] Open
Abstract
Topiroxostat is a novel inhibitor of xanthine oxidase, and is postulated to exert a renoprotective effect. Puromycin aminonucleoside nephrosis (PAN) is a rat model of minimal change nephrotic syndrome. In this study, we examined whether topiroxostat ameliorates the kidney injury in PAN rats that was induced by a single intraperitoneal injection of PA (100 mg/kg body weight). Rats were divided into four groups: control rats, PAN rats, control rats treated with topiroxostat (1.0 mg/kg/day), and PAN rats treated with topiroxostat. Topiroxostat significantly reduced the amount of uric acid in the kidney cortex, while serum UA concentration remained unaffected by this treatment. Urinary protein excretion decreased significantly on day 10 in PAN rats upon topiroxostat treatment. Podocyte injury in PAN rats, as indicated by the reduction in WT‐1‐positive cell numbers and podocin immunoreactivity and foot process effacement, was partially yet significantly alleviated with topiroxostat treatment. In the kidney cortex, the increase in oxidative stress markers such as nitrotyrosine and 8‐hydroxy‐2‐deoxyguanosine (8‐OHdG) and the enhanced expressions of xanthine oxidase and NADPH oxidase 4 (NOX4) in PAN rats were significantly ameliorated by topiroxostat. Using cultured podocytes NOX4 expression was upregulated by adding 12 mg/dL UA into the culture medium. These results suggest that topiroxostat ameliorates proteinuria and kidney injury in PAN rats by lowering oxidative stress and tissue UA concentration. The renoprotective effects of topiroxostat could be attributed to its potential to inhibit xanthine oxidase and NOX4 in concert with suppression of intracellular UA production.
Collapse
Affiliation(s)
- Yosuke Kawamorita
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takeshi Shiraishi
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan.,Support for Community Medicine Endowed Chair, Teikyo University School of Medicine, Tokyo, Japan
| | - Yoshifuru Tamura
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takanori Kumagai
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan.,Support for Community Medicine Endowed Chair, Teikyo University School of Medicine, Tokyo, Japan
| | - Shigeru Shibata
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Yoshihide Fujigaki
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Makoto Hosoyamada
- Department of Human Physiology and Pathology, Faculty of Pharma-Sciences, Teikyo University, Tokyo, Japan
| | - Takahiko Nakagawa
- Industry-Academia-Government Association Promotion Center, Nara Medical University, Nara, Japan
| | - Shunya Uchida
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
25
|
Schiavone S, Camerino GM, Mhillaj E, Zotti M, Colaianna M, De Giorgi A, Trotta A, Cantatore FP, Conte E, Bove M, Tucci P, Morgese MG, Trabace L. Visceral Fat Dysfunctions in the Rat Social Isolation Model of Psychosis. Front Pharmacol 2017; 8:787. [PMID: 29167640 PMCID: PMC5682313 DOI: 10.3389/fphar.2017.00787] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/17/2017] [Indexed: 01/18/2023] Open
Abstract
Medication with neuroleptics has been associated with adipose tissue dysfunctions and, in particular, with increased visceral fat amount. However, several studies suggested that antipsychotic treatment might not be the main responsible of fat mass accumulation, as this has been also described in not treated psychotic patients. One of the most used “drug-free” rodent models of psychosis is the social isolation rearing of young adult rats, which provides a non-pharmacologic method of inducing long-term alterations reminiscent of symptoms seen in psychotic patients. Recent data highlighted a crucial role of redox imbalance in adipose tissue dysfunctions, in terms of decreased antioxidant defense and increased reactive oxygen species (ROS). Here, we investigated possible oxidative stress-related biomolecular alterations associated with visceral fat increase in 7 week isolated rats. To this purpose, we quantified total and visceral fat amount by using dual-energy X-ray (DEXA) absorptiometry. On visceral fat, we analyzed the expression of specific ROS-producer genes (Nox1, Nox4, Hmox-1), antioxidant enzymes (Prdx1 and Ucp-1) and oxidative stress-induced damage markers (Cidea, Slc2a4, and Acacb). The impact of oxidative stress on beta3-adrenergic receptors (Adrb3), at both mRNA and protein level, was also assessed. We found that 7 weeks of social isolation induced an increase in total and visceral fat, associated with a decrease in Prdx1 (mRNA and protein) as well as Ucp-1 mRNA levels and an enhanced expression of Nox1 (mRNA and protein) and Hmox-1 mRNA. No differences were detected in Nox4 mRNA levels between grouped and isolated animals. Elevations in Cidea, Slc2a4, and Acacb expression in visceral fat of isolated animals accounted for oxidative stress-related damage in this tissue, further associated with a significant increase in Adrb3 mRNA and protein. Our results provide a novel understanding of the pathological link existing among psychosocial stress-induced psychosis, adipose tissue dysfunctions and redox imbalance, opening new therapeutic perspectives for the treatment of alterations in peripheral tissues associated with this mental disorder.
Collapse
Affiliation(s)
- Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Giulia M Camerino
- Department of Pharmacy and Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Emanuela Mhillaj
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| | - Margherita Zotti
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Marilena Colaianna
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Angelo De Giorgi
- Dual Diagnosis Unit, Azienda Sanitaria Locale della Provincia di Foggia, Foggia, Italy
| | - Antonello Trotta
- Rheumatology Unit, Foggia City Hospital "Ospedali Riuniti", Foggia, Italy
| | | | - Elena Conte
- Department of Pharmacy and Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Maria Bove
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria G Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
26
|
Brun J, Berthou F, Trajkovski M, Maechler P, Foti M, Bonnet N. Bone Regulates Browning and Energy Metabolism Through Mature Osteoblast/Osteocyte PPARγ Expression. Diabetes 2017; 66:2541-2554. [PMID: 28687706 DOI: 10.2337/db17-0116] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/29/2017] [Indexed: 11/13/2022]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a master regulator of energy metabolism. In bone, it is known to regulate osteoblast differentiation and osteoclast activity. Whether PPARγ expression in bone cells, particularly osteocytes, regulates energy metabolism remains unknown. Here, we show that mature osteoblast/osteocyte-specific ablation of PPARγ in mice (Ocy-PPARγ-/-) alters body composition with age, namely, to produce less fat and more lean mass, and enhances insulin sensitivity and energy expenditure compared with wild-type mice. In addition, Ocy-PPARγ-/- mice exhibit more bone density, structure, and strength by uncoupling bone formation from resorption. When challenged with a high-fat diet, Ocy-PPARγ-/- mice retain glycemic control, with increased browning of the adipose tissue, decreased gluconeogenesis, and less hepatic steatosis. Moreover, these metabolic effects, particularly an increase in fatty acid oxidation, cannot be explained by decarboxylated osteocalcin changes, suggesting existence of other osteokines that are under the control of PPARγ. We further identify bone morphogenetic protein 7 as one of them. Hence, osteocytes coregulate bone and glucose homeostasis through a PPARγ regulatory pathway, and its inhibition could be clinically relevant for the prevention of glucose metabolic disorders.
Collapse
Affiliation(s)
- Julia Brun
- Division of Bone Diseases, Department of Internal Medicine Specialties, Faculty of Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Flavien Berthou
- Department of Cell Physiology and Metabolism, University of Geneva, and Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Mirko Trajkovski
- Department of Cell Physiology and Metabolism, University of Geneva, and Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva, and Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Michanlegelo Foti
- Department of Cell Physiology and Metabolism, University of Geneva, and Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Nicolas Bonnet
- Division of Bone Diseases, Department of Internal Medicine Specialties, Faculty of Medicine, Geneva University Hospital, Geneva, Switzerland
| |
Collapse
|
27
|
Bundalo M, Djordjevic A, Bursac B, Zivkovic M, Koricanac G, Stanković A. Fructose-rich diet differently affects angiotensin II receptor content in the nucleus and a plasma membrane fraction of visceral adipose tissue. Appl Physiol Nutr Metab 2017; 42:1254-1263. [PMID: 28772089 DOI: 10.1139/apnm-2016-0725] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The adipose tissue renin-angiotensin system (RAS) is proposed to be a pathophysiological link between adipose tissue dysregulation and metabolic disorders induced by a fructose-rich diet (FRD). RAS can act intracellularly. We hypothesized that adipocyte nuclear membranes possess angiotensin receptor types 1 and 2 (AT1R and AT2R), which couple to nuclear signaling pathways and regulate oxidative gene expression under FRD conditions. We analyzed the effect of consumption of 10% fructose solution for 9 weeks on biochemical parameters, adipocyte morphology, and expression of AT1R, AT2R, AT1R-associated protein (ATRAP), NADPH oxidase 4 (NOX4), matrix metalloproteinase-9 (MMP-9), and manganese superoxide dismutase (MnSOD) in adipose tissue of Wistar rats. We detected AT1R and AT2R in the nuclear fraction. FRD reduced the level of angiotensin receptors in the nucleus, while increased AT1R and decreased AT2R levels were observed in the plasma membrane. FRD increased the ATRAP mRNA level and decreased MnSOD mRNA and protein levels. No significant differences were observed for MMP-9 and NOX4 mRNA levels. These findings coincided with hyperleptinemia, elevated blood pressure and triglycerides, and unchanged visceral adipose tissue mass and morphology in FRD rats. Besides providing evidence for nuclear localization of angiotensin receptors in visceral adipose tissue, this study demonstrates the different effects of FRD on AT1R expression in different cellular compartments. Elevated blood pressure and decreased antioxidant capacity in visceral fat of fructose-fed rats were accompanied by an increased AT1R level in the plasma membrane, while upregulation of ATRAP and a decrease of nuclear membrane AT1R suggest an increased capacity for attenuation of excessive AT1R signaling and visceral adiposity.
Collapse
Affiliation(s)
- Maja Bundalo
- a Laboratory for Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Ana Djordjevic
- b Department of Biochemistry, Institute for Biological Research "Siniša Stanković", University of Belgrade, 142 Despot Stefan Blvd., 11000 Belgrade, Serbia
| | - Biljana Bursac
- b Department of Biochemistry, Institute for Biological Research "Siniša Stanković", University of Belgrade, 142 Despot Stefan Blvd., 11000 Belgrade, Serbia
| | - Maja Zivkovic
- a Laboratory for Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Goran Koricanac
- c Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Aleksandra Stanković
- a Laboratory for Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| |
Collapse
|
28
|
Jankovic A, Korac A, Buzadzic B, Stancic A, Otasevic V, Ferdinandy P, Daiber A, Korac B. Targeting the NO/superoxide ratio in adipose tissue: relevance to obesity and diabetes management. Br J Pharmacol 2017; 174:1570-1590. [PMID: 27079449 PMCID: PMC5446578 DOI: 10.1111/bph.13498] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 12/21/2022] Open
Abstract
Insulin sensitivity and metabolic homeostasis depend on the capacity of adipose tissue to take up and utilize excess glucose and fatty acids. The key aspects that determine the fuel-buffering capacity of adipose tissue depend on the physiological levels of the small redox molecule, nitric oxide (NO). In addition to impairment of NO synthesis, excessive formation of the superoxide anion (О2•- ) in adipose tissue may be an important interfering factor diverting the signalling of NO and other reactive oxygen and nitrogen species in obesity, resulting in metabolic dysfunction of adipose tissue over time. Besides its role in relief from superoxide burst, enhanced NO signalling may be responsible for the therapeutic benefits of different superoxide dismutase mimetics, in obesity and experimental diabetes models. This review summarizes the role of NO in adipose tissue and highlights the effects of NO/О2•- ratio 'teetering' as a promising pharmacological target in the metabolic syndrome. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Aleksandra Jankovic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Aleksandra Korac
- Faculty of Biology, Center for Electron MicroscopyUniversity of BelgradeBelgradeSerbia
| | - Biljana Buzadzic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Ana Stancic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Vesna Otasevic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Péter Ferdinandy
- Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- Pharmahungary GroupSzegedHungary
| | - Andreas Daiber
- Center for Cardiology ‐ Cardiology 1, Molecular CardiologyUniversity Medical CenterMainzGermany
| | - Bato Korac
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| |
Collapse
|
29
|
Friederich-Persson M, Nguyen Dinh Cat A, Persson P, Montezano AC, Touyz RM. Brown Adipose Tissue Regulates Small Artery Function Through NADPH Oxidase 4–Derived Hydrogen Peroxide and Redox-Sensitive Protein Kinase G-1α. Arterioscler Thromb Vasc Biol 2017; 37:455-465. [DOI: 10.1161/atvbaha.116.308659] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 12/06/2016] [Indexed: 12/30/2022]
Abstract
Objective—
Biomedical interest in brown adipose tissue (BAT) has increased since the discovery of functionally active BAT in adult humans. Although white adipose tissue (WAT) influences vascular function, vascular effects of BAT are elusive. Thus, we investigated the regulatory role and putative vasoprotective effects of BAT, focusing on hydrogen peroxide, nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4), and redox-sensitive signaling.
Approach and Results—
Vascular reactivity was assessed in wild-type and Nox4-knockout mice (Nox4
−/−
) by wire myography in the absence and presence of perivascular adipose tissue of different phenotypes from various adipose depots: (1) mixed WAT/BAT (inguinal adipose tissue) and (2) WAT (epididymal visceral fat) and BAT (intrascapular fat). In wild-type mice, epididymal visceral fat and perivascular adipose tissue increased EC
50
to noradrenaline without affecting maximum contraction. BAT increased EC
50
and significantly decreased maximum contraction, which were prevented by a hydrogen peroxide scavenger (polyethylene glycated catalase) and a specific cyclic GMP–dependent protein kinase G type-1α inhibitor (DT-3), but not by inhibition of endothelial nitric oxide synthase or guanylate cyclase. BAT induced dimerization of cyclic GMP–dependent protein kinase G type-1α and reduced phosphorylation of myosin light chain phosphatase subunit 1 and myosin light chain 20. BAT from Nox4-knockout mice displayed reduced hydrogen peroxide levels and no anticontractile effects. Perivascular adipose tissue from β
3
agonist–treated mice displayed browned perivascular adipose tissue and an increased anticontractile effect.
Conclusions—
We identify a novel vasoprotective action of BAT through an anticontractile effect that is mechanistically different to WAT. Specifically, BAT, via Nox4-derived hydrogen peroxide, induces cyclic GMP–dependent protein kinase G type-1α activation, resulting in reduced vascular contractility. BAT may constitute an interesting therapeutic target to restore vascular function and prevent vascular complications in cardiovascular diseases.
Collapse
Affiliation(s)
- Malou Friederich-Persson
- From the Institute of Cardiovascular Medicine and Sciences, University of Glasgow, United Kingdom
| | - Aurelie Nguyen Dinh Cat
- From the Institute of Cardiovascular Medicine and Sciences, University of Glasgow, United Kingdom
| | - Patrik Persson
- From the Institute of Cardiovascular Medicine and Sciences, University of Glasgow, United Kingdom
| | - Augusto C. Montezano
- From the Institute of Cardiovascular Medicine and Sciences, University of Glasgow, United Kingdom
| | - Rhian M. Touyz
- From the Institute of Cardiovascular Medicine and Sciences, University of Glasgow, United Kingdom
| |
Collapse
|
30
|
The Pathogenesis of Obesity-Associated Adipose Tissue Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:221-245. [DOI: 10.1007/978-3-319-48382-5_9] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
31
|
de Villiers D, Potgieter M, Ambele MA, Adam L, Durandt C, Pepper MS. The Role of Reactive Oxygen Species in Adipogenic Differentiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1083:125-144. [PMID: 29139087 DOI: 10.1007/5584_2017_119] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Interest in reactive oxygen species and adipocyte differentiation/adipose tissue function is steadily increasing. This is due in part to a search for alternative avenues for combating obesity, which results from the excess accumulation of adipose tissue. Obesity is a major risk factor for complex disorders such as cancer, type 2 diabetes, and cardiovascular diseases. The ability of mesenchymal stromal/stem cells (MSCs) to differentiate into adipocytes is often used as a model for studying adipogenesis in vitro. A key focus is the effect of both intra- and extracellular reactive oxygen species (ROS) on adipogenesis. The consensus from the majority of studies is that ROS, irrespective of the source, promote adipogenesis.The effect of ROS on adipogenesis is suppressed by antioxidants or ROS scavengers. Reactive oxygen species are generated during the process of adipocyte differentiation as well as by other cell metabolic processes. Despite many studies in this field, it is still not possible to state with certainty whether ROS measured during adipocyte differentiation are a cause or consequence of this process. In addition, it is still unclear what the exact sources are of the ROS that initiate and/or drive adipogenic differentiation in MSCs in vivo. This review provides an overview of our understanding of the role of ROS in adipocyte differentiation as well as how certain ROS scavengers and antioxidants might affect this process.
Collapse
Affiliation(s)
- Danielle de Villiers
- Department of Immunology and Institute for Cellular and Molecular Medicine; SAMRC Extramural Unit for Stem Cell Research and Therapy; Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Marnie Potgieter
- Department of Immunology and Institute for Cellular and Molecular Medicine; SAMRC Extramural Unit for Stem Cell Research and Therapy; Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Center for Microbial Ecology and Genomics, Department of Genetics, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| | - Melvin A Ambele
- Department of Immunology and Institute for Cellular and Molecular Medicine; SAMRC Extramural Unit for Stem Cell Research and Therapy; Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Department of Oral Pathology and Oral Biology, School of Dentistry, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ladislaus Adam
- Department of Immunology and Institute for Cellular and Molecular Medicine; SAMRC Extramural Unit for Stem Cell Research and Therapy; Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Chrisna Durandt
- Department of Immunology and Institute for Cellular and Molecular Medicine; SAMRC Extramural Unit for Stem Cell Research and Therapy; Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Michael S Pepper
- Department of Immunology and Institute for Cellular and Molecular Medicine; SAMRC Extramural Unit for Stem Cell Research and Therapy; Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
32
|
Ribou AC. Synthetic Sensors for Reactive Oxygen Species Detection and Quantification: A Critical Review of Current Methods. Antioxid Redox Signal 2016; 25:520-33. [PMID: 27225539 DOI: 10.1089/ars.2016.6741] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
SIGNIFICANCE Redox reactions play important roles in both physiological and pathological processes, highlighting the importance of quantifying and localizing intracellular redox-active components. Most research has focused on direct investigation of reactive oxygen species (ROS). Intensity-based fluorescent methods are very sensitive and easy to use, but they lack specificity and can produce artifacts. In this article, we focus on synthetic sensors, describing experimental pitfalls associated with their use. We also present alternative methods for the detection of free radicals. RECENT ADVANCES New approaches have been developed to overcome the main artifact of intensity-based methods: spurious changes in fluorescence intensity caused by oxidation. These new approaches are based on analytical measurements of the oxidized sensors or techniques that are not susceptible to oxidation, such as electron spin resonance and fluorescence lifetime-based methods. Regardless of the approach, the need for detection of ROS on the subcellular level, especially in the mitochondria, has motivated the development of new probes. CRITICAL ISSUES Flow cytometry systems and confocal microscopes are now available to the majority of biologists, and commercially available probes are, therefore, more widely used. The fact that these new applications are cited in thousands of publications makes these sensors even more attractive. FUTURE DIRECTIONS The field of ROS detection by synthetic sensors continues to expand, bringing needed additional research to the development of robust techniques that are applicable both in vitro and in vivo. Antioxid. Redox Signal. 25, 520-533.
Collapse
Affiliation(s)
- Anne-Cécile Ribou
- Institute of Modeling and Analysis in Geo-Environmental and Health (IMAGES_ESPACE-DEV), University of Perpignan Via Domitia , Perpignan, France
| |
Collapse
|
33
|
Li Z, Sheng Y, Liu C, Li K, Huang X, Huang J, Xu K. Nox4 has a crucial role in uric acid‑induced oxidative stress and apoptosis in renal tubular cells. Mol Med Rep 2016; 13:4343-8. [PMID: 27052425 DOI: 10.3892/mmr.2016.5083] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 03/08/2016] [Indexed: 11/06/2022] Open
Abstract
The purpose of the present study was to evaluate the effects of uric acid in promoting tubular cell apoptosis and verify the role of nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4)‑induced oxidative stress in this process. HK‑2 cells were used as a human proximal tubular cell model and co‑cultured with various concentrations of uric acid with or without pre‑treatment with the Nox4 inhibitor diphenylene iodonium (DPI). The apoptotic rate and the amount of reactive oxygen species (ROS) were examined by flow cytometry. Furthermore, levels of Nox4, phosphorylated (p)‑P38, p‑extracellular signal‑regulated kinase (ERK), B‑cell lymphoma 2 (Bcl‑2) and Bcl‑2‑extra large (Bax) were detected by western blot analysis. The results showed that treatment with uric acid decreased HK‑2 cell viability and promoted apoptosis in a dose‑dependent manner. This was paralleled with an upregulation of Nox4 as well as ROS overproduction, which activated the phosphorylation of P38/ERK and caused an imbalance of Bax/Bcl‑2 in HK‑2 cells. Of note, inhibition of Nox4 with DPI prevented uric acid‑induced cell injury by suppressing ROS generation and P38/ERK activation. In conclusion, it was demonstrated that elevated uric acid promoted ROS‑induced tubular cell apoptosis by upregulating Nox4 expression. The present study therefore provided possible mechanisms and a potential therapeutic target of uric acid‑induced chronic kidney disease.
Collapse
Affiliation(s)
- Zhuohang Li
- Department of Urology, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yiyu Sheng
- Department of Urology, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Cheng Liu
- Department of Urology, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Kuiqing Li
- Department of Urology, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xin Huang
- Department of Urology, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jian Huang
- Department of Urology, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Kewei Xu
- Department of Urology, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
34
|
Stockler-Pinto MB, Saldanha JF, Yi D, Mafra D, Fouque D, Soulage CO. The uremic toxin indoxyl sulfate exacerbates reactive oxygen species production and inflammation in 3T3-L1 adipose cells. Free Radic Res 2016; 50:337-44. [PMID: 26617268 DOI: 10.3109/10715762.2015.1125996] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Inflammation and oxidative stress are common features of patients with chronic kidney disease (CKD) and many uremic solutes retained in these patients could be involved in these processes, among which protein-bound solutes such as indoxyl sulfate (IS). White adipose tissue recently gained attention as an important source of inflammation and oxidative stress. To examine the effect of IS on adipocytes, 3T3-L1 adipose cells were incubated with IS to mimic the conditions encountered in uremic patients. Incubation of adipose cells with IS increased reactive oxygen species production generated mainly through activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase since it was prevented by the NADPH oxidase inhibitor apocynin. Exposure to IS furthermore exacerbated the secretion of tumor necrosis factor-α and interleukin-6 by adipose cells. This inflammatory response was prevented by NADPH oxidase inhibition pinpointing the pivotal role of intracellular oxidative stress. IS induces adipocyte perturbation and promotes inflammatory state mainly through induction of oxidative stress. IS, a uremic toxin, accumulates in CKD patients could, therefore, be an important mediator of adipocyte dysfunction in these patients.
Collapse
Affiliation(s)
- Milena B Stockler-Pinto
- a Cardiovascular Sciences Graduate Program, Federal Fluminense University , Niterói , Brazil
| | - Juliana F Saldanha
- b Medical Sciences Graduate Program, Federal Fluminense University , Niterói , Brazil
| | - Dan Yi
- c Université De Lyon, INSA De Lyon, CarMeN, INSERM U1060 , Villeurbanne , France
| | - Denise Mafra
- a Cardiovascular Sciences Graduate Program, Federal Fluminense University , Niterói , Brazil ;,b Medical Sciences Graduate Program, Federal Fluminense University , Niterói , Brazil
| | - Denis Fouque
- c Université De Lyon, INSA De Lyon, CarMeN, INSERM U1060 , Villeurbanne , France ;,d Department of Nephrology , Centre Hopitalier Lyon Sud , Pierre-Bénite , France
| | - Christophe O Soulage
- c Université De Lyon, INSA De Lyon, CarMeN, INSERM U1060 , Villeurbanne , France
| |
Collapse
|
35
|
Koh EJ, Kim KJ, Choi J, Jeon HJ, Seo MJ, Lee BY. Ginsenoside Rg1 suppresses early stage of adipocyte development via activation of C/EBP homologous protein-10 in 3T3-L1 and attenuates fat accumulation in high fat diet-induced obese zebrafish. J Ginseng Res 2015; 41:23-30. [PMID: 28123318 PMCID: PMC5223064 DOI: 10.1016/j.jgr.2015.12.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/19/2015] [Accepted: 12/04/2015] [Indexed: 01/11/2023] Open
Abstract
Background Ginsenoside Rg1 is a class of steroid glycoside and triterpene saponin in Panax ginseng. Many studies suggest that Rg1 suppresses adipocyte differentiation in 3T3-L1. However, the detail molecular mechanism of Rg1 on adipogenesis in 3T3-L1 is still not fully understood. Methods 3T3-L1 preadipocyte was used to evaluate the effect of Rg1 on adipocyte development in the differentiation in a stage-dependent manner in vitro. Oil Red O staining and Nile red staining were conducted to measure intracellular lipid accumulation and superoxide production, respectively. We analyzed the protein expression using Western blot in vitro. The zebrafish model was used to investigate whether Rg1 suppresses the early stage of fat accumulation in vivo. Results Rg1 decreased lipid accumulation in early-stage differentiation of 3T3-L1 compared with intermediate and later stages of adipocyte differentiation. Rg1 dramatically increased CAAT/enhancer binding protein (C/EBP) homologous protein-10 (CHOP10) and subsequently reduced the C/EBPβ transcriptional activity that prohibited the initiation of adipogenic marker expression as well as triglyceride synthase. Rg1 decreased the expression of extracellular signal-regulated kinase 1/2 and glycogen synthase kinase 3β, which are also essential for stimulating the expression of CEBPβ. Rg1 also reduced reactive oxygen species production because of the downregulated protein level of nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) oxidase 4 (NOX4). While Rg1 increased the endogenous antioxidant enzymes, it also dramatically decreased the accumulation of lipid and triglyceride in high fat diet-induced obese zebrafish. Conclusion We demonstrated that Rg1 suppresses early-stage differentiation via the activation of CHOP10 and attenuates fat accumulation in vivo. These results indicate that Rg1 might have the potential to reduce body fat accumulation in the early stage of obesity.
Collapse
Affiliation(s)
- Eun-Jeong Koh
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam, Korea
| | - Kui-Jin Kim
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam, Korea
| | - Jia Choi
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam, Korea
| | - Hui Jeon Jeon
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam, Korea
| | - Min-Jung Seo
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam, Korea
| | - Boo-Yong Lee
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam, Korea
| |
Collapse
|
36
|
Wang X, Hai C. Redox modulation of adipocyte differentiation: hypothesis of "Redox Chain" and novel insights into intervention of adipogenesis and obesity. Free Radic Biol Med 2015; 89:99-125. [PMID: 26187871 DOI: 10.1016/j.freeradbiomed.2015.07.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/19/2015] [Accepted: 07/08/2015] [Indexed: 02/08/2023]
Abstract
In view of the global prevalence of obesity and obesity-associated disorders, it is important to clearly understand how adipose tissue forms. Accumulating data from various laboratories implicate that redox status is closely associated with energy metabolism. Thus, biochemical regulation of the redox system may be an attractive alternative for the treatment of obesity-related disorders. In this work, we will review the current data detailing the role of the redox system in adipocyte differentiation, as well as identifying areas for further research. The redox system affects adipogenic differentiation in an extensive way. We propose that there is a complex and interactive "redox chain," consisting of a "ROS-generating enzyme chain," "combined antioxidant chain," and "transcription factor chain," which contributes to fine-tune the regulation of ROS level and subsequent biological consequences. The roles of the redox system in adipocyte differentiation are paradoxical. The redox system exerts a "tridimensional" mechanism in the regulation of adipocyte differentiation, including transcriptional, epigenetic, and posttranslational modulations. We suggest that redoxomic techniques should be extensively applied to understand the biological effects of redox alterations in a more integrated way. A stable and standardized "redox index" is urgently needed for the evaluation of the general redox status. Therefore, more effort should be made to establish and maintain a general redox balance rather than to conduct simple prooxidant or antioxidant interventions, which have comprehensive implications.
Collapse
Affiliation(s)
- Xin Wang
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| | - Chunxu Hai
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
37
|
Jankovic A, Korac A, Buzadzic B, Otasevic V, Stancic A, Daiber A, Korac B. Redox implications in adipose tissue (dys)function--A new look at old acquaintances. Redox Biol 2015; 6:19-32. [PMID: 26177468 PMCID: PMC4511633 DOI: 10.1016/j.redox.2015.06.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 06/25/2015] [Accepted: 06/30/2015] [Indexed: 12/15/2022] Open
Abstract
Obesity is an energy balance disorder associated with dyslipidemia, insulin resistance and diabetes type 2, also summarized with the term metabolic syndrome or syndrome X. Increasing evidence points to “adipocyte dysfunction”, rather than fat mass accretion per se, as the key pathophysiological factor for metabolic complications in obesity. The dysfunctional fat tissue in obesity characterizes a failure to safely store metabolic substrates into existing hypertrophied adipocytes and/or into new preadipocytes recruited for differentiation. In this review we briefly summarize the potential of redox imbalance in fat tissue as an instigator of adipocyte dysfunction in obesity. We reveal the challenge of the adipose redox changes, insights in the regulation of healthy expansion of adipose tissue and its reduction, leading to glucose and lipids overflow. Adipose tissue (AT) buffers nutrient excess determining overall metabolic health. Redox insight in lipid storage and adipogenesis of AT is reviewed. Redox modulation of AT as therapeutic target in obesity/syndrome X is considered.
Collapse
Affiliation(s)
- Aleksandra Jankovic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Aleksandra Korac
- University of Belgrade, Faculty of Biology, Center for Electron Microscopy, Belgrade, Serbia
| | - Biljana Buzadzic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Vesna Otasevic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Ana Stancic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Andreas Daiber
- 2nd Medical Department, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Bato Korac
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia.
| |
Collapse
|
38
|
Ryu JM, Lee HJ, Jung YH, Lee KH, Kim DI, Kim JY, Ko SH, Choi GE, Chai II, Song EJ, Oh JY, Lee SJ, Han HJ. Regulation of Stem Cell Fate by ROS-mediated Alteration of Metabolism. Int J Stem Cells 2015; 8:24-35. [PMID: 26019752 PMCID: PMC4445707 DOI: 10.15283/ijsc.2015.8.1.24] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 04/14/2015] [Indexed: 02/06/2023] Open
Abstract
Stem cells have attracted much attention due to their distinct features that support infinite self-renewal and differentiation into the cellular derivatives of three lineages. Recent studies have suggested that many stem cells both embryonic and adult stem cells reside in a specialized niche defined by hypoxic condition. In this respect, distinguishing functional differences arising from the oxygen concentration is important in understanding the nature of stem cells and in controlling stem cell fate for therapeutic purposes. ROS act as cellular signaling molecules involved in the propagation of signaling and the translation of environmental cues into cellular responses to maintain cellular homeostasis, which is mediated by the coordination of various cellular processes, and to adapt cellular activity to available bioenergetic sources. Thus, in this review, we describe the physiological role of ROS in stem cell fate and its effect on the metabolic regulation of stem cells.
Collapse
Affiliation(s)
- Jung Min Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Ki Hoon Lee
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Dah Ihm Kim
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Jeong Yeon Kim
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - So Hee Ko
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Gee Euhn Choi
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Ing Ing Chai
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Eun Ju Song
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Ji Young Oh
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Sei-Jung Lee
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
39
|
Atashi F, Modarressi A, Pepper MS. The role of reactive oxygen species in mesenchymal stem cell adipogenic and osteogenic differentiation: a review. Stem Cells Dev 2015; 24:1150-63. [PMID: 25603196 PMCID: PMC4424969 DOI: 10.1089/scd.2014.0484] [Citation(s) in RCA: 454] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising candidates for tissue engineering and regenerative medicine. The multipotent stem cell component of MSC isolates is able to differentiate into derivatives of the mesodermal lineage including adipocytes, osteocytes, chondrocytes, and myocytes. Many common pathways have been described in the regulation of adipogenesis and osteogenesis. However, stimulation of osteogenesis appears to suppress adipogenesis and vice-versa. Increasing evidence implicates a tight regulation of these processes by reactive oxygen species (ROS). ROS are short-lived oxygen-containing molecules that display high chemical reactivity toward DNA, RNA, proteins, and lipids. Mitochondrial complexes I and III, and the NADPH oxidase isoform NOX4 are major sources of ROS production during MSC differentiation. ROS are thought to interact with several pathways that affect the transcription machinery required for MSC differentiation including the Wnt, Hedgehog, and FOXO signaling cascades. On the other hand, elevated levels of ROS, defined as oxidative stress, lead to arrest of the MSC cell cycle and apoptosis. Tightly regulated levels of ROS are therefore critical for MSC terminal differentiation, although the precise sources, localization, levels and the exact species of ROS implicated remain to be determined. This review provides a detailed overview of the influence of ROS on adipogenic and osteogenic differentiation in MSCs.
Collapse
Affiliation(s)
- Fatemeh Atashi
- 1 Department of Plastic, Reconstructive & Aesthetic Surgery, University Hospitals of Geneva , University of Geneva, Geneva, Switzerland
| | | | | |
Collapse
|
40
|
Ye ZW, Zhang J, Townsend DM, Tew KD. Oxidative stress, redox regulation and diseases of cellular differentiation. Biochim Biophys Acta Gen Subj 2014; 1850:1607-21. [PMID: 25445706 DOI: 10.1016/j.bbagen.2014.11.010] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 10/31/2014] [Accepted: 11/10/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Within cells, there is a narrow concentration threshold that governs whether reactive oxygen species (ROS) induce toxicity or act as second messengers. SCOPE OF REVIEW We discuss current understanding of how ROS arise, facilitate cell signaling, cause toxicities and disease related to abnormal cell differentiation and those (primarily) sulfur based pathways that provide nucleophilicity to offset these effects. PRIMARY CONCLUSIONS Cellular redox homeostasis mediates a plethora of cellular pathways that determine life and death events. For example, ROS intersect with GSH based enzyme pathways to influence cell differentiation, a process integral to normal hematopoiesis, but also affecting a number of diverse cell differentiation related human diseases. Recent attempts to manage such pathologies have focused on intervening in some of these pathways, with the consequence that differentiation therapy targeting redox homeostasis has provided a platform for drug discovery and development. GENERAL SIGNIFICANCE The balance between electrophilic oxidative stress and protective biomolecular nucleophiles predisposes the evolution of modern life forms. Imbalances of the two can produce aberrant redox homeostasis with resultant pathologies. Understanding the pathways involved provides opportunities to consider interventional strategies. This article is part of a Special Issue entitled Redox regulation of differentiation and de-differentiation.
Collapse
Affiliation(s)
- Zhi-Wei Ye
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President St., DD410, Charleston, SC 29425, USA
| | - Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President St., DD410, Charleston, SC 29425, USA
| | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, 274 Calhoun Street MSC 141, Charleston, SC 29425-1410, USA
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President St., DD410, Charleston, SC 29425, USA.
| |
Collapse
|
41
|
Abstract
SIGNIFICANCE Stem cells are characterized by the properties of self-renewal and the ability to differentiate into multiple cell types, and thus maintain tissue homeostasis. Reactive oxygen species (ROS) are a natural byproduct of aerobic metabolism and have roles in cell signaling. Regulation of ROS has a vital role in maintaining "stemness" and differentiation of the stem cells, as well as in progression of stem-cell-associated diseases. RECENT ADVANCES As of late, much research has been done on the adverse effects of ROS in stem cells. However, recently it has become apparent that in some cases redox status of the stem cell does have a role in maintaining its identity as such. Both pluripotent and multipotent stem cell types have been reported to possess enzymatic and nonenzymatic mechanisms for detoxification of ROS and to correct oxidative damage to the genome as well as the proteome. CRITICAL ISSUES Although context dependent and somewhat varied among different stem cell types, the correlation seems to exist between antioxidant defense level and stem cell fate change (i.e., proliferation, differentiation, and death). Changes in stem cell redox regulation may affect the pathogenesis of various human diseases. FUTURE DIRECTIONS Dissecting the defined roles of ROS in distinct stem cell types will greatly enhance their basic and translational applications. Here, we discuss the various roles of ROS in adult, embryonic, and induced pluripotent stem cells.
Collapse
Affiliation(s)
- Pooja Chaudhari
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | | | | |
Collapse
|
42
|
Murdolo G, Bartolini D, Tortoioli C, Piroddi M, Iuliano L, Galli F. Lipokines and oxysterols: novel adipose-derived lipid hormones linking adipose dysfunction and insulin resistance. Free Radic Biol Med 2013; 65:811-820. [PMID: 23954331 DOI: 10.1016/j.freeradbiomed.2013.08.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 08/07/2013] [Accepted: 08/08/2013] [Indexed: 02/07/2023]
Abstract
The expansion of adipose tissue (AT) is, by definition, a hallmark of obesity. However, not all increases in fat mass are associated with pathophysiological cues. Indeed, whereas a "healthy" fat mass accrual, mainly in the subcutaneous depots, preserves metabolic homeostasis, explaining the occurrence of the metabolically healthy obese phenotype, "unhealthy" AT expansion is importantly associated with insulin resistance/type 2 diabetes and the metabolic syndrome. The development of a dysfunctional adipose organ may find mechanistic explanation in a reduced ability to recruit new and functional (pre)adipocytes from undifferentiated precursor cells. Such a failure of the adipogenic process underlies the "AT expandability" paradigm. The inability of AT to expand further to store excess nutrients, rather than obesity per se, induces a diabetogenic milieu by promoting the overflow and the ectopic deposition of fatty acids in insulin-dependent organs (i.e., lipotoxicity), the secretion of various metabolically detrimental adipose-derived hormones (i.e., adipokines and lipokines), and the occurrence of local and systemic inflammation and oxidative stress. Hitherto, fatty acids (i.e., lipokines) and the oxidation by-products of cholesterol and polyunsaturated fatty acids, such as nonenzymatic oxysterols and reactive aldehyde species, respectively, emerge as key modulators of (pre)adipocyte signaling through Wnt/β-catenin and MAPK pathways and potential regulators of glucose homeostasis. These and other mechanistic insights linking adipose dysfunction, oxidative stress, and impairment of glucose homeostasis are discussed in this review article, which focuses on adipose peroxidation as a potential instigator of, and a putative therapeutic target for, obesity-associated metabolic dysfunctions.
Collapse
Affiliation(s)
- Giuseppe Murdolo
- Department of Internal Medicine, Assisi Hospital, I-06081 Assisi, Perugia, Italy; Section of Internal Medicine, Endocrine, and Metabolic Sciences, Italy.
| | - Desirée Bartolini
- Section of Applied Biochemistry and Nutritional Sciences, Department of Internal Medicine, Perugia University, Perugia, Italy
| | | | - Marta Piroddi
- Section of Applied Biochemistry and Nutritional Sciences, Department of Internal Medicine, Perugia University, Perugia, Italy
| | - Luigi Iuliano
- Unit of Vascular Medicine, Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Francesco Galli
- Section of Applied Biochemistry and Nutritional Sciences, Department of Internal Medicine, Perugia University, Perugia, Italy
| |
Collapse
|
43
|
Gratas-Delamarche A, Derbré F, Vincent S, Cillard J. Physical inactivity, insulin resistance, and the oxidative-inflammatory loop. Free Radic Res 2013; 48:93-108. [PMID: 24060092 DOI: 10.3109/10715762.2013.847528] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Epidemiological data indicate that physical inactivity, a main factor of global energetic imbalance, is involved in the worldwide epidemic of obesity and metabolic disorders such as insulin resistance. Although the complex pathogenesis of insulin resistance is not fully understood, literature data accumulated during the past decades clearly indicate that the activation of the oxidative-inflammatory loop plays a major role. By activating the oxidative-inflammatory loop in insulin-sensitive tissues, fat gain and adipose tissue dysfunction likely contribute to induce insulin resistance during chronic and prolonged physical inactivity. However, in the past years, evidence has emerged showing that early insulin resistance also occurs after very short-term exposure to physical inactivity (1-7 days) without any fat gain or energetic imbalance. The possible role of liver disturbances or endothelial dysfunction is suggested, but further studies are necessary to really conclude. Inactive skeletal muscle probably constitutes the primary triggering tissue for the development of early insulin resistance. In the present review, we discuss on the current knowledge about the effect of physical inactivity on whole-body and peripheral insulin sensitivity, and how local inflammation and oxidative stress arising with physical inactivity could potentially induce insulin resistance. We assume that early muscle insulin resistance allows the excess nutrients to shift in the storage tissues to withstand starvation through energy storage. We also consider when chronic and prolonged, physical inactivity over an extended period of time is an underestimated contributor to pathological insulin resistance and hence indirectly to numerous chronic diseases.
Collapse
Affiliation(s)
- A Gratas-Delamarche
- University Rennes 2 - ENS Cachan - Antenne de Bretagne, Laboratory "Movement, Sport and Health Sciences" (M2S) , Rennes , France
| | | | | | | |
Collapse
|
44
|
Sedeek M, Nasrallah R, Touyz RM, Hébert RL. NADPH oxidases, reactive oxygen species, and the kidney: friend and foe. J Am Soc Nephrol 2013; 24:1512-8. [PMID: 23970124 DOI: 10.1681/asn.2012111112] [Citation(s) in RCA: 348] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Reactive oxygen species (ROS) play an important role in normal cellular physiology. They regulate different biologic processes such as cell defense, hormone synthesis and signaling, activation of G protein-coupled receptors, and ion channels and kinases/phosphatases. ROS are also important regulators of transcription factors and gene expression. On the other hand, in pathologic conditions, a surplus of ROS in tissue results in oxidative stress with various injurious consequences such as inflammation and fibrosis. NADPH oxidases are one of the many sources of ROS in biologic systems, and there are seven isoforms (Nox1-5, Duox1, Duox2). Nox4 is the predominant form in the kidney, although Nox2 is also expressed. Nox4 has been implicated in the basal production of ROS in the kidney and in pathologic conditions such as diabetic nephropathy and CKD; upregulation of Nox4 may be important in renal oxidative stress and kidney injury. Although there is growing evidence indicating the involvement of NADPH oxidase in renal pathology, there is a paucity of information on the role of NADPH oxidase in the regulation of normal renal function. Here we provide an update on the role of NADPH oxidases and ROS in renal physiology and pathology.
Collapse
Affiliation(s)
- Mona Sedeek
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | |
Collapse
|
45
|
vinh quốc Lu'o'ng K, Nguyễn LTH. The beneficial role of vitamin D in obesity: possible genetic and cell signaling mechanisms. Nutr J 2013; 12:89. [PMID: 23800102 PMCID: PMC3702462 DOI: 10.1186/1475-2891-12-89] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 06/21/2013] [Indexed: 02/06/2023] Open
Abstract
The prevalence rates of overweight and obesity are considered an important public issue in the United States, and both of these conditions are increasing among both children and adults. There is evidence of aberrations in the vitamin D-endocrine system in obese subjects. Vitamin D deficiency is highly prevalent in patients with obesity, and many studies have demonstrated the significant effect of calcitriol on adipocytes. Genetic studies have provided an opportunity to determine which proteins link vitamin D to obesity pathology, including the vitamin D receptor, toll-like receptors, the renin-angiotensin system, apolipoprotein E, vascular endothelial growth factor, and poly (ADP-ribose) polymerase-1. Vitamin D also exerts its effect on obesity through cell-signaling mechanisms, including matrix metalloproteinases, mitogen-activated protein kinase pathways, the reduced form of nicotinamide adenine dinucleotide phosphate, prostaglandins, reactive oxygen species, and nitric oxide synthase. In conclusion, vitamin D may have a role in obesity. The best form of vitamin D for use in the obese individuals is calcitriol because it is the active form of the vitamin D3 metabolite, its receptors are present in adipocytes, and modulates inflammatory cytokine expression.
Collapse
Affiliation(s)
- Khanh vinh quốc Lu'o'ng
- Vietnamese American Medical Research Foundation, 14971 Brookhurst Street, Westminster, CA 92683, USA.
| | | |
Collapse
|
46
|
Murdolo G, Piroddi M, Luchetti F, Tortoioli C, Canonico B, Zerbinati C, Galli F, Iuliano L. Oxidative stress and lipid peroxidation by-products at the crossroad between adipose organ dysregulation and obesity-linked insulin resistance. Biochimie 2012; 95:585-94. [PMID: 23274128 DOI: 10.1016/j.biochi.2012.12.014] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 12/13/2012] [Indexed: 12/11/2022]
Abstract
Obesity has been proposed as an energy balance disorder in which the expansion of adipose tissue (AT) leads to unfavorable health outcomes. Even though adiposity represents the most powerful driving force for the development of insulin resistance (IR) and type 2 diabetes, mounting evidence points to "adipose dysregulation", rather than fat mass accrual per se, as a key pathophysiological trigger of the obesity-linked metabolic complications. The dysfunctional fat, besides hypertrophic adipose cells and inflammatory cues, displays a reduced ability to form new adipocytes from the undifferentiated precursor cells (ie, the preadipocytes). The failure of adipogenesis poses a "diabetogenic" milieu either by promoting the ectopic overflow/deposition of lipids in non-adipose targets (lipotoxicity) or by inducing a dysregulated secretion of different adipose-derived hormones (ie, adipokines and lipokines). This novel and provocative paradigm ("expandability hypothesis") further extends current "adipocentric view" implicating a reduced adipogenic capacity as a missing link between "unhealthy" fat expansion and impairment of metabolic homeostasis. Hitherto, reactive oxygen species have been implicated in multiple forms of IR. However, the effects of stress on adipogenesis remain controversial. Compelling circumstantial data indicate that lipid peroxidation by-products (ie, oxysterols and 4-hydrononenal) may detrimentally affect adipose homeostasis partly by impairing (pre)adipocyte differentiation. In this scenario, it is tempting to speculate that a fine tuning of the adipose redox status may provide new mechanistic insights at the interface between fat dysregulation and development of metabolic dysfunctions. Yet, in humans, the molecular "signatures" of oxidative stress in the dysregulated fat as well as the pathophysiological effects of adipose (per)oxidation on glucose homeostasis remain poorly investigated. In this review we will summarize the potential mechanisms by which increased oxidative stress in fat may impair (pre)adipocyte differentiation and promote the adipose dysfunction. We will also attempt to highlight the conundrum with the adipose redox changes and the regulation of glucose homeostasis. Finally, we will briefly discuss the scientific rationale for proposing the adipose redox state as a potential target for novel therapeutic strategies to curb/prevent adiposity-linked insulin resistance.
Collapse
Affiliation(s)
- Giuseppe Murdolo
- Department of Internal Medicine, Assisi Hospital, Via Valentin Muller 1, Assisi, Perugia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Liu GS, Chan EC, Higuchi M, Dusting GJ, Jiang F. Redox mechanisms in regulation of adipocyte differentiation: beyond a general stress response. Cells 2012; 1:976-93. [PMID: 24710538 PMCID: PMC3901142 DOI: 10.3390/cells1040976] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 10/26/2012] [Accepted: 10/31/2012] [Indexed: 02/07/2023] Open
Abstract
In this review, we summarize advances in our understanding of redox-sensitive mechanisms that regulate adipogenesis. Current evidence indicates that reactive oxygen species may act to promote both the initiation of adipocyte lineage commitment of precursor or stem cells, and the terminal differentiation of preadipocytes to mature adipose cells. These can involve redox regulation of pathways mediated by receptor tyrosine kinases, peroxisome proliferator-activated receptor γ (PPARγ), PPARγ coactivator 1α (PGC-1α), AMP-activated protein kinase (AMPK), and CCAAT/enhancer binding protein β (C/EBPβ). However, the precise roles of ROS in adipogenesis in vivo remain controversial. More studies are needed to delineate the roles of reactive oxygen species and redox signaling mechanisms, which could be either positive or negative, in the pathogenesis of obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Guei-Sheung Liu
- Centre for Eye Research Australia, University of Melbourne, Victoria 3002, Australia.
| | - Elsa C Chan
- Centre for Eye Research Australia, University of Melbourne, Victoria 3002, Australia.
| | - Masayoshi Higuchi
- Centre for Eye Research Australia, University of Melbourne, Victoria 3002, Australia.
| | - Gregory J Dusting
- Centre for Eye Research Australia, University of Melbourne, Victoria 3002, Australia.
| | - Fan Jiang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan 250-012, Shandong, China.
| |
Collapse
|
48
|
Altenhöfer S, Kleikers PWM, Radermacher KA, Scheurer P, Rob Hermans JJ, Schiffers P, Ho H, Wingler K, Schmidt HHHW. The NOX toolbox: validating the role of NADPH oxidases in physiology and disease. Cell Mol Life Sci 2012; 69:2327-43. [PMID: 22648375 PMCID: PMC3383958 DOI: 10.1007/s00018-012-1010-9] [Citation(s) in RCA: 287] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 04/18/2012] [Accepted: 04/20/2012] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are cellular signals but also disease triggers; their relative excess (oxidative stress) or shortage (reductive stress) compared to reducing equivalents are potentially deleterious. This may explain why antioxidants fail to combat diseases that correlate with oxidative stress. Instead, targeting of disease-relevant enzymatic ROS sources that leaves physiological ROS signaling unaffected may be more beneficial. NADPH oxidases are the only known enzyme family with the sole function to produce ROS. Of the catalytic NADPH oxidase subunits (NOX), NOX4 is the most widely distributed isoform. We provide here a critical review of the currently available experimental tools to assess the role of NOX and especially NOX4, i.e. knock-out mice, siRNAs, antibodies, and pharmacological inhibitors. We then focus on the characterization of the small molecule NADPH oxidase inhibitor, VAS2870, in vitro and in vivo, its specificity, selectivity, and possible mechanism of action. Finally, we discuss the validation of NOX4 as a potential therapeutic target for indications including stroke, heart failure, and fibrosis.
Collapse
Affiliation(s)
- Sebastian Altenhöfer
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Pamela W. M. Kleikers
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Kim A. Radermacher
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | | | - J. J. Rob Hermans
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Paul Schiffers
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Heidi Ho
- National Stroke Research Institute, Melbourne, VIC Australia
| | - Kirstin Wingler
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Harald H. H. W. Schmidt
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Vascular Drug Discovery Group, Faculty of Medicine, Health and Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
49
|
Lassègue B, San Martín A, Griendling KK. Biochemistry, physiology, and pathophysiology of NADPH oxidases in the cardiovascular system. Circ Res 2012; 110:1364-90. [PMID: 22581922 PMCID: PMC3365576 DOI: 10.1161/circresaha.111.243972] [Citation(s) in RCA: 610] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 03/09/2012] [Indexed: 02/07/2023]
Abstract
The NADPH oxidase (Nox) enzymes are critical mediators of cardiovascular physiology and pathophysiology. These proteins are expressed in virtually all cardiovascular cells, and regulate such diverse functions as differentiation, proliferation, apoptosis, senescence, inflammatory responses and oxygen sensing. They target a number of important signaling molecules, including kinases, phosphatases, transcription factors, ion channels, and proteins that regulate the cytoskeleton. Nox enzymes have been implicated in many different cardiovascular pathologies: atherosclerosis, hypertension, cardiac hypertrophy and remodeling, angiogenesis and collateral formation, stroke, and heart failure. In this review, we discuss in detail the biochemistry of Nox enzymes expressed in the cardiovascular system (Nox1, 2, 4, and 5), their roles in cardiovascular cell biology, and their contributions to disease development.
Collapse
Affiliation(s)
- Bernard Lassègue
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
50
|
Li Y, Mouche S, Sajic T, Veyrat-Durebex C, Supale R, Pierroz D, Ferrari S, Negro F, Hasler U, Feraille E, Moll S, Meda P, Deffert C, Montet X, Krause KH, Szanto I. Deficiency in the NADPH oxidase 4 predisposes towards diet-induced obesity. Int J Obes (Lond) 2012; 36:1503-13. [PMID: 22430302 DOI: 10.1038/ijo.2011.279] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE NADPH oxidase 4 (NOX4) is a reactive oxygen species (ROS) producing NADPH oxidase that regulates redox homeostasis in diverse insulin-sensitive cell types. In particular, NOX4-derived ROS is a key modulator of adipocyte differentiation and mediates insulin receptor signaling in mature adipocytes in vitro. Our study was aimed at investigating the role of NOX4 in adipose tissue differentiation, whole body metabolic homeostasis and insulin sensitivity in vivo. DESIGN Mice with genetic ablation of NOX4 (NOX4-deficient mice) were subjected to chow or high-fat-containing diet for 12 weeks. Body weight gain, adiposity, insulin sensitivity, and adipose tissue and liver gene and protein expression were analyzed and compared with similarly treated wild-type mice. RESULTS Here, we report that NOX4-deficient mice display latent adipose tissue accumulation and are susceptible to diet-induced obesity and early onset insulin resistance. Obesity results from accelerated adipocyte differentiation and hypertrophy, and an increase in whole body energy efficiency. Insulin resistance is associated with increased adipose tissue hypoxia, inflammation and adipocyte apoptosis. In the liver, more severe diet-induced steatosis was observed due to the lack of proper upregulation of mitochondrial fatty acid β-oxidation. CONCLUSION These findings identify NOX4 as a regulator of metabolic homeostasis. Moreover, they indicate an anti-adipogenic role for NOX4 in vivo and reveal its function as a protector against the development of diet-induced obesity, insulin resistance and hepatosteatosis.
Collapse
Affiliation(s)
- Y Li
- Department of Cellular Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|