1
|
Yang Y, Wang TT, Xie HA, Hu PP, Li P. Experimental cell models of insulin resistance: overview and appraisal. Front Endocrinol (Lausanne) 2024; 15:1469565. [PMID: 39749015 PMCID: PMC11693592 DOI: 10.3389/fendo.2024.1469565] [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: 07/25/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Insulin resistance, a key factor in the development of type 2 diabetes mellitus (T2DM), is defined as a defect in insulin-mediated control of glucose metabolism in tissues such as liver, fat and muscle. Insulin resistance is a driving force behind various metabolic diseases, such as T2DM, hyperlipidemia, hypertension, coronary heart disease and fatty liver. Therefore, improving insulin sensitivity can be considered as an effective strategy for the prevention and treatment of these complex metabolic diseases. Cell-based models are extensively employed for the study of pathological mechanisms and drug screening, particularly in relation to insulin resistance in T2DM. Currently, numerous methods are available for the establishment of in vitro insulin resistance models, a comprehensive review of these models is required and can serve as an excellent introduction or understanding for researchers undertaking studies in this filed. This review examines and discusses the primary methods for establishing and evaluating insulin resistance cell models. Furthermore, it highlights key issues and suggestions on cell selection, establishment, evaluation and drug screening of insulin resistance, thereby providing valuable references for the future research efforts.
Collapse
Affiliation(s)
- Ying Yang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Research Laboratory for Drug Metabolism, Chongqing Medical University, Chongqing, China
| | - Ting-ting Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Research Laboratory for Drug Metabolism, Chongqing Medical University, Chongqing, China
| | - Hu-ai Xie
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Research Laboratory for Drug Metabolism, Chongqing Medical University, Chongqing, China
| | - Ping Ping Hu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Research Laboratory for Drug Metabolism, Chongqing Medical University, Chongqing, China
| | - Pan Li
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Research Laboratory for Drug Metabolism, Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Terzo M, Iantomasi M, Tsiani E. Effects of Resveratrol on Adipocytes: Evidence from In Vitro and In Vivo Studies. Molecules 2024; 29:5359. [PMID: 39598748 PMCID: PMC11596734 DOI: 10.3390/molecules29225359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Obesity, a prevalent global health issue, arises from an imbalance between caloric intake and energy expenditure, leading to the expansion of adipose tissue and metabolic dysfunction. White adipose tissue (WAT) stores energy as lipids, while brown adipose tissue (BAT) plays a pivotal role in energy dissipation through adaptive thermogenesis. Recent research initiatives have focused on finding strategies to decrease adipogenesis and fat mass accumulation and increase thermogenesis. Finding chemicals with anti-obesity properties would be beneficial. Resveratrol, a polyphenolic compound abundantly found in the skin of grapes and red wine, possesses anti-oxidant, anti-inflammatory, anti-cancer, and anti-obesity properties. This literature review examines the effects of resveratrol on adipocytes in culture and adipose tissue in animal models of obesity. The existing evidence indicates that resveratrol may exert its anti-obesity effects by inhibiting adipogenesis, promoting the apoptosis of mature adipocytes, reducing lipid accumulation, and increasing thermogenesis. Further research utilizing animal and clinical studies is required to understand in detail the anti-obesity potential of resveratrol.
Collapse
Affiliation(s)
- Matthew Terzo
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Michael Iantomasi
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| |
Collapse
|
3
|
Wang F, Huynh PM, An YA. Mitochondrial Function and Dysfunction in White Adipocytes and Therapeutic Implications. Compr Physiol 2024; 14:5581-5640. [PMID: 39382163 DOI: 10.1002/cphy.c230009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
For a long time, white adipocytes were thought to function as lipid storages due to the sizeable unilocular lipid droplet that occupies most of their space. However, recent discoveries have highlighted the critical role of white adipocytes in maintaining energy homeostasis and contributing to obesity and related metabolic diseases. These physiological and pathological functions depend heavily on the mitochondria that reside in white adipocytes. This article aims to provide an up-to-date overview of the recent research on the function and dysfunction of white adipocyte mitochondria. After briefly summarizing the fundamental aspects of mitochondrial biology, the article describes the protective role of functional mitochondria in white adipocyte and white adipose tissue health and various roles of dysfunctional mitochondria in unhealthy white adipocytes and obesity. Finally, the article emphasizes the importance of enhancing mitochondrial quantity and quality as a therapeutic avenue to correct mitochondrial dysfunction, promote white adipocyte browning, and ultimately improve obesity and its associated metabolic diseases. © 2024 American Physiological Society. Compr Physiol 14:5581-5640, 2024.
Collapse
Affiliation(s)
- Fenfen Wang
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Phu M Huynh
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Department of Biochemistry and Molecular Biology, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
4
|
Li S, Xu R, Yao Y, Rousseau D. ATAD3 is a limiting factor in mitochondrial biogenesis and adipogenesis of white adipocyte-like 3T3-L1 cells. Cell Biol Int 2024; 48:1473-1489. [PMID: 38923254 DOI: 10.1002/cbin.12206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
ATAD3 is a vital ATPase of the inner mitochondrial membrane of pluri-cellular eukaryotes, with largely unknown functions but early required for organism development as necessary for mitochondrial biogenesis. ATAD3 knock-down in C. elegans inhibits at first the development of adipocyte-like intestinal tissue so we used mouse adipocyte model 3T3-L1 cells to analyze ATAD3 functions during adipogenesis and lipogenesis in a mammalian model. ATAD3 function was studied by stable and transient modulation of ATAD3 expression in adipogenesis- induced 3T3-L1 cells using Knock-Down and overexpression strategies, exploring different steps of adipocyte differentiation and lipogenesis. We show that (i) an increase in ATAD3 is preceding differentiation-induced mitochondrial biogenesis; (ii) downregulation of ATAD3 inhibits adipogenesis, lipogenesis, and impedes overexpression of many mitochondrial proteins; (iii) ATAD3 re-expression rescues the phenotype of ATAD3 KD, and (iv) differentiation and lipogenesis are accelerated by ATAD3 overexpression, but inhibited by expression of a dominant-negative mutant. We further show that the ATAD3 KD phenotype is not due to altered insulin signal but involves a limitation of mitochondrial biogenesis linked to Drp1. These results demonstrate that ATAD3 is limiting for in vitro mitochondrial biogenesis and adipogenesis/lipogenesis and therefore that ATAD3 mutation/over- or under-expression could be involved in adipogenic and lipogenic pathologies.
Collapse
Affiliation(s)
- Shuijie Li
- Department of Biology, University Grenoble Alpes, Grenoble, France
| | - Rui Xu
- Institute of Biochemistry and Cell Biology of Shanghai Institutes for Biological Sciences Chinese Academy of Sciences, Shanghai, China
| | - Yao Yao
- Institute of Biochemistry and Cell Biology of Shanghai Institutes for Biological Sciences Chinese Academy of Sciences, Shanghai, China
| | - Denis Rousseau
- Department of Biology, University Grenoble Alpes, Grenoble, France
- Laboratoire des Matériaux et du Génie Physique-Interfaces entre Matériaux et Matière Biologique -Institut National Polytechnique-Centre National de la Recherche Scientifique - Unité Mixte de Recherche, Grenoble, France
| |
Collapse
|
5
|
de Luxán-Delgado B, Potes Y, Rubio-González A, Solano JJ, Boga JA, Antuña E, Cachán-Vega C, Bermejo-Millo JC, Menéndez-Coto N, García-González C, Pereira GC, Caballero B, Coto-Montes A, Vega-Naredo I. Melatonin Alleviates Liver Mitochondrial Dysfunction in Leptin-Deficient Mice. Int J Mol Sci 2024; 25:8677. [PMID: 39201365 PMCID: PMC11354344 DOI: 10.3390/ijms25168677] [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: 07/23/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Despite efforts to elucidate the cellular adaptations induced by obesity, cellular bioenergetics is currently considered a crucial target. New strategies to delay the onset of the hazardous adaptations induced by obesity are needed. Therefore, we evaluated the effects of 4 weeks of melatonin treatment on mitochondrial function and lipid metabolism in the livers of leptin-deficient mice. Our results revealed that the absence of leptin increased lipid storage in the liver and induced significant mitochondrial alterations, which were ultimately responsible for defective ATP production and reactive oxygen species overproduction. Moreover, leptin deficiency promoted mitochondrial biogenesis, fusion, and outer membrane permeabilization. Melatonin treatment reduced the bioenergetic deficit found in ob/ob mice, alleviating some mitochondrial alterations in the electron transport chain machinery, biogenesis, dynamics, respiration, ATP production, and mitochondrial outer membrane permeabilization. Given the role of melatonin in maintaining mitochondrial homeostasis, it could be used as a therapeutic agent against adipogenic steatosis.
Collapse
Affiliation(s)
- Beatriz de Luxán-Delgado
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
| | - Yaiza Potes
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Adrian Rubio-González
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Juan José Solano
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Geriatrics Service, Monte Naranco Hospital, 33012 Oviedo, Spain
| | - José Antonio Boga
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Microbiology Department, Hospital Universitario Central de Asturias, Avenida de Roma s/n, 33011 Oviedo, Spain
| | - Eduardo Antuña
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Cristina Cachán-Vega
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Juan Carlos Bermejo-Millo
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Nerea Menéndez-Coto
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
| | - Claudia García-González
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
| | - Gonçalo C. Pereira
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK;
| | - Beatriz Caballero
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Ana Coto-Montes
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Ignacio Vega-Naredo
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| |
Collapse
|
6
|
Aldehoff AS, Karkossa I, Goerdeler C, Krieg L, Schor J, Engelmann B, Wabitsch M, Landgraf K, Hackermüller J, Körner A, Rolle-Kampczyk U, Schubert K, von Bergen M. Unveiling the dynamics of acetylation and phosphorylation in SGBS and 3T3-L1 adipogenesis. iScience 2024; 27:109711. [PMID: 38840842 PMCID: PMC11152682 DOI: 10.1016/j.isci.2024.109711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/11/2023] [Accepted: 04/06/2024] [Indexed: 06/07/2024] Open
Abstract
Obesity, characterized by enlarged and dysfunctional adipose tissue, is among today's most pressing global public health challenges with continuously increasing prevalence. Despite the importance of post-translational protein modifications (PTMs) in cellular signaling, knowledge of their impact on adipogenesis remains limited. Here, we studied the temporal dynamics of transcriptome, proteome, central carbon metabolites, and the acetyl- and phosphoproteome during adipogenesis using LC-MS/MS combined with PTM enrichment strategies on human (SGBS) and mouse (3T3-L1) adipocyte models. Both cell lines exhibited unique PTM profiles during adipogenesis, with acetylated proteins being enriched for central energy metabolism, while phosphorylated proteins related to insulin signaling and organization of cellular structures. As candidates with strong correlation to the adipogenesis timeline we identified CD44 and the acetylation sites FASN_K673 and IDH_K272. While results generally aligned between SGBS and 3T3-L1 cells, details appeared cell line specific. Our datasets on SGBS and 3T3-L1 adipogenesis dynamics are accessible for further mining.
Collapse
Affiliation(s)
- Alix Sarah Aldehoff
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Isabel Karkossa
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Cornelius Goerdeler
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Laura Krieg
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Jana Schor
- Department of Computational Biology and Chemistry, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Beatrice Engelmann
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, University Hospital for Children and Adolescents Ulm, Ulm, Germany
| | - Kathrin Landgraf
- University Hospital for Children and Adolescents, Center for Pediatric Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Jörg Hackermüller
- Department of Computational Biology and Chemistry, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
- Department of Computer Science, University of Leipzig, Leipzig, Germany
| | - Antje Körner
- University Hospital for Children and Adolescents, Center for Pediatric Research, Medical Faculty, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI-MAG) of the Helmholtz-Centre Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
- LIFE–Leipzig Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Kristin Schubert
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| |
Collapse
|
7
|
Abstract
Obesity is a severe health problem worldwide due to its association with various adverse health consequences. The present study aims to evaluate the anti-obesity effects of resveratrol, as a natural polyphenol, on the 3T3-L1 adipocytes. PubMed, Scopus, ScienceDirect, Web of Sciences, and Google Scholar databases were searched up to March 2022 using relevant keywords. All original articles, written in English, evaluating the anti-obesity effects of resveratrol on the 3T3-L1 adipocytes were eligible for this review. Initially, 4361 records were found in the electronic search databases. After removing duplicates and irrelevant studies according to the title and abstract, the full text of the 51 articles was critically screened and 38 in vitro studies were included in this review. Except for one case, all of these studies reported that different doses (ranged 1-200 μM) of resveratrol treatment have anti-obesity effects on 3T3L1 adipocytes through various mechanisms such as induction of apoptosis, a decrease of fat accumulation and adipogenesis, promotion of white adipocytes browning, inhibition of preadipocyte proliferation and consequent differentiation, and up-regulation of miRNA that involved in the antiadipogenic and triacylglycerol metabolism in white adipose tissue. The findings indicate that resveratrol has anti-obesity effects. Therefore, resveratrol treatment could be used to prevent and treat obesity and its related disorders. Well-designed randomized clinical trials with different doses of resveratrol are recommended to be performed on obese subjects.
Collapse
Affiliation(s)
- Roghayeh Molani-Gol
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Rafraf
- Nutrition Research Center, Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Flori L, Piragine E, Spezzini J, Citi V, Calderone V, Martelli A. Influence of Polyphenols on Adipose Tissue: Sirtuins as Pivotal Players in the Browning Process. Int J Mol Sci 2023; 24:ijms24119276. [PMID: 37298226 DOI: 10.3390/ijms24119276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Adipose tissue (AT) can be classified into two different types: (i) white adipose tissue (WAT), which represents the largest amount of total AT, and has the main function of storing fatty acids for energy needs and (ii) brown adipose tissue (BAT), rich in mitochondria and specialized in thermogenesis. Many exogenous stimuli, e.g., cold, exercise or pharmacological/nutraceutical tools, promote the phenotypic change of WAT to a beige phenotype (BeAT), with intermediate characteristics between BAT and WAT; this process is called "browning". The modulation of AT differentiation towards WAT or BAT, and the phenotypic switch to BeAT, seem to be crucial steps to limit weight gain. Polyphenols are emerging as compounds able to induce browning and thermogenesis processes, potentially via activation of sirtuins. SIRT1 (the most investigated sirtuin) activates a factor involved in mitochondrial biogenesis, peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), which, through peroxisome proliferator-activated receptor γ (PPAR-γ) modulation, induces typical genes of BAT and inhibits genes of WAT during the transdifferentiation process in white adipocytes. This review article aims to summarize the current evidence, from pre-clinical studies to clinical trials, on the ability of polyphenols to promote the browning process, with a specific focus on the potential role of sirtuins in the pharmacological/nutraceutical effects of natural compounds.
Collapse
Affiliation(s)
- Lorenzo Flori
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | - Jacopo Spezzini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Valentina Citi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Center "Nutrafood: Nutraceutica e Alimentazione per la Salute", University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Center "Biology and Pathology of Ageing", University of Pisa, 56126 Pisa, Italy
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Center "Nutrafood: Nutraceutica e Alimentazione per la Salute", University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Center "Biology and Pathology of Ageing", University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
9
|
Santos AL, Sinha S. Ageing, Metabolic Dysfunction, and the Therapeutic Role of Antioxidants. Subcell Biochem 2023; 103:341-435. [PMID: 37120475 DOI: 10.1007/978-3-031-26576-1_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
The gradual ageing of the world population has been accompanied by a dramatic increase in the prevalence of obesity and metabolic diseases, especially type 2 diabetes. The adipose tissue dysfunction associated with ageing and obesity shares many common physiological features, including increased oxidative stress and inflammation. Understanding the mechanisms responsible for adipose tissue dysfunction in obesity may help elucidate the processes that contribute to the metabolic disturbances that occur with ageing. This, in turn, may help identify therapeutic targets for the treatment of obesity and age-related metabolic disorders. Because oxidative stress plays a critical role in these pathological processes, antioxidant dietary interventions could be of therapeutic value for the prevention and/or treatment of age-related diseases and obesity and their complications. In this chapter, we review the molecular and cellular mechanisms by which obesity predisposes individuals to accelerated ageing. Additionally, we critically review the potential of antioxidant dietary interventions to counteract obesity and ageing.
Collapse
Affiliation(s)
- Ana L Santos
- IdISBA - Fundación de Investigación Sanitaria de las Islas Baleares, Palma, Spain.
| | | |
Collapse
|
10
|
Kurawaka M, Sasaki N, Yamazaki Y, Shimura F. Near-Physiological Concentrations of Extracellular Pyruvate Stimulated Glucose Utilization along with Triglyceride Accumulation and Mitochondrial Activity in HepG2 Cells. J Nutr Sci Vitaminol (Tokyo) 2023; 69:314-325. [PMID: 37940572 DOI: 10.3177/jnsv.69.314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Pyruvate, a key intermediate in energy and nutrient metabolism, probably plays important roles in these regulations. In previous reports using cell lines, extracellular pyruvate of supraphysiological concentrations inhibited the glucose uptake by myotubes while being stimulated by adipocytes. As the effect of pyruvate on the glucose utilization is unclear in cultured hepatocytes. We have investigated the effects of extracellular pyruvate on the glucose utilization and the subsequent metabolic changes using the cell line HepG2. In a 24 h culture, pyruvate enhanced the glucose consumption more potently than 1 μM insulin, and this enhancement was detectable at a near-physiological concentrations of ≤1 mM. For metabolic changes following glucose consumption, the conversion ratio of glucose and pyruvate to extracellular lactate was approximately 1.0 without extracellular pyruvate. The addition of pyruvate decreased the conversion ratio to approximately 0.7, indicating that the glycolytic reaction switched from being an anaerobic to a partially aerobic feature. Consistent with this finding, pyruvate increased the accumulation of intracellular triglycerides which are produced through substrate supply from the mitochondria. Furthermore, pyruvate stimulated mitochondria activity as evidenced by increases in ATP content, mitochondrial DNA copy number, enhanced mitochondria-specific functional imaging and oxygen consumption. Interestingly, 1 mM pyruvate increased oxygen consumption immediately after addition. In this study, we found that near-physiological concentrations of extracellular pyruvate exerted various changes in metabolic events, including glucose influx, lactate conversion rations, TG accumulation, and mitochondrial activity in HepG2 cells.
Collapse
Affiliation(s)
- Misaki Kurawaka
- Department of Food and Nutritional Sciences, Graduate School of Human Life Sciences, Jumonji University Graduate School
| | - Naho Sasaki
- Department of Health and Nutrition, Faculty of Human Life, Jumonji University
| | - Yuko Yamazaki
- Department of Food and Nutrition, Faculty of Human Life, Jumonji University
| | - Fumio Shimura
- Department of Food and Nutritional Sciences, Graduate School of Human Life Sciences, Jumonji University Graduate School
| |
Collapse
|
11
|
Khine HEE, Sungthong R, Sritularak B, Prompetchara E, Chaotham C. Untapped Pharmaceutical Potential of 4,5,4'-Trihydroxy-3,3'-dimethoxybibenzyl for Regulating Obesity: A Cell-Based Study with a Focus on Terminal Differentiation in Adipogenesis. JOURNAL OF NATURAL PRODUCTS 2022; 85:1591-1602. [PMID: 35679136 DOI: 10.1021/acs.jnatprod.2c00213] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Obesity and its global prevalence has become a threat to human health, while its pharmacotherapy via the application of natural products is still underdeveloped. Here, we probed how 4,5,4'-trihydroxy-3,3'-dimethoxybibenzyl (TDB) derived from an orchid (Dendrobium ellipsophyllum) could exert its roles on the differentiation and function of murine (3T3-L1) and human (PCS-210-010) pre-adipocytes and offer some implications to modulate obesity. Cytotoxic effects of TDB on adipocytes were 2-fold lower than those detected with pre-adipocytes, and no significant difference was detected in cytotoxic profiles between both cell lineages. TDB in a dose-dependent manner decreased cellular lipid accumulation and enhanced lipolysis of both cell lines assessed at early differentiation and during maturation. Underlining molecular mechanisms proved that TBD paused the cell cycle progression by regulating inducers and inhibitors in mitotic clonal expansion, leading to growth arrest of pre-adipocytes at the G0/G1 phase. The compound also governed adipocyte differentiation by repressing expressions of crucial adipogenic regulators and effectors through deactivating the AKT/GSK-3β signaling pathway and activating the AMPK-ACC pathway. To this end, TDB has shown its pharmaceutical potential for modulating adipocyte development and function, and it would be a promising candidate for further assessments as a therapeutic agent to defeat obesity.
Collapse
Affiliation(s)
- Hnin Ei Ei Khine
- Pharmaceutical Sciences and Technology Graduate Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Rungroch Sungthong
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, U.K
| | - Boonchoo Sritularak
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Eakachai Prompetchara
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chatchai Chaotham
- Pharmaceutical Sciences and Technology Graduate Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
12
|
Segrestin B, Delage P, Nemeth A, Seyssel K, Disse E, Nazare JA, Lambert-Porcheron S, Meiller L, Sauvinet V, Chanon S, Simon C, Ratiney H, Beuf O, Pralong F, Yassin NAH, Boizot A, Gachet M, Burton-Pimentel KJ, Vidal H, Meugnier E, Vionnet N, Laville M. Polyphenol Supplementation Did Not Affect Insulin Sensitivity and Fat Deposition During One-Month Overfeeding in Randomized Placebo-Controlled Trials in Men and in Women. Front Nutr 2022; 9:854255. [PMID: 35614978 PMCID: PMC9125251 DOI: 10.3389/fnut.2022.854255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/05/2022] [Indexed: 12/30/2022] Open
Abstract
Two randomized placebo-controlled double-blind paralleled trials (42 men in Lyon, 19 women in Lausanne) were designed to test 2 g/day of a grape polyphenol extract during 31 days of high calorie-high fructose overfeeding. Hyperinsulinemic-euglycemic clamps and test meals with [1,1,1-13C3]-triolein were performed before and at the end of the intervention. Changes in body composition were assessed by dual-energy X-ray absorptiometry (DEXA). Fat volumes of the abdominal region and liver fat content were determined in men only, using 3D-magnetic resonance imaging (MRI) and magnetic resonance spectroscopy (MRS) at 3T. Adipocyte's size was measured in subcutaneous fat biopsies. Bodyweight and fat mass increased during overfeeding, in men and in women. While whole body insulin sensitivity did not change, homeostasis model assessment of insulin resistance (HOMA-IR) and the hepatic insulin resistance index (HIR) increased during overfeeding. Liver fat increased in men. However, grape polyphenol supplementation did not modify the metabolic and anthropometric parameters or counteract the changes during overfeeding, neither in men nor in women. Polyphenol intake was associated with a reduction in adipocyte size in women femoral fat. Grape polyphenol supplementation did not counteract the moderated metabolic alterations induced by one month of high calorie-high fructose overfeeding in men and women. The clinical trials are registered under the numbers NCT02145780 and NCT02225457 at ClinicalTrials.gov and available at https://clinicaltrials.gov/ct2/show/NCT02145780 and https://clinicaltrials.gov/ct2/show/NCT02225457.
Collapse
Affiliation(s)
- Bérénice Segrestin
- INSERM, INRAe, CarMeN Laboratory, Claude Bernard Lyon 1 University, Lyon, France.,CRNH-RA, INSERM, INRAe, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Lyon, France.,Centre Hospitalier Lyon-Sud Service d'Endocrinologie Diabète Nutrition Lyon, Hospices Civils de Lyon, Lyon, France
| | - Pauline Delage
- INSERM, INRAe, CarMeN Laboratory, Claude Bernard Lyon 1 University, Lyon, France
| | - Angéline Nemeth
- CNRS, INSERM, CREATIS, Université de Lyon, INSA-Lyon, Claude Bernard Lyon 1 University, UJM-Saint Etienne, Lyon, France
| | - Kevin Seyssel
- INSERM, INRAe, CarMeN Laboratory, Claude Bernard Lyon 1 University, Lyon, France.,CRNH-RA, INSERM, INRAe, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Lyon, France
| | - Emmanuel Disse
- INSERM, INRAe, CarMeN Laboratory, Claude Bernard Lyon 1 University, Lyon, France.,CRNH-RA, INSERM, INRAe, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Lyon, France.,Centre Hospitalier Lyon-Sud Service d'Endocrinologie Diabète Nutrition Lyon, Hospices Civils de Lyon, Lyon, France
| | - Julie-Anne Nazare
- INSERM, INRAe, CarMeN Laboratory, Claude Bernard Lyon 1 University, Lyon, France.,CRNH-RA, INSERM, INRAe, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Lyon, France
| | | | - Laure Meiller
- CRNH-RA, INSERM, INRAe, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Lyon, France
| | - Valerie Sauvinet
- CRNH-RA, INSERM, INRAe, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Lyon, France
| | - Stéphanie Chanon
- INSERM, INRAe, CarMeN Laboratory, Claude Bernard Lyon 1 University, Lyon, France
| | - Chantal Simon
- INSERM, INRAe, CarMeN Laboratory, Claude Bernard Lyon 1 University, Lyon, France.,CRNH-RA, INSERM, INRAe, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Lyon, France
| | - Hélène Ratiney
- CNRS, INSERM, CREATIS, Université de Lyon, INSA-Lyon, Claude Bernard Lyon 1 University, UJM-Saint Etienne, Lyon, France
| | - Olivier Beuf
- CNRS, INSERM, CREATIS, Université de Lyon, INSA-Lyon, Claude Bernard Lyon 1 University, UJM-Saint Etienne, Lyon, France
| | - François Pralong
- Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Naba-Al-Huda Yassin
- Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Alexia Boizot
- Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Mélanie Gachet
- Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Kathryn J Burton-Pimentel
- Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Hubert Vidal
- INSERM, INRAe, CarMeN Laboratory, Claude Bernard Lyon 1 University, Lyon, France.,CRNH-RA, INSERM, INRAe, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Lyon, France
| | - Emmanuelle Meugnier
- INSERM, INRAe, CarMeN Laboratory, Claude Bernard Lyon 1 University, Lyon, France
| | - Nathalie Vionnet
- Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Martine Laville
- INSERM, INRAe, CarMeN Laboratory, Claude Bernard Lyon 1 University, Lyon, France.,CRNH-RA, INSERM, INRAe, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Lyon, France.,Centre Hospitalier Lyon-Sud Service d'Endocrinologie Diabète Nutrition Lyon, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
13
|
Sankaran H, Negi S, McShane LM, Zhao Y, Krushkal J. Pharmacogenomics of in vitro response of the NCI-60 cancer cell line panel to Indian natural products. BMC Cancer 2022; 22:512. [PMID: 35525914 PMCID: PMC9077913 DOI: 10.1186/s12885-022-09580-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/20/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Indian natural products have been anecdotally used for cancer treatment but with limited efficacy. To better understand their mechanism, we examined the publicly available data for the activity of Indian natural products in the NCI-60 cell line panel. METHODS We examined associations of molecular genomic features in the well-characterized NCI-60 cancer cell line panel with in vitro response to treatment with 75 compounds derived from Indian plant-based natural products. We analyzed expression measures for annotated transcripts, lncRNAs, and miRNAs, and protein-changing single nucleotide variants in cancer-related genes. We also examined the similarities between cancer cell line response to Indian natural products and response to reference anti-tumor compounds recorded in a U.S. National Cancer Institute (NCI) Developmental Therapeutics Program database. RESULTS Hierarchical clustering based on cell line response measures identified clustering of Phyllanthus and cucurbitacin products with known anti-tumor agents with anti-mitotic mechanisms of action. Curcumin and curcuminoids mostly clustered together. We found associations of response to Indian natural products with expression of multiple genes, notably including SLC7A11 involved in solute transport and ATAD3A and ATAD3B encoding mitochondrial ATPase proteins, as well as significant associations with functional single nucleotide variants, including BRAF V600E. CONCLUSION These findings suggest potential mechanisms of action and novel associations of in vitro response with gene expression and some cancer-related mutations that increase our understanding of these Indian natural products.
Collapse
Affiliation(s)
- Hari Sankaran
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD, 20850, USA.
| | - Simarjeet Negi
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD, 20850, USA
| | - Lisa M McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD, 20850, USA
| | - Yingdong Zhao
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD, 20850, USA
| | - Julia Krushkal
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD, 20850, USA.
| |
Collapse
|
14
|
Pereira AC, De Pascale J, Resende R, Cardoso S, Ferreira I, Neves BM, Carrascal MA, Zuzarte M, Madeira N, Morais S, Macedo A, do Carmo A, Moreira PI, Cruz MT, Pereira CF. ER-mitochondria communication is involved in NLRP3 inflammasome activation under stress conditions in the innate immune system. Cell Mol Life Sci 2022; 79:213. [PMID: 35344105 PMCID: PMC11072401 DOI: 10.1007/s00018-022-04211-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022]
Abstract
Endoplasmic reticulum (ER) stress and mitochondrial dysfunction, which are key events in the initiation and/or progression of several diseases, are correlated with alterations at ER-mitochondria contact sites, the so-called "Mitochondria-Associated Membranes" (MAMs). These intracellular structures are also implicated in NLRP3 inflammasome activation which is an important driver of sterile inflammation, however, the underlying molecular basis remains unclear. This work aimed to investigate the role of ER-mitochondria communication during ER stress-induced NLRP3 inflammasome activation in both peripheral and central innate immune systems, by using THP-1 human monocytes and BV2 microglia cells, respectively, as in vitro models. Markers of ER stress, mitochondrial dynamics and mass, as well as NLRP3 inflammasome activation were evaluated by Western Blot, IL-1β secretion was measured by ELISA, and ER-mitochondria contacts were quantified by transmission electron microscopy. Mitochondrial Ca2+ uptake and polarization were analyzed with fluorescent probes, and measurement of aconitase and SOD2 activities monitored mitochondrial ROS accumulation. ER stress was demonstrated to activate the NLRP3 inflammasome in both peripheral and central immune cells. Studies in monocytes indicate that ER stress-induced NLRP3 inflammasome activation occurs by a Ca2+-dependent and ROS-independent mechanism, which is coupled with upregulation of MAMs-resident chaperones, closer ER-mitochondria contacts, as well as mitochondrial depolarization and impaired dynamics. Moreover, enhanced ER stress-induced NLRP3 inflammasome activation in the immune system was found associated with pathological conditions since it was observed in monocytes derived from bipolar disorder (BD) patients, supporting a pro-inflammatory status in BD. In conclusion, by demonstrating that ER-mitochondria communication plays a key role in the response of the innate immune cells to ER stress, this work contributes to elucidate the molecular mechanisms underlying NLRP3 inflammasome activation under stress conditions, and to disclose novel potential therapeutic targets for diseases associated with sterile inflammation.
Collapse
Affiliation(s)
- Ana Catarina Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Jessica De Pascale
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
| | - Rosa Resende
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Susana Cardoso
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Isabel Ferreira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University Coimbra, Coimbra, Portugal
| | - Bruno Miguel Neves
- iBiMED-Department of Medical Sciences and Institute for Biomedicine, University Aveiro, Aveiro, Portugal
| | - Mylène A Carrascal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Tecnimede Group, Sintra, Portugal
| | - Mónica Zuzarte
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- iCBR-Institute for Clinical and Biomedical Research, University Coimbra, Coimbra, Portugal
| | - Nuno Madeira
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- CIBIT-Coimbra Institute for Biomedical Imaging and Translational Research, University Coimbra, Coimbra, Portugal
- Department of Psychiatry, CHUC-UC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Sofia Morais
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Department of Psychiatry, CHUC-UC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - António Macedo
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Department of Psychiatry, CHUC-UC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Anália do Carmo
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Department of Clinical Pathology, CHUC-UC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Maria Teresa Cruz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University Coimbra, Coimbra, Portugal
| | - Cláudia F Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal.
- Faculty of Medicine, University Coimbra, Coimbra, Portugal.
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal.
- , Coimbra, Portugal.
| |
Collapse
|
15
|
Borah AK, Sharma P, Singh A, Kalita KJ, Saha S, Chandra Borah J. Adipose and non-adipose perspectives of plant derived natural compounds for mitigation of obesity. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114410. [PMID: 34273447 DOI: 10.1016/j.jep.2021.114410] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/07/2021] [Accepted: 07/10/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Phyto-preparations and phyto-compounds, by their natural origin, easy availability, cost-effectiveness, and fruitful traditional uses based on accumulated experiences, have been extensively explored to mitigate the global burden of obesity. AIM OF THIS REVIEW The review aimed to analyse and critically summarize the prospect of future anti-obesity drug leads from the extant array of phytochemicals for mitigation of obesity, using adipose related targets (adipocyte formation, lipid metabolism, and thermogenesis) and non-adipose targets (hepatic lipid metabolism, appetite, satiety, and pancreatic lipase activity). Phytochemicals as inhibitors of adipocyte differentiation, modulators of lipid metabolism, and thermogenic activators of adipocytes are specifically discussed with their non-adipose anti-obesogenic targets. MATERIALS AND METHODS PubMed, Google Scholar, Scopus, and SciFinder were accessed to collect data on traditional medicinal plants, compounds derived from plants, their reported anti-obesity mechanisms, and therapeutic targets. The taxonomically accepted name of each plant in this review has been vetted from "The Plant List" (www.theplantlist.org) or MPNS (http://mpns.kew.org). RESULTS Available knowledge of a large number of phytochemicals, across a range of adipose and non-adipose targets, has been critically analysed and delineated by graphical and tabular depictions, towards mitigation of obesity. Neuro-endocrinal modulation in non-adipose targets brought into sharp dual focus, both non-adipose and adipose targets as the future of anti-obesity research. Numerous phytochemicals (Berberine, Xanthohumol, Ursolic acid, Guggulsterone, Tannic acid, etc.) have been found to be effectively reducing weight through lowered adipocyte formation, increased lipolysis, decreased lipogenesis, and enhanced thermogenesis. They have been affirmed as potential anti-obesity drugs of future because of their effectiveness yet having no threat to adipose or systemic insulin sensitivity. CONCLUSION Due to high molecular diversity and a greater ratio of benefit to risk, plant derived compounds hold high therapeutic potential to tackle obesity and associated risks. This review has been able to generate fresh perspectives on the anti-diabetic/anti-hyperglycemic/anti-obesity effect of phytochemicals. It has also brought into the focus that many phytochemicals demonstrating in vitro anti-obesogenic effects are yet to undergo in vivo investigation which could lead to potential phyto-molecules for dedicated anti-obesity action.
Collapse
Affiliation(s)
- Anuj Kumar Borah
- Dept. of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, 784028, Assam, India
| | - Pranamika Sharma
- Laboratory of Chemical Biology, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India
| | - Archana Singh
- Dept. of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, 784028, Assam, India
| | - Kangkan Jyoti Kalita
- Laboratory of Chemical Biology, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India
| | - Sougata Saha
- Dept. of Biotechnology, NIT Durgapur, West Bengal, 713209, India
| | - Jagat Chandra Borah
- Laboratory of Chemical Biology, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India.
| |
Collapse
|
16
|
Rabbani M, Pezeshki A, Ahmadi R, Mohammadi M, Tabibiazar M, Ahmadzadeh Nobari Azar F, Ghorbani M. Phytosomal nanocarriers for encapsulation and delivery of resveratrol- Preparation, characterization, and application in mayonnaise. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.112093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
17
|
Zhou S, Riadh D, Sakamoto K. Grape Extract Promoted α-MSH-Induced Melanogenesis in B16F10 Melanoma Cells, Which Was Inverse to Resveratrol. Molecules 2021; 26:molecules26195959. [PMID: 34641503 PMCID: PMC8512250 DOI: 10.3390/molecules26195959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
Melanin is a natural pigment produced by cells to prevent damage caused by ultraviolet radiation. Previously, resveratrol was shown to reduce melanin synthesis. As a natural polyphenol with various biological activities, resveratrol occurs in a variety of beverages and plant foods, such as grapes. Therefore, we investigated whether grape extracts containing resveratrol also had the ability to regulate melanin synthesis. In this study, we used mouse B16F10 melanoma cells as a model for melanin synthesis with the melanogenesis-inducing α-melanocyte-stimulating hormone (α-MSH) as a positive control. Our results confirmed previous reports that resveratrol reduces melanin synthesis by reducing the activity of the rate-limiting enzyme tyrosinase. In contrast, the grape extract could not reduce melanin synthesis, and in fact promoted melanogenesis in the presence of α-MSH. The expression of genes related to melanin synthesis, such as tyrosinase, tyrosinase-related protein-1, tyrosinase-related protein-2, and microphthalmia-associated transcription factor, also supports these phenomena, which means that even in the presence of resveratrol, grape extract will strengthen the function of α-MSH in promoting melanin synthesis. Therefore, these results also provide a point of view for research on cosmetics.
Collapse
|
18
|
Benbouguerra N, Hornedo-Ortega R, Garcia F, El Khawand T, Saucier C, Richard T. Stilbenes in grape berries and wine and their potential role as anti-obesity agents: A review. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.03.060] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
19
|
Jhanji M, Rao CN, Sajish M. Towards resolving the enigma of the dichotomy of resveratrol: cis- and trans-resveratrol have opposite effects on TyrRS-regulated PARP1 activation. GeroScience 2021; 43:1171-1200. [PMID: 33244652 PMCID: PMC7690980 DOI: 10.1007/s11357-020-00295-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
Unlike widely perceived, resveratrol (RSV) decreased the average lifespan and extended only the replicative lifespan in yeast. Similarly, although not widely discussed, RSV is also known to evoke neurite degeneration, kidney toxicity, atherosclerosis, premature senescence, and genotoxicity through yet unknown mechanisms. Nevertheless, in vivo animal models of diseases and human clinical trials demonstrate inconsistent protective and beneficial effects. Therefore, the mechanism of action of RSV that elicits beneficial effects remains an enigma. In a previously published work, we demonstrated structural similarities between RSV and tyrosine amino acid. RSV acts as a tyrosine antagonist and competes with it to bind to human tyrosyl-tRNA synthetase (TyrRS). Interestingly, although both isomers of RSV bind to TyrRS, only the cis-isomer evokes a unique structural change at the active site to promote its interaction with poly-ADP-ribose polymerase 1 (PARP1), a major determinant of cellular NAD+-dependent stress response. However, retention of trans-RSV in the active site of TyrRS mimics its tyrosine-bound conformation that inhibits the auto-poly-ADP-ribos(PAR)ylation of PARP1. Therefore, we proposed that cis-RSV-induced TyrRS-regulated auto-PARylation of PARP1 would contribute, at least in part, to the reported health benefits of RSV through the induction of protective stress response. This observation suggested that trans-RSV would inhibit TyrRS/PARP1-mediated protective stress response and would instead elicit an opposite effect compared to cis-RSV. Interestingly, most recent studies also confirmed the conversion of trans-RSV and its metabolites to cis-RSV in the physiological context. Therefore, the finding that cis-RSV and trans-RSV induce two distinct conformations of TyrRS with opposite effects on the auto-PARylation of PARP1 provides a potential molecular basis for the observed dichotomic effects of RSV under different experimental paradigms. However, the fact that natural RSV exists as a diastereomeric mixture of its cis and trans isomers and cis-RSV is also a physiologically relevant isoform has not yet gained much scientific attention.
Collapse
Affiliation(s)
- Megha Jhanji
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Chintada Nageswara Rao
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Mathew Sajish
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA.
| |
Collapse
|
20
|
Reguero M, Gómez de Cedrón M, Wagner S, Reglero G, Quintela JC, Ramírez de Molina A. Precision Nutrition to Activate Thermogenesis as a Complementary Approach to Target Obesity and Associated-Metabolic-Disorders. Cancers (Basel) 2021; 13:cancers13040866. [PMID: 33670730 PMCID: PMC7922953 DOI: 10.3390/cancers13040866] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Regarding the pandemic of obesity and chronic diseases associated to metabolic alterations that occur nowadays worldwide, here, we review the most recent studies related to bioactive compounds and diet derived ingredients with potential effects to augment the systemic energy expenditure. We specifically focus in two processes: the activation of thermogenesis in adipose tissue and the enhancement of the mitochondrial oxidative phosphorylation capacity in muscles. This may provide relevant information to develop diets and supplements to conduct nutritional intervention studies with the objective to ameliorate the metabolic and chronic inflammation in the course of obesity and related disorders. Abstract Obesity is associated to increased incidence and poorer prognosis in multiple cancers, contributing to up to 20% of cancer related deaths. These associations are mainly driven by metabolic and inflammatory changes in the adipose tissue during obesity, which disrupt the physiologic metabolic homeostasis. The association between obesity and hypercholesterolemia, hypertension, cardiovascular disease (CVD) and type 2 diabetes mellitus (T2DM) is well known. Importantly, the retrospective analysis of more than 1000 epidemiological studies have also shown the positive correlation between the excess of fatness with the risk of cancer. In addition, more important than weight, it is the dysfunctional adipose tissue the main driver of insulin resistance, metabolic syndrome and all cause of mortality and cancer deaths, which also explains why normal weight individuals may behave as “metabolically unhealthy obese” individuals. Adipocytes also have direct effects on tumor cells through paracrine signaling. Downregulation of adiponectin and upregulation of leptin in serum correlate with markers of chronic inflammation, and crown like structures (CLS) associated to the adipose tissue disfunction. Nevertheless, obesity is a preventable risk factor in cancer. Lifestyle interventions might contribute to reduce the adverse effects of obesity. Thus, Mediterranean diet interventional studies have been shown to reduce to circulation inflammatory factors, insulin sensitivity and cardiovascular function, with durable responses of up to 2 years in obese patients. Mediterranean diet supplemented with extra-virgin olive oil reduced the incidence of breast cancer compared with a control diet. Physical activity is another important lifestyle factor which may also contribute to reduced systemic biomarkers of metabolic syndrome associated to obesity. In this scenario, precision nutrition may provide complementary approaches to target the metabolic inflammation associated to “unhealthy obesity”. Herein, we first describe the different types of adipose tissue -thermogenic active brown adipose tissue (BAT) versus the energy storing white adipose tissue (WAT). We then move on precision nutrition based strategies, by mean of natural extracts derived from plants and/or diet derived ingredients, which may be useful to normalize the metabolic inflammation associated to “unhealthy obesity”. More specifically, we focus on two axis: (1) the activation of thermogenesis in BAT and browning of WAT; (2) and the potential of augmenting the oxidative capacity of muscles to dissipate energy. These strategies may be particularly relevant as complementary approaches to alleviate obesity associated effects on chronic inflammation, immunosuppression, angiogenesis and chemotherapy resistance in cancer. Finally, we summarize main studies where plant derived extracts, mainly, polyphenols and flavonoids, have been applied to increase the energy expenditure.
Collapse
Affiliation(s)
- Marina Reguero
- Molecular Oncology Group, Precision Nutrition and Health, IMDEA Food Institute, CEI UAM + CSIC, Ctra. de Cantoblanco 8, 28049 Madrid, Spain; (M.R.); (S.W.)
- NATAC BIOTECH, Electronica 7, Alcorcón, 28923 Madrid, Spain;
| | - Marta Gómez de Cedrón
- Molecular Oncology Group, Precision Nutrition and Health, IMDEA Food Institute, CEI UAM + CSIC, Ctra. de Cantoblanco 8, 28049 Madrid, Spain; (M.R.); (S.W.)
- Correspondence: (M.G.d.C.); (A.R.d.M.)
| | - Sonia Wagner
- Molecular Oncology Group, Precision Nutrition and Health, IMDEA Food Institute, CEI UAM + CSIC, Ctra. de Cantoblanco 8, 28049 Madrid, Spain; (M.R.); (S.W.)
- Medicinal Gardens SL, Marqués de Urquijo 47, 28008 Madrid, Spain
| | - Guillermo Reglero
- Production and Characterization of Novel Foods Department, Institute of Food Science Research CIAL, CEI UAM + CSIC, 28049 Madrid, Spain;
| | | | - Ana Ramírez de Molina
- Molecular Oncology Group, Precision Nutrition and Health, IMDEA Food Institute, CEI UAM + CSIC, Ctra. de Cantoblanco 8, 28049 Madrid, Spain; (M.R.); (S.W.)
- Correspondence: (M.G.d.C.); (A.R.d.M.)
| |
Collapse
|
21
|
Wang X, Liang F, Fan H, Wang Z, Gou X, Ning Y. Proteinase-activated receptor 2 promotes 3T3-L1 preadipocyte differentiation through activation of the PI3K/AKT signalling pathway and MAT2A gene expression. Arch Physiol Biochem 2020; 126:468-475. [PMID: 32990471 DOI: 10.1080/13813455.2020.1817094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The present study aimed to investigate the function and mechanisms of PAR2 in preadipocyte differentiation. This study found that the expression level of PAR2 was increased during 3T3-L1 mouse preadipocyte differentiation towards adipocytes. In addition, PAR2 overexpression significantly stimulated the expression of adipogenic proteins including ACC1, PPARγ, and SREBF1. Moreover, PAR2 overexpression increased the content of triglyceride (TG) in 3T3-L1 preadipocytes. Knockdown of PAR2 suppressed 3T3-L1 preadipocyte differentiation and adipogenesis. Mechanistically, PAR2 promoted 3T3-L1 preadipocyte differentiation and TG production through activation of the PI3K/AKT signalling pathway and MAT2A gene expression. The research sheds light on the adipogenic effects of PAR2 and its underlying mechanisms. Thus, PAR2 may have therapeutic significance for obesity.
Collapse
Affiliation(s)
- Xiaojuan Wang
- Nursing Department, Xianyang Central Hospital, Xianyang, China
| | - Fang Liang
- CPC Committee, The First People's Hospital of Xianyang, Xianyang, China
| | - Hui Fan
- Nursing Department, Xianyang Central Hospital, Xianyang, China
| | - Zhijie Wang
- The First People's Hospital of Xianyang, Xianyang, China
| | - Xiaolin Gou
- Department of Endocrinology, The First People's Hospital of Xianyang, Xianyang, China
| | - Yu Ning
- Department of Endocrinology, The First People's Hospital of Xianyang, Xianyang, China
| |
Collapse
|
22
|
Kässner F, Kirstein A, Händel N, Schmid GL, Landgraf K, Berthold A, Tannert A, Schaefer M, Wabitsch M, Kiess W, Körner A, Garten A. A new human adipocyte model with PTEN haploinsufficiency. Adipocyte 2020; 9:290-301. [PMID: 32579864 PMCID: PMC7469440 DOI: 10.1080/21623945.2020.1785083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Few human cell strains are suitable and readily available as in vitro adipocyte models. We used resected lipoma tissue from a patient with germline phosphatase and tensin homolog (PTEN) haploinsufficiency to establish a preadipocyte cell strain termed LipPD1 and aimed to characterize cellular functions and signalling pathway alterations in comparison to the established adipocyte model Simpson-Golabi-Behmel-Syndrome (SGBS) and to primary stromal-vascular fraction cells. We found that both cellular life span and the capacity for adipocyte differentiation as well as adipocyte-specific functions were preserved in LipPD1 and comparable to SGBS adipocytes. Basal and growth factor-stimulated activation of the PI3 K/AKT signalling pathway was increased in LipPD1 preadipocytes, corresponding to reduced PTEN levels in comparison to SGBS cells. Altogether, LipPD1 cells are a novel primary cell model with a defined genetic lesion suitable for the study of adipocyte biology.
Collapse
Affiliation(s)
- Franziska Kässner
- Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Anna Kirstein
- Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | | | - Gordian L. Schmid
- Department for General Practise, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Kathrin Landgraf
- Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Antje Berthold
- Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | | | - Michael Schaefer
- Rudolf Boehm Institute for Pharmacology and Toxicology, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, University Hospital for Children and Adolescents, Ulm University, Germany
| | - Wieland Kiess
- Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Antje Körner
- Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Antje Garten
- Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
- Institute for Metabolism and Systems Research, College for Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
23
|
Aranaz P, Navarro-Herrera D, Zabala M, Romo-Hualde A, López-Yoldi M, Vizmanos JL, Milagro FI, González-Navarro CJ. Phenolic Compounds Reduce the Fat Content in Caenorhabditis elegans by Affecting Lipogenesis, Lipolysis, and Different Stress Responses. Pharmaceuticals (Basel) 2020; 13:E355. [PMID: 33143060 PMCID: PMC7693530 DOI: 10.3390/ph13110355] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/22/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
Supplementation with bioactive compounds capable of regulating energy homeostasis is a promising strategy to manage obesity. Here, we have screened the ability of different phenolic compounds (myricetin, kaempferol, naringin, hesperidin, apigenin, luteolin, resveratrol, curcumin, and epicatechin) and phenolic acids (p-coumaric, ellagic, ferulic, gallic, and vanillic acids) regulating C. elegans fat accumulation. Resveratrol exhibited the strongest lipid-reducing activity, which was accompanied by the improvement of lifespan, oxidative stress, and aging, without affecting worm development. Whole-genome expression microarrays demonstrated that resveratrol affected fat mobilization, fatty acid metabolism, and unfolded protein response of the endoplasmic reticulum (UPRER), mimicking the response to calorie restriction. Apigenin induced the oxidative stress response and lipid mobilization, while vanillic acid affected the unfolded-protein response in ER. In summary, our data demonstrates that phenolic compounds exert a lipid-reducing activity in C. elegans through different biological processes and signaling pathways, including those related with lipid mobilization and fatty acid metabolism, oxidative stress, aging, and UPR-ER response. These findings open the door to the possibility of combining them in order to achieve complementary activity against obesity-related disorders.
Collapse
Affiliation(s)
- Paula Aranaz
- Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (P.A.); (D.N.-H.); (M.Z.); (A.R.-H.); (M.L.-Y.); (F.I.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain;
| | - David Navarro-Herrera
- Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (P.A.); (D.N.-H.); (M.Z.); (A.R.-H.); (M.L.-Y.); (F.I.M.)
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
| | - María Zabala
- Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (P.A.); (D.N.-H.); (M.Z.); (A.R.-H.); (M.L.-Y.); (F.I.M.)
| | - Ana Romo-Hualde
- Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (P.A.); (D.N.-H.); (M.Z.); (A.R.-H.); (M.L.-Y.); (F.I.M.)
| | - Miguel López-Yoldi
- Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (P.A.); (D.N.-H.); (M.Z.); (A.R.-H.); (M.L.-Y.); (F.I.M.)
| | - José Luis Vizmanos
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain;
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
| | - Fermín I. Milagro
- Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (P.A.); (D.N.-H.); (M.Z.); (A.R.-H.); (M.L.-Y.); (F.I.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carlos J. González-Navarro
- Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (P.A.); (D.N.-H.); (M.Z.); (A.R.-H.); (M.L.-Y.); (F.I.M.)
| |
Collapse
|
24
|
Lang L, Loveless R, Teng Y. Emerging Links between Control of Mitochondrial Protein ATAD3A and Cancer. Int J Mol Sci 2020; 21:E7917. [PMID: 33113782 PMCID: PMC7663417 DOI: 10.3390/ijms21217917] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/17/2022] Open
Abstract
Spanning from the mitochondria's outer surface to the inner membrane, the nuclear-encoded protein ATAD3A maintains vital roles in regulating mitochondrial dynamics, homeostasis, metabolism, and interactions with the endoplasmic reticulum. Recently, elevated levels of ATAD3A have been reported in several types of cancer and to be tightly correlated with cancer development and progression, including increased cancer cell potential of proliferation, metastasis, and resistance to chemotherapy and radiotherapy. In the current review, we reveal ATAD3A as the link between mitochondrial functions and cancer biology and the accumulating evidence presenting ATAD3A as an attractive target for the development of novel cancer therapy to inhibit aberrant cancer metabolism and progression.
Collapse
Affiliation(s)
- Liwei Lang
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.L.); (R.L.)
| | - Reid Loveless
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.L.); (R.L.)
| | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.L.); (R.L.)
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Medical Laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
25
|
Goktas Z, Zu Y, Abbasi M, Galyean S, Wu D, Fan Z, Wang S. Recent Advances in Nanoencapsulation of Phytochemicals to Combat Obesity and Its Comorbidities. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:8119-8131. [PMID: 32633507 PMCID: PMC8507418 DOI: 10.1021/acs.jafc.0c00131] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
An increasing epidemic of obesity has become a serious public health concern primarily because it contributes to pathogenesis of many chronic diseases including type 2 diabetes, cardiovascular disease, hepatobiliary disease, obstructive sleep apnea, kidney disease, some types of cancer, among others. Consumption of a variety of phytochemicals has emerged as a promising potential for combating obesity and its comorbidities. However, the generally low aqueous solubility, stability, bioavailability, and target specificity of phytochemicals, along with their side-effects and toxicity seen when used at high doses, have restricted their clinical applications. As a solution, phytochemicals can be encapsulated into nanoparticles to increase their stability and solubility, enhance their bioavailability, protect them from premature degradation in the body, prolong their circulation time, and thus enhance their antiobesity activity. In this perspective, we summarize the problems and limitations of the prominent phytochemicals (epigallocatechin gallate, trans-resveratrol, curcumin, and quercetin), the major biocompatible and biodegradable nanoparticles, and the efficacy of nanoencapsulated forms of these phytochemicals in combating obesity and its comorbidities.
Collapse
Affiliation(s)
- Zeynep Goktas
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Hacettepe University, 06100 Ankara, Turkey
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas 79409, United States
| | - Mehrnaz Abbasi
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas 79409, United States
| | - Shannon Galyean
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas 79409, United States
| | - Dayong Wu
- Nutrition Immunology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts 02111, United States
| | - Zhaoyang Fan
- Department of Electrical & Computer Engineering and Nano Tech Center, Texas Tech University, Lubbock, Texas 79409, United States
| | - Shu Wang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas 79409, United States
| |
Collapse
|
26
|
Frazier AE, Compton AG, Kishita Y, Hock DH, Welch AE, Amarasekera SSC, Rius R, Formosa LE, Imai-Okazaki A, Francis D, Wang M, Lake NJ, Tregoning S, Jabbari JS, Lucattini A, Nitta KR, Ohtake A, Murayama K, Amor DJ, McGillivray G, Wong FY, van der Knaap MS, Jeroen Vermeulen R, Wiltshire EJ, Fletcher JM, Lewis B, Baynam G, Ellaway C, Balasubramaniam S, Bhattacharya K, Freckmann ML, Arbuckle S, Rodriguez M, Taft RJ, Sadedin S, Cowley MJ, Minoche AE, Calvo SE, Mootha VK, Ryan MT, Okazaki Y, Stroud DA, Simons C, Christodoulou J, Thorburn DR. Fatal perinatal mitochondrial cardiac failure caused by recurrent de novo duplications in the ATAD3 locus. MED 2020; 2:49-73. [PMID: 33575671 DOI: 10.1016/j.medj.2020.06.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background In about half of all patients with a suspected monogenic disease, genomic investigations fail to identify the diagnosis. A contributing factor is the difficulty with repetitive regions of the genome, such as those generated by segmental duplications. The ATAD3 locus is one such region, in which recessive deletions and dominant duplications have recently been reported to cause lethal perinatal mitochondrial diseases characterized by pontocerebellar hypoplasia or cardiomyopathy, respectively. Methods Whole exome, whole genome and long-read DNA sequencing techniques combined with studies of RNA and quantitative proteomics were used to investigate 17 subjects from 16 unrelated families with suspected mitochondrial disease. Findings We report six different de novo duplications in the ATAD3 gene locus causing a distinctive presentation including lethal perinatal cardiomyopathy, persistent hyperlactacidemia, and frequently corneal clouding or cataracts and encephalopathy. The recurrent 68 Kb ATAD3 duplications are identifiable from genome and exome sequencing but usually missed by microarrays. The ATAD3 duplications result in the formation of identical chimeric ATAD3A/ATAD3C proteins, altered ATAD3 complexes and a striking reduction in mitochondrial oxidative phosphorylation complex I and its activity in heart tissue. Conclusions ATAD3 duplications appear to act in a dominant-negative manner and the de novo inheritance infers a low recurrence risk for families, unlike most pediatric mitochondrial diseases. More than 350 genes underlie mitochondrial diseases. In our experience the ATAD3 locus is now one of the five most common causes of nuclear-encoded pediatric mitochondrial disease but the repetitive nature of the locus means ATAD3 diagnoses may be frequently missed by current genomic strategies. Funding Australian NHMRC, US Department of Defense, Japanese AMED and JSPS agencies, Australian Genomics Health Alliance and Australian Mito Foundation.
Collapse
Affiliation(s)
- Ann E Frazier
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia.,These authors contributed equally: A.E. Frazier, A.G. Compton
| | - Alison G Compton
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia.,These authors contributed equally: A.E. Frazier, A.G. Compton
| | - Yoshihito Kishita
- Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Juntendo University, Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Daniella H Hock
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC 3052, Australia
| | - AnneMarie E Welch
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia
| | - Sumudu S C Amarasekera
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Rocio Rius
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Luke E Formosa
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Atsuko Imai-Okazaki
- Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Juntendo University, Graduate School of Medicine, Tokyo, 113-8421, Japan.,Division of Genomic Medicine Research, Medical Genomics Center, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - David Francis
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia
| | - Min Wang
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia
| | - Nicole J Lake
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia.,Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Simone Tregoning
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia
| | - Jafar S Jabbari
- Australian Genome Research Facility Ltd, Victorian Comprehensive Cancer Centre, Melbourne VIC 3052, Australia
| | - Alexis Lucattini
- Australian Genome Research Facility Ltd, Victorian Comprehensive Cancer Centre, Melbourne VIC 3052, Australia
| | - Kazuhiro R Nitta
- Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Juntendo University, Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Akira Ohtake
- Department of Pediatrics & Clinical Genomics, Saitama Medical University Hospital, Saitama, 350-0495, Japan
| | - Kei Murayama
- Department of Metabolism, Chiba Children's Hospital, Chiba, 266-0007, Japan
| | - David J Amor
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
| | - George McGillivray
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia
| | - Flora Y Wong
- Ritchie Centre, Hudson Institute of Medical Research; Department of Paediatrics, Monash University; and Monash Newborn, Monash Children's Hospital, Melbourne, VIC 3168, Australia
| | - Marjo S van der Knaap
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands.,Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit and Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - R Jeroen Vermeulen
- Department of Neurology, Maastricht University Medical Center, 6229 HX, Maastricht, The Netherlands
| | - Esko J Wiltshire
- Department of Paediatrics and Child Health, University of Otago Wellington and Capital and Coast District Health Board, Wellington 6021, New Zealand
| | - Janice M Fletcher
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA 5000, Australia
| | - Barry Lewis
- Department of Clinical Biochemistry, PathWest Laboratory Medicine Western Australia, Nedlands, WA 6009, Australia
| | - Gareth Baynam
- Western Australian Register of Developmental Anomalies and Genetic Services of Western Australia and King Edward Memorial Hospital for Women Perth, Subiaco, WA 6008, Australia.,Telethon Kids Institute and School of Paediatrics and Child Health, The University of Western Australia, Perth, WA 6009, Australia
| | - Carolyn Ellaway
- Genetic Metabolic Disorders Service, Sydney Children's Hospital Network, The Children's Hospital at Westmead, Sydney, NSW 2145, Australia.,Disciplines of Genomic Medicine and Child and Adolescent Health, Sydney Medical School, University of Sydney, NSW 2145, Australia
| | - Shanti Balasubramaniam
- Genetic Metabolic Disorders Service, Sydney Children's Hospital Network, The Children's Hospital at Westmead, Sydney, NSW 2145, Australia
| | - Kaustuv Bhattacharya
- Genetic Metabolic Disorders Service, Sydney Children's Hospital Network, The Children's Hospital at Westmead, Sydney, NSW 2145, Australia.,Disciplines of Genomic Medicine and Child and Adolescent Health, Sydney Medical School, University of Sydney, NSW 2145, Australia
| | | | - Susan Arbuckle
- Department of Histopathology, The Children's Hospital at Westmead, Sydney Children's Hospital Network, Sydney, NSW 2145, Australia
| | - Michael Rodriguez
- Discipline of Pathology, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Simon Sadedin
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia
| | - Mark J Cowley
- Children's Cancer Institute, Kensington, NSW 2750, Australia; St Vincent's Clinical School, UNSW Sydney, Darlinghurst, NSW 2010, Australia.,Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - André E Minoche
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Sarah E Calvo
- Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02446, USA
| | - Vamsi K Mootha
- Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02446, USA
| | - Michael T Ryan
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Yasushi Okazaki
- Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Juntendo University, Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - David A Stroud
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Cas Simons
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072 Australia
| | - John Christodoulou
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia.,Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Disciplines of Genomic Medicine and Child and Adolescent Health, Sydney Medical School, University of Sydney, NSW 2145, Australia
| | - David R Thorburn
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia.,Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.,Lead contact
| |
Collapse
|
27
|
Chang CC, Chang CY, Lin PC, Huang JP, Chen KH, Yen TH, Hung LM. Administration of low-dose resveratrol attenuated hepatic inflammation and lipid accumulation in high cholesterol-fructose diet-induced rat model of nonalcoholic fatty liver disease. CHINESE J PHYSIOL 2020; 63:149-155. [PMID: 32859881 DOI: 10.4103/cjp.cjp_43_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Resveratrol (RSV) has been demonstrated to ameliorate nonalcoholic fatty liver disease (NAFLD) in animal studies. However, RSV was given with the dosage that ranged from 7 to 300 mg/kg body weight (BW). Hence, the study aimed to investigate the efficacy of RSV at a lower dosage on high cholesterol-fructose diet (HCFD)-induced rat model of NAFLD. In the study, male Sprague-Dawley rats were fed with HCFD for 15 weeks. RSV was also given at a daily dose of 1 mg/kg BW for 15 days or 15 weeks by oral delivery. At sacrifice, plasma and liver specimens were acquired for detections of alanine and aspartate aminotransferases, proinflammatory cytokines, and lipid contents. Histological examinations and Western blotting analysis were performed using liver tissues. The results showed that RSV administration reduced plasma levels of aminotransferases and proinflammatory cytokines including interleukin-1 beta (IL-1β), IL-6, and tumor necrosis factor-alpha (TNF-α) in HCFD-induced NAFLD. RSV also mitigated hepatic lipid accumulation and expression of IL-1β, IL-6, and TNF-α. Besides, phosphorylation of signal transducer and activator of transcription 3 (STAT3) was reduced with RSV supplementation in the liver of HCFD-fed rats. We concluded that low-dose RSV supplementation attenuated hepatic inflammation and lipid accumulation in HCFD-induced NAFLD. The ameliorative effect of RSV on NAFLD could be associated with downregulation of phosphorylated STAT3.
Collapse
Affiliation(s)
- Chih-Chun Chang
- Department of Clinical Pathology, Far Eastern Memorial Hospital, New Taipei; Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, Yilan, Taiwan
| | - Chieh-Yu Chang
- Department of Internal Medicine, Division of Cardiology, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Pei-Chun Lin
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Jiung-Pang Huang
- Department and Graduate Institute of Biomedical Sciences, College of Medicine; Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan
| | - Kuan-Hsing Chen
- Healthy Aging Research Center, Chang Gung University; Kidney Research Center, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Tzung-Hai Yen
- Kidney Research Center; Department of Nephrology, Clinical Poison Center, Chang Gung Memorial Hospital; College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Li-Man Hung
- Department and Graduate Institute of Biomedical Sciences, College of Medicine; Healthy Aging Research Center, Chang Gung University; Kidney Research Center, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| |
Collapse
|
28
|
Chen Y, Cai GH, Xia B, Wang X, Zhang CC, Xie BC, Shi XC, Liu H, Lu JF, Zhang RX, Zhu MQ, Liu M, Yang SZ, Yang Zhang D, Chu XY, Khan R, Wang YL, Wu JW. Mitochondrial aconitase controls adipogenesis through mediation of cellular ATP production. FASEB J 2020; 34:6688-6702. [PMID: 32212192 DOI: 10.1096/fj.201903224rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 11/11/2022]
Abstract
Mitochondrial aconitase (Aco2) catalyzes the conversion of citrate to isocitrate in the TCA cycle, which produces NADH and FADH2, driving synthesis of ATP through OXPHOS. In this study, to explore the relationship between adipogenesis and mitochondrial energy metabolism, we hypothesize that Aco2 may play a key role in the lipid synthesis. Here, we show that overexpression of Aco2 in 3T3-L1 cells significantly increased lipogenesis and adipogenesis, accompanied by elevated mitochondrial biogenesis and ATP production. However, when ATP is depleted by rotenone, an inhibitor of the respiratory chain, the promotive role of Aco2 in adipogenesis is abolished. In contrast to Aco2 overexpression, deficiency of Aco2 markedly reduced lipogenesis and adipogenesis, along with the decreased mitochondrial biogenesis and ATP production. Supplementation of isocitrate efficiently rescued the inhibitory effect of Aco2 deficiency. Similarly, the restorative effect of isocitrate was abolished in the presence of rotenone. Together, these results show that Aco2 sustains normal adipogenesis through mediating ATP production, revealing a potential mechanistic link between TCA cycle enzyme and lipid synthesis. Our work suggest that regulation of adipose tissue mitochondria function may be a potential way for combating abnormal adipogenesis related diseases such as obesity and lipodystrophy.
Collapse
Affiliation(s)
- Yan Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Guo He Cai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Bo Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xin Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Cong Cong Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Bao Cai Xie
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiao Chen Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Huan Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jun Feng Lu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Rui Xin Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Meng Qing Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Min Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Shi Zhen Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Dan Yang Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xin Yi Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Rajwali Khan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yong Liang Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jiang Wei Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
29
|
Resveratrol Modifies Lipid Composition of Two Cancer Cell Lines. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5393041. [PMID: 32149115 PMCID: PMC7053465 DOI: 10.1155/2020/5393041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/30/2019] [Indexed: 01/01/2023]
Abstract
Resveratrol (Resv) offers health benefits in cancer and has been reported to modulate important enzymes of lipid metabolism. Studies of its effects on lipid composition in different subtypes of breast-cancer cells are scarce. Thus, we investigated the alterations in phospholipids (PL), fatty acids (FA), and lipid metabolism enzymes in two breast-cancer cell lines after Resv treatment. MCF-7 and MDA-MB-231 cells were treated with 80 and 200 μM of Resv, respectively, for 24 hours. We analyzed PL with radiolabeled inorganic phosphate (32Pi) by thin-layer chromatography, FA by gas chromatography-mass spectrometry, and lipid metabolism enzymes (DGAT2, FAS, ρACCβ, pAMPKα, and AMPK) by Western blot. Resv treated MDA-MB-231 phospholipids showed a reduction in phosphatidylcholine (63%) and phosphatidylethanolamine (35%). We observed an increase in eicosapentaenoic acid (EPA) (73%) and docosahexaenoic acid (DHA) (65%) in MCF-7 cells after Resv treatment. Interestingly, the same treatment caused 50% and 90% increases in EPA and DHA, respectively, in MDA-MB-231 cells. In MCF-7 cells, Resv increased the expression of ρACCβ (3.3-fold) and AMPKα/ρAMPKα (1.5-fold) and in MDA-MB-231 cells it inhibited the expression of ρACCβ (111.8-fold) and AMPKα/ρAMPKα (1.2 fold). Our results show that Resv modified PL and saturated and unsaturated FA especially in MDA-MB-231 cells, and open new perspectives to the understanding of the reported anticancer effect of Resv on these cells.
Collapse
|
30
|
Koushki M, Zare M, Shabani M, Teimouri M, Hosseini H, Babaei Khorzoughi R, Meshkani R. Resveratrol Reduces Lipid Accumulation through Upregulating the Expression of MicroRNAs Regulating Fatty Acid Bet Oxidation in Liver Cells: Evidence from In-vivo and In-vitro Studies. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 19:333-340. [PMID: 33224240 PMCID: PMC7667538 DOI: 10.22037/ijpr.2019.111745.13332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
MicroRNAs have been shown to regulate lipogenesis in liver. The aim of the present study was to investigate whether the effects of resveratrol (RSV) on lipogenesis are associated with the changes in the expression of two miRNAs (miR-107 and miR-10b) that regulate lipogenic pathways. 30 wild type C57BL/6j male mice were randomly fed three diets: a standard chow diet (ND), a high fat diet (HFD, 60% fat) and the high fat diet supplemented with 0.4% RSV (HFD-RSV) for 16 weeks. HepG2 cells were treated with high glucose (33 mM) and RSV (20 µM) for 24 h. The expression of the genes and miRNAs were measured by real-time PCR. Triglyceride level was increased in the liver of mice and HepG2 cells. In both animal and In-vitro experiments, triglyceride level was significantly decreased in groups treated with RSV. The expression of the miR-107 and miR-10b was significantly upregulated in the liver of HFD mice, whereas HFD-RSV group demonstrated a significant lower expression of both miRNAs compared to HFD group. In addition, RSV treatment significantly upregulated the expression of CPT-1a and PPARα genes in the liver of HFD mice. Moreover, treatment with RSV could reduce the expression of miR-107 and miR-10b and increase the expression of CPT-1a and PPARα in HG-treated HepG2 cells. These evidence, as a whole, suggest that RSV could exert its anti-lipogenic effect partially through alterations in the expression of miR-107 and miR-10b in liver cells.
Collapse
Affiliation(s)
- Mehdi Koushki
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mina Zare
- Recombinant Protein Laboratory, Department of Biochemistry, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Maryam Shabani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Teimouri
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hossein Hosseini
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Reyhaneh Babaei Khorzoughi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Effect of resveratrol on adipokines and myokines involved in fat browning: Perspectives in healthy weight against obesity. Pharmacol Res 2019; 148:104411. [PMID: 31449976 DOI: 10.1016/j.phrs.2019.104411] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/22/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023]
Abstract
Obesity is a globally widespread metabolic disorder, characterized by immoderate fat accumulation in the body. There are different types of body fats such as white adipose tissue (WAT), which stores surplus energy in the body, and brown adipose tissue (BAT) which utilize energy to produce heat during metabolism. BAT acts many beneficial functions in metabolic disorders including type 2 diabetes and obesity. Recent studies have investigated methods for promoting the fat browning process of WAT in obesity because of various reasons such as the improvement of insulin resistance, and weight loss. Among natural polyphenolic compounds, resveratrol has been highlighted due to its anti-oxidant and anti-obesity as well as anti-inflammation and anti-cancer properties. Recent studies have paid a lot of attention to that resveratrol may act as a fat browning activator, involved in the secretion of many myokines and adipokines. Here, we reviewed the role of resveratrol in fat browning and also the association between resveratrol and adipokines/myokines in the fat browning process. Our review may provide novel insight into the role of resveratrol in fat browning, leading to the maintenance of a healthy weight against obesity.
Collapse
|
32
|
Park U, Hwang J, Youn H, Kim E, Um S. Piperine inhibits adipocyte differentiation via dynamic regulation of histone modifications. Phytother Res 2019; 33:2429-2439. [DOI: 10.1002/ptr.6434] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 06/03/2019] [Accepted: 06/08/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Ui‐Hyun Park
- Department of Integrative Bioscience and BiotechnologySejong University 209 Neungdong‐ro, Gwangjin‐gu Seoul 05006 Korea
| | - Jin‐Taek Hwang
- Korea Food Research InstituteResearch Group of Healthcare 245 Nongsaengmyeong‐ro Jeonju Jeonbuk 55365 Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and BiotechnologySejong University 209 Neungdong‐ro, Gwangjin‐gu Seoul 05006 Korea
| | - Eun‐Joo Kim
- Department of Molecular BiologyDankook University Cheonan Chungnam 31116 Korea
| | - Soo‐Jong Um
- Department of Integrative Bioscience and BiotechnologySejong University 209 Neungdong‐ro, Gwangjin‐gu Seoul 05006 Korea
| |
Collapse
|
33
|
Resveratrol Anti-Obesity Effects: Rapid Inhibition of Adipocyte Glucose Utilization. Antioxidants (Basel) 2019; 8:antiox8030074. [PMID: 30917543 PMCID: PMC6466544 DOI: 10.3390/antiox8030074] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 12/11/2022] Open
Abstract
Studies in animal models of diabetes and obesity have shown that resveratrol mitigates complications of metabolic diseases, beyond those resulting from oxidative stress. Furthermore, results obtained with cultured preadipocytes have also revealed that prolonged resveratrol treatment impairs adipogenesis. Considering the role of adipocytes in the hypertrophy of fat stores, and keeping in mind that insulin is the main trigger of excessive energy storage during post-prandial periods, the present study aimed to investigate how short-term effects of resveratrol can limit glucose disposal in a gut-adipose tissue axis. We found that resveratrol exhibits a more potent inhibitory capacity towards α-glucosidase than pancreatic lipase activity. Resveratrol also rapidly blunts glucose transport in mature fat cells by counteracting the effect of insulin and insulin-like lipogenic agents. Within two hours, resveratrol also inhibited the incorporation of glucose into lipids of adipocytes, which was unaffected by membrane cholesterol depletion. Moreover, the comparison between adipocytes with invalidated semicarbazide-sensitive amine oxidase activity and their control, or between resveratrol and several inhibitors, did not indicate that the recently described interaction of resveratrol with amine oxidases was involved in its antilipogenic effect. Caffeine and piceatannol, previously said to interact with glucose carriers, also inhibit lipogenesis in adipocytes, whereas other antioxidant phytochemicals do not reproduce such an antilipogenic effect. This study highlights the diverse first steps by which resveratrol impairs excessive fat accumulation, indicating that this natural molecule and its derivatives deserve further studies to develop their potential anti-obesity properties.
Collapse
|
34
|
Li AN, Chen JJ, Li QQ, Zeng GY, Chen QY, Chen JL, Liao ZM, Jin P, Wang KS, Yang ZC. Alpha-glucosidase inhibitor 1-Deoxynojirimycin promotes beige remodeling of 3T3-L1 preadipocytes via activating AMPK. Biochem Biophys Res Commun 2019; 509:1001-1007. [DOI: 10.1016/j.bbrc.2019.01.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/05/2019] [Indexed: 12/11/2022]
|
35
|
Wiciński M, Leis K, Szyperski P, Węclewicz M, Mazur E, Pawlak-Osińska K. Impact of resveratrol on exercise performance: A review. Sci Sports 2018. [DOI: 10.1016/j.scispo.2018.02.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
36
|
Jeung WH, Shim JJ, Woo SW, Sim JH, Lee JL. Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032 Cell Extracts Inhibit Adipogenesis in 3T3-L1 and HepG2 Cells. J Med Food 2018; 21:876-886. [PMID: 30148699 DOI: 10.1089/jmf.2017.4157] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Some lactic acid bacteria (LAB) and their cellular components have antiobesity effects. In this study, we evaluated the antiadipogenic effects of a mixture of two LAB-Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032-using 3T3-L1 preadipocytes and HepG2 hepatocarcinoma cells. 3T3-L1 cells treated with a 1:1 ratio of HY7601 and KY1032 during differentiation showed reduced lipid accumulation by Oil Red O staining, as well as decreased leptin secretion and mRNA expression of peroxisome proliferator-activated receptor-γ and CCAAT/enhancer binding protein-α. HY7601 and KY1032 treatment also suppressed mitochondrial biogenesis and inhibited the expression of genes encoding mitochondrial transcription factors, as well as those related to fatty acid synthesis in HepG2 cells. The antiadipogenic effects of LAB were associated with the cell membrane fraction. These results demonstrate that a mixture of two LAB (HY7601 and KY1032) inhibits adipogenesis in preadipocytes and liver cells and is a potential therapeutic strategy for the treatment of obesity.
Collapse
Affiliation(s)
| | - Jae-Jung Shim
- 1 R&BD Center , Korea Yakult Co. Ltd., Yongin, Korea.,2 College of Agriculture and Life Sciences, Seoul National University , Seoul, Korea
| | - Seon-Wook Woo
- 1 R&BD Center , Korea Yakult Co. Ltd., Yongin, Korea
| | - Jae-Hun Sim
- 1 R&BD Center , Korea Yakult Co. Ltd., Yongin, Korea
| | - Jung-Lyoul Lee
- 1 R&BD Center , Korea Yakult Co. Ltd., Yongin, Korea.,3 College of Veterinary Medicine, Konkuk University , Seoul, Korea
| |
Collapse
|
37
|
Carpéné C, Pejenaute H, Del Moral R, Boulet N, Hijona E, Andrade F, Villanueva-Millán MJ, Aguirre L, Arbones-Mainar JM. The Dietary Antioxidant Piceatannol Inhibits Adipogenesis of Human Adipose Mesenchymal Stem Cells and Limits Glucose Transport and Lipogenic Activities in Adipocytes. Int J Mol Sci 2018; 19:ijms19072081. [PMID: 30018277 PMCID: PMC6073844 DOI: 10.3390/ijms19072081] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/20/2022] Open
Abstract
Phenolic compounds are among the most investigated herbal remedies, as is especially the case for resveratrol. Many reports have shown its anti-aging properties and the ability to reduce obesity and diabetes induced by high-fat diet in mice. However, such beneficial effects hardly translate from animal models to humans. The scientific community has therefore tested whether other plant phenolic compounds may surpass the effects of resveratrol. In this regard, it has been reported that piceatannol reproduces in rodents the anti-obesity actions of its parent polyphenol. However, the capacity of piceatannol to inhibit adipocyte differentiation in humans has not been characterized so far. Here, we investigated whether piceatannol was antiadipogenic and antilipogenic in human preadipocytes. Human mesenchymal stem cells (hMSC), isolated from adipose tissues of lean and obese individuals, were differentiated into mature adipocytes with or without piceatannol, and their functions were explored. Fifty µM of piceatannol deeply limited synthesis/accumulation of lipids in both murine and hMSC-derived adipocytes. Interestingly, this phenomenon occurred irrespective of being added at the earlier or later stages of adipocyte differentiation. Moreover, piceatannol lowered glucose transport into adipocytes and decreased the expression of key elements of the lipogenic pathway (PPARγ, FAS, and GLUT4). Thus, the confirmation of the antiadipogenic properties of piceatanol in vitro warrants the realization of clinical studies for the application of this compound in the treatment of the metabolic complications associated with obesity.
Collapse
Affiliation(s)
- Christian Carpéné
- INSERM U1048, Institute of Metabolic and Cardiovascular Diseases (I2MC), Paul Sabatier University, 31059 Toulouse, France.
| | - Héctor Pejenaute
- Adipocyte and Fat Biology Laboratory (AdipoFat), Unidad de Investigación Traslacional, Instituto Aragonés de Ciencias de la Salud (IACS), Instituto de Investigación Sanitaria (IIS) Aragon, 50009 Zaragoza, Spain.
| | - Raquel Del Moral
- Adipocyte and Fat Biology Laboratory (AdipoFat), Unidad de Investigación Traslacional, Instituto Aragonés de Ciencias de la Salud (IACS), Instituto de Investigación Sanitaria (IIS) Aragon, 50009 Zaragoza, Spain.
| | - Nathalie Boulet
- INSERM U1048, Institute of Metabolic and Cardiovascular Diseases (I2MC), Paul Sabatier University, 31059 Toulouse, France.
| | - Elizabeth Hijona
- Department of Gastroenterology, University of Basque Country (UPV/EHU), Biodonostia Research Institute, 20014 San Sebastián, Spain.
| | - Fernando Andrade
- Division of Metabolism, Cruces University Hospital and BioCruces Health Research Institute, Plaza de Cruces s/n, 48903 Barakaldo, Spain.
| | - Maria Jesùs Villanueva-Millán
- HIV and Associated Metabolic Alterations Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain.
| | - Leixuri Aguirre
- Nutrition and Obesity Group, Department of Nutrition and Food Science, Faculty of Pharmacy and Lucio Lascaray Research Center, University of the Basque Country (UPV/EHU), 01006 Vitoria, Spain.
- CIBEROBN Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain.
| | - José Miguel Arbones-Mainar
- Adipocyte and Fat Biology Laboratory (AdipoFat), Unidad de Investigación Traslacional, Instituto Aragonés de Ciencias de la Salud (IACS), Instituto de Investigación Sanitaria (IIS) Aragon, 50009 Zaragoza, Spain.
- CIBEROBN Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain.
| |
Collapse
|
38
|
Teplova VV, Isakova EP, Klein OI, Dergachova DI, Gessler NN, Deryabina YI. Natural Polyphenols: Biological Activity, Pharmacological Potential, Means of Metabolic Engineering (Review). APPL BIOCHEM MICRO+ 2018. [DOI: 10.1134/s0003683818030146] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
39
|
|
40
|
Chen D, Yuan X, Liu L, Zhang M, Qu B, Zhen Z, Gao X. Mitochondrial ATAD3A regulates milk biosynthesis and proliferation of mammary epithelial cells from dairy cow via the mTOR pathway. Cell Biol Int 2018; 42:533-542. [DOI: 10.1002/cbin.10929] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/26/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Dongying Chen
- The Key Laboratory of Dairy Science of Education Ministry; Northeast Agricultural University; Harbin 150030 China
| | - Xiaohan Yuan
- The Key Laboratory of Dairy Science of Education Ministry; Northeast Agricultural University; Harbin 150030 China
| | - Lijie Liu
- The Key Laboratory of Dairy Science of Education Ministry; Northeast Agricultural University; Harbin 150030 China
| | - Minghui Zhang
- The Key Laboratory of Dairy Science of Education Ministry; Northeast Agricultural University; Harbin 150030 China
| | - Bo Qu
- The Key Laboratory of Dairy Science of Education Ministry; Northeast Agricultural University; Harbin 150030 China
| | - Zhen Zhen
- The Key Laboratory of Dairy Science of Education Ministry; Northeast Agricultural University; Harbin 150030 China
| | - Xuejun Gao
- The Key Laboratory of Dairy Science of Education Ministry; Northeast Agricultural University; Harbin 150030 China
| |
Collapse
|
41
|
Geng YW, Zhang Z, Liu MY, Hu WP. Differentiation of human dental pulp stem cells into neuronal by resveratrol. Cell Biol Int 2017; 41:1391-1398. [PMID: 28782906 DOI: 10.1002/cbin.10835] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 08/03/2017] [Indexed: 01/09/2023]
Abstract
Dental pulp stem cells (DPSCs) have been proposed as a promising source of stem cells in nerve regeneration due to their close embryonic origin and ease of harvest. Resveratrol (RSV) is a natural polyphenolic and possesses many biological functions such as anti-inflammatory activity and protection against atherosclerosis and neuroprotective activities. There is increasing evidence showing that RSV plays a pivotal role in neuron protection and neuronal differentiation. In this study, we isolated DPSCs from impacted third molars and investigated whether RSV induces neuronal differentiation of DPSCs. To avoid loss of DPSCs multipotency, all the experiments were conducted on cells at early passages. RT-PCR results showed that RSV-treated DPSCs (RSV-DPSCs) significantly increased the expression of the neuroprogenitor marker Nestin. When RSV-DPSCs were differentiated with neuronal induction media (RSV-dDPSCs), they showed a cell morphology similar to neurons. The expression of neuronal-specific marker genes Nestin, Musashi, and NF-M in RSV-dDPSCs was significantly increased. Immunocytochemical staining and Western blot analysis showed that the expression of neuronal marker proteins, Nestin, and NF-M, was significantly increased in RSV-dDPSCs. Therefore, we have shown that RSV treatment, along with the use of neuronal induction media, effectively promotes neuronal cell differentiation of DPSCs.
Collapse
Affiliation(s)
- Ya-Wei Geng
- Department of Prosthodontics, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, China
| | - Zhen Zhang
- Oral and Maxillofacial Surgery, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, China
| | - Ming-Yue Liu
- Department of Prosthodontics, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, China
| | - Wei-Ping Hu
- Department of Prosthodontics, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, China
| |
Collapse
|
42
|
Gomez-Zorita S, Belles C, Briot A, Fernández-Quintela A, Portillo MP, Carpéné C. Pterostilbene Inhibits Lipogenic Activity similar to Resveratrol or Caffeine but Differently Modulates Lipolysis in Adipocytes. Phytother Res 2017. [PMID: 28627722 DOI: 10.1002/ptr.5852] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The anti-obesity effects of resveratrol shown in rodents are not transposed into an efficient therapy of human obesity. Consequently, the search for molecules mimicking or surpassing resveratrol actions is ongoing. The natural phenolic compound pterostilbene exhibits beneficial health effects and has the capacity to limit fat mass in animal models. In this study, we tested whether pterostilbene modulates triacylglycerol accumulation/breakdown. Prolonged exposure to pterostilbene or resveratrol inhibited adipocyte differentiation in 3T3-F442A preadipocytes. Acute effects on lipolysis, antilipolysis and lipogenesis were determined for pterostilbene in mouse adipocytes, and compared with resveratrol. Pterostilbene was also tested on glycerol release and glucose uptake in subcutaneous human adipocytes. Dose-response analyses did not reveal a clear lipolytic effect in both species. The antilipolytic effect of insulin was improved by pterostilbene at 1-10 μM in mouse fat cells only, while at 1 mM, the phenolic compound was antilipolytic in human fat cells in a manner not additive to insulin. Pterostilbene dose-dependently inhibited glucose incorporation into lipids similarly to resveratrol and caffeine. However, only the former did not inhibit insulin-stimulated glucose uptake. Indeed, pterostilbene abolished the insulin lipogenic effect without inhibiting its antilipolytic action and rapid activation of glucose uptake. Pterostilbene therefore exhibits a unique panel of direct interactions with adipocytes that relies on its reported anti-obesity and antidiabetic properties. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Saioa Gomez-Zorita
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut. National de la Santé et de la Recherche Médicale and Université Paul Sabatier, Toulouse, France.,Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Centre, Vitoria, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), Spain
| | - Chloé Belles
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut. National de la Santé et de la Recherche Médicale and Université Paul Sabatier, Toulouse, France.,DIVA expertise, Centre Pierre Potier, Toulouse, France
| | - Anaïs Briot
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut. National de la Santé et de la Recherche Médicale and Université Paul Sabatier, Toulouse, France
| | - Alfredo Fernández-Quintela
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Centre, Vitoria, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), Spain
| | - Maria P Portillo
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Centre, Vitoria, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), Spain
| | - Christian Carpéné
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut. National de la Santé et de la Recherche Médicale and Université Paul Sabatier, Toulouse, France
| |
Collapse
|
43
|
Kim YJ, Chung SO, Kim JK, Park SU. Recent studies on resveratrol and its biological and pharmacological activity. EXCLI JOURNAL 2017; 16:602-608. [PMID: 28694761 PMCID: PMC5491918 DOI: 10.17179/excli2017-253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 04/11/2017] [Indexed: 02/03/2023]
Affiliation(s)
- Yong Joo Kim
- Department of Biosystems Machinery Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
| | - Sun Ok Chung
- Department of Biosystems Machinery Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
| | - Jae Kwang Kim
- Department of Crop Science, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
| | - Sang Un Park
- Division of Life Sciences and Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Korea
| |
Collapse
|
44
|
Chen L, Wang T, Chen G, Wang N, Gui L, Dai F, Fang Z, Zhang Q, Lu Y. Influence of resveratrol on endoplasmic reticulum stress and expression of adipokines in adipose tissues/adipocytes induced by high-calorie diet or palmitic acid. Endocrine 2017; 55:773-785. [PMID: 28070709 DOI: 10.1007/s12020-016-1212-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 12/15/2016] [Indexed: 01/18/2023]
Abstract
PURPOSE This study aimed to determine whether resveratrol treatment alleviates endoplasmic reticulum stress and changes the expression of adipokines in adipose tissues and cells. METHODS 8-week-old male C57BL/6 mice were fed a high-calorie diet (HCD group) or high-calorie diet supplemented with resveratrol (high-calorie diet + resveratrol group) for 3 months. Insulin resistance, serum lipids and proinflammatory indices, the size and inflammatory cell infiltration in subcutaneous and visceral adipose tissues were analyzed. The gene expressions of endoplasmic reticulum stress, adipokines, and inflammatory cytokines were determined. The induced mature 3T3-L1 cells were pretreated with resveratrol and then palmitic acid, and the gene expressions of endoplasmic reticulum stress, adipokines, and inflammatory cytokines were determined. RESULTS Subcutaneous and visceral adipose tissues in the high-calorie diet-fed mice exhibited adipocyte hypertrophy, inflammatory activation, and endoplasmic reticulum stress. Resveratrol alleviated high-calorie diet-induced insulin resistance and endoplasmic reticulum stress, increased expression of SIRT1, and reversed expression of adipokines in varying degrees in both subcutaneous and visceral adipose tissues. The effects of resveratrol on palmitic acid-treated adipocytes were similar to those shown in the tissues. CONCLUSIONS Resveratrol treatment obviously reversed adipocyte hypertrophy and insulin resistance by attenuating endoplasmic reticulum stress and inflammation, thus increasing the expression of SIRT1 and inverting the expression of adipokines in vivo and in vitro.
Collapse
Affiliation(s)
- Li Chen
- Clinical Laboratory, Anhui Provincial Hospital, Hefei, Anhui, 230001, China
| | - Ting Wang
- Endocrinology Department, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Guanjun Chen
- Department of Chemistry, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Nuojin Wang
- Endocrinology Department, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Li Gui
- The Comprehensive Laboratory, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Fang Dai
- Endocrinology Department, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Zhaohui Fang
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230032, China
| | - Qiu Zhang
- Endocrinology Department, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Yunxia Lu
- The Comprehensive Laboratory, Anhui Medical University, Hefei, Anhui, 230032, China.
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui, 230031, China.
| |
Collapse
|
45
|
Varshney P, Dey CS. Resveratrol regulates neuronal glucose uptake and insulin sensitivity via P21-activated kinase 2 (PAK2). Biochem Biophys Res Commun 2017; 485:372-378. [PMID: 28216158 DOI: 10.1016/j.bbrc.2017.02.070] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 02/13/2017] [Indexed: 02/06/2023]
Abstract
We have recently reported P21-activated kinase 2 (PAK2), a serine/threonine kinase as a negative regulator of neuronal glucose uptake and insulin sensitivity. Resveratrol (RSV), a natural polyphenol with anti-oxidative, anti-inflammatory and anti-diabetic properties, regulates PAK2 activity in HepG2 and ESC-B5 cell apoptosis. However, regulation of PAK2 by RSV in neuronal insulin signaling pathway, if any, is still unknown. In the present study, RSV treatment significantly increased PAK2 activity under insulin-sensitive and insulin-resistant condition, along with a marked decrease in glucose uptake in differentiated N2A cells. Pretreatment with AMPK inhibitor, followed by RSV treatment resulted in reduction in PAK2 activity whereas glucose uptake showed an increase. However, pretreatment with Akt inhibitor and then RSV exposure significantly increased PAK2 activity, with a corresponding decrease in glucose uptake. RSV treatment increased AMPK activity and decreased Akt activity. In conclusion, RSV negatively regulates neuronal glucose uptake and insulin sensitivity via PAK2.
Collapse
Affiliation(s)
- Pallavi Varshney
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi 110016, India.
| |
Collapse
|
46
|
Ding H, Xu X, Qin X, Yang C, Feng Q. Resveratrol promotes differentiation of mouse embryonic stem cells to cardiomyocytes. Cardiovasc Ther 2017; 34:283-9. [PMID: 27225714 DOI: 10.1111/1755-5922.12200] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIM Embryonic stem cells (ESCs) are capable to differentiate into cardiomyocytes, with the potential to treat cardiovascular diseases. However, directed differentiation is still a challenge faced by scientists. As a natural substance in grapes, resveratrol (RV) is important for cardiovascular protection. The studies of RV and its effects on ESC differentiation have potential clinical applications. METHODS Using mouse embryonic stem cells (mESCs), we investigated the effects of different concentrations of RV (5, 10, 20, 50, and 100 μmol/L) exposure on mESCs viability, expression levels of cardiac marker genes in embryoid bodies (EBs) derived from mESCs, expression levels of maturity indicative cardiac markers in cardiomyocytes derived from mESCs, and the beating properties of EBs. RESULTS About 10 μmol/L of RV showed no toxicity on cell viability and was the optimal concentration to promote mESC differentiation, induce mESC differentiation to cardiomyocytes, and gain the beating properties of EBs. CONCLUSION RV can successfully direct the differentiation of mESCs into cardiomyocytes, shedding light on its future applications to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Hong Ding
- Department of Cardiology, Affiliated Hospital of Nanjing Medical University, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu, China
| | - Xin Xu
- Department of Cardiology, Affiliated Hospital of Nanjing Medical University, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu, China
| | - Xian Qin
- Department of Cardiology, Affiliated Hospital of Nanjing Medical University, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu, China
| | - Chengjian Yang
- Department of Cardiology, Affiliated Hospital of Nanjing Medical University, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu, China
| | - Qiuting Feng
- Department of Cardiology, Affiliated Hospital of Nanjing Medical University, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu, China
| |
Collapse
|
47
|
Zou T, Chen D, Yang Q, Wang B, Zhu MJ, Nathanielsz PW, Du M. Resveratrol supplementation of high-fat diet-fed pregnant mice promotes brown and beige adipocyte development and prevents obesity in male offspring. J Physiol 2017; 595:1547-1562. [PMID: 27891610 DOI: 10.1113/jp273478] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/15/2016] [Indexed: 01/01/2023] Open
Abstract
KEY POINTS Maternal high-fat diet impairs brown adipocyte function and correlates with obesity in offspring. Maternal resveratrol administration recovers metabolic activity of offspring brown adipose tissue. Maternal resveratrol promotes beige adipocyte development in offspring white adipose tissue. Maternal resveratrol intervention protects offspring against high-fat diet-induced obesity. ABSTRACT Promoting beige/brite adipogenesis and thermogenic activity is considered as a promising therapeutic approach to reduce obesity and metabolic syndrome. Maternal obesity impairs offspring brown adipocyte function and correlates with obesity in offspring. We previously found that dietary resveratrol (RES) induces beige adipocyte formation in adult mice. Here, we evaluated further the effect of resveratrol supplementation of pregnant mice on offspring thermogenesis and energy expenditure. Female C57BL/6 J mice were fed a control diet (CON) or a high-fat diet (HFD) with or without 0.2% (w/w) RES during pregnancy and lactation. Male offspring were weaned onto a HFD and maintained on this diet for 11 weeks. The offspring thermogenesis and related regulatory factors in adipose tissue were evaluated. At weaning, HFD offspring had lower thermogenesis in brown and white adipose tissues compared with CON offspring, which was recovered by maternal RES supplementation, along with the appearance of multilocular brown/beige adipocytes and elevated thermogenic gene expression. Adult offspring of RES-treated mothers showed increased energy expenditure and insulin sensitivity when on an obesogenic diet compared with HFD offspring. The elevated metabolic activity was correlated with enhanced brown adipose function and white adipose tissue browning in HFD+RES compared with HFD offspring. In conclusion, RES supplementation of HFD-fed dams during pregnancy and lactation promoted white adipose browning and thermogenesis in offspring at weaning accompanied by persistent beneficial effects in protecting against HFD-induced obesity and metabolic disorders.
Collapse
Affiliation(s)
- Tiande Zou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.,Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Daiwen Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Qiyuan Yang
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Bo Wang
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Mei-Jun Zhu
- School of Food Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Peter W Nathanielsz
- Wyoming Pregnancy and Life Course Health Centre, Department of Animal Science, University of Wyoming, Laramie, WY, 82071, USA
| | - Min Du
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, 99164, USA.,Beijing Advanced Innovation Centre for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, 100194, China
| |
Collapse
|
48
|
Lai CS, Wu JC, Ho CT, Pan MH. Chemoprevention of obesity by dietary natural compounds targeting mitochondrial regulation. Mol Nutr Food Res 2016; 61. [DOI: 10.1002/mnfr.201600721] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/02/2016] [Accepted: 11/07/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Ching-Shu Lai
- Department of Seafood Science; National Kaohsiung Marine University; Kaohsiung Taiwan
| | - Jia-Ching Wu
- Institute of Food Science and Technology; National Taiwan University; Taipei Taiwan
| | - Chi-Tang Ho
- Department of Food Science; Rutgers University; New Brunswick NJ USA
| | - Min-Hsiung Pan
- Institute of Food Science and Technology; National Taiwan University; Taipei Taiwan
- Department of Medical Research, China Medical University Hospital; China Medical University; Taichung Taiwan
- Department of Health and Nutrition Biotechnology; Asia University; Taichung Taiwan
| |
Collapse
|
49
|
Kong X, Guan J, Li J, Wei J, Wang R. P66 Shc-SIRT1 Regulation of Oxidative Stress Protects Against Cardio-cerebral Vascular Disease. Mol Neurobiol 2016; 54:5277-5285. [PMID: 27578018 DOI: 10.1007/s12035-016-0073-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/18/2016] [Indexed: 10/21/2022]
Abstract
Growing evidence shows that acute and chronic overproduction of reactive oxygen species (ROS) and increased oxidants under pathophysiologic circumstances are of vital importance in the development of cardio-cerebral vascular diseases (CCVDs). It has been revealed that the impact of ROS can be suppressed by sirtuin 1 (SIRT1), a member of the highly conserved nicotinamide adenine dinucleotide-dependent class III histone deacetylases through protecting endothelial cells from oxidative injury. Plenty of evidences indicate that p66Shc stimulates mitochondrial ROS generation through its oxidoreductase activity and plays a vital role in the pathophysiology of CCVDs. The link between SIRT and p66Shc, though not very clear yet, may be generally illustrated like this: SIRT1 negatively regulates the expression of p66Shc in transcriptional level. In this review, the authors aimed to discuss the link between the pathogenesis of CCVDs, the regulation of ROS, the interrelation between SIRT1 and p66Shc, and the protective effect of the proper regulation of p66Shc/SIRT1 on CCVDs. The imbalance between the elimination and production of ROS can lead to oxidative stress (OS). More and more evidence suggest that ROS pathological overproduction is closely connected to the genesis and growth of CCVDs. P66shc is a gene that controls ROS level, apoptosis induction, and lifespan. Lots of evidence also indicate a role for SIRT1 mediating OS responses through several ways including directly deacetylating some transcription factors that control anti-OS genes. SIRT1 downregulation can lead to a decreased deacetylation of p66shc gene promoter and can then result in p66shc transcription. SIRT1 binds to the promoter of p66Shc where it can deacetylate histone H3, which weakens the transcription and translation of p66shc.
Collapse
Affiliation(s)
- Xiangyi Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, People's Republic of China.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Harvard University, 55 Fruit Street, Boston, MA, 02114-3117, USA
| | - Jian Guan
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Jun Li
- Department of Neurosurgery, Tangshan Gongren Hospital, Hebei Medical University, 27 Wenhua Road, Tangshan, 063000, People's Republic of China
| | - Junji Wei
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, People's Republic of China.
| |
Collapse
|
50
|
Willson CM, Grundmann O. In vitro assays in natural products research - a matter of concentration and relevance to in vivo administration using resveratrol, α-mangostin/γ-mangostin and xanthohumol as examples. Nat Prod Res 2016; 31:492-506. [PMID: 27234135 DOI: 10.1080/14786419.2016.1190721] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Herbal or botanical dietary supplements are an ever increasingly popular category of products in the United States and around the world. In vitro data can provide meaningful insight into the potential target and mechanism of action for a proposed active compound but may also be misused to promote a supplement to consumers with unverified health claims. In vitro data need to be considered alongside pharmacokinetic and pharmacodynamic data in preclinical animal and clinical human trials. While considerable activity of compounds and extracts in vitro may lead to further testing in vivo, in many instances, concentrations tested in cell lines or isolated targets are not achievable at the target site in vivo. Thus, whether the in vitro data are relevant to humans after oral administration is questionable. This review will discuss this discrepancy using in vitro and in vivo data of resveratrol, xanthones (α-mangostin and γ-mangostin) and xanthohumol.
Collapse
Affiliation(s)
- C M Willson
- a Department of Medicinal Chemistry , College of Pharmacy, University of Florida , Gainesville , FL , USA
| | - O Grundmann
- a Department of Medicinal Chemistry , College of Pharmacy, University of Florida , Gainesville , FL , USA
| |
Collapse
|