1
|
Roy S, Ghosh A, Majie A, Karmakar V, Das S, Dinda SC, Bose A, Gorain B. Terpenoids as potential phytoconstituent in the treatment of diabetes: From preclinical to clinical advancement. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155638. [PMID: 38728916 DOI: 10.1016/j.phymed.2024.155638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/21/2024] [Accepted: 04/13/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Diabetes mellitus, a hyperglycemic condition associated with multitudinous organ dysfunction, is a hallmark of the metabolic disorder. This life-threatening condition affects millions of individuals globally, harming them financially, physically and psychologically in the course of therapy. PURPOSES The course therapy for illnesses has undergone ground-breaking transformations due to recent technical advances and insights. Alternatively, the administration of hyperglycemia-reducing agents results in several complications, including severe cardiovascular disease, kidney failure, hepatic problems, and several dermatological conditions. Consideration of alternate diabetic therapy having minimal side effects or no adverse reactions has been driven by such problems. STUDY DESIGN An extensive literature study was conducted in authoritative scientific databases such as PubMed, Scopus, and Web of Science to identify the studies elucidating the bioactivities of terpenoids in diabetic conditions. METHODS Keywords including 'terpenoids', 'monoterpenes', 'diterpenes', 'sesquiterpenes', 'diabetes', 'diabetes mellitus', 'clinical trials', 'preclinical studies', and 'increased blood glucose' were used to identify the relevant research articles. The exclusion criteria, such as English language, duplication, open access, abstract only, and studies not involving preclinical and clinical research, were set. Based on these criteria, 937 relevant articles were selected for further evaluation. RESULTS Triterpenes can serve as therapeutic agents for diabetic retinopathy, peripheral neuropathy, and kidney dysfunction by inhibiting several pathways linked to hyperglycemia and its complications. Therefore, it is essential to draw special attention to these compounds' therapeutic effectiveness and provide scientific professionals with novel data. CONCLUSION This study addressed recent progress in research focussing on mechanisms of terpenoid, its by-products, physiological actions, and therapeutic applications, particularly in diabetic and associated disorders.
Collapse
Affiliation(s)
- Sukanta Roy
- School of Pharmacy, The Neotia University, Diamond Harbour Rd, Sarisha, West Bengal, India
| | - Arya Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Ankit Majie
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Varnita Karmakar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Sourav Das
- School of Pharmacy, The Neotia University, Diamond Harbour Rd, Sarisha, West Bengal, India
| | - Subas Chandra Dinda
- School of Pharmacy, The Neotia University, Diamond Harbour Rd, Sarisha, West Bengal, India
| | - Anirbandeep Bose
- School of Medical Science, Adamas University, Barbaria, Jagannathpur, Kolkata, India.
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India.
| |
Collapse
|
2
|
Skoczyńska A, Ołdakowska M, Dobosz A, Adamiec R, Gritskevich S, Jonkisz A, Lebioda A, Adamiec-Mroczek J, Małodobra-Mazur M, Dobosz T. PPARs in Clinical Experimental Medicine after 35 Years of Worldwide Scientific Investigations and Medical Experiments. Biomolecules 2024; 14:786. [PMID: 39062500 PMCID: PMC11275227 DOI: 10.3390/biom14070786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
This year marks the 35th anniversary of Professor Walter Wahli's discovery of the PPARs (Peroxisome Proliferator-Activated Receptors) family of nuclear hormone receptors. To mark the occasion, the editors of the scientific periodical Biomolecules decided to publish a special issue in his honor. This paper summarizes what is known about PPARs and shows how trends have changed and how research on PPARs has evolved. The article also highlights the importance of PPARs and what role they play in various diseases and ailments. The paper is in a mixed form; essentially it is a review article, but it has been enriched with the results of our experiments. The selection of works was subjective, as there are more than 200,000 publications in the PubMed database alone. First, all papers done on an animal model were discarded at the outset. What remained was still far too large to describe directly. Therefore, only papers that were outstanding, groundbreaking, or simply interesting were described and briefly commented on.
Collapse
Affiliation(s)
- Anna Skoczyńska
- Department of Internal and Occupational Medicine and Hypertension, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Monika Ołdakowska
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| | - Agnieszka Dobosz
- Department of Basic Medical Sciences and Immunology, Division of Basic Medical Sciences, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland
| | - Rajmund Adamiec
- Department of Diabetology and Internal Medicine, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
- Department of Internal Medicine, Faculty of Medical and Technical Sciences, Karkonosze University of Applied Sciences, Lwówiecka 18, 58-506 Jelenia Góra, Poland
| | - Sofya Gritskevich
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| | - Anna Jonkisz
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| | - Arleta Lebioda
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| | - Joanna Adamiec-Mroczek
- Department of Ophthalmology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Małgorzata Małodobra-Mazur
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| | - Tadeusz Dobosz
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| |
Collapse
|
3
|
Bui TT, Aasa J, Abass K, Ågerstrand M, Beronius A, Castro M, Escrivá L, Galizia A, Gliga A, Karlsson O, Whaley P, Yost E, Rudén C. Applying a modified systematic review and integrated assessment framework (SYRINA) - a case study on triphenyl phosphate. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2024; 26:380-399. [PMID: 38205707 PMCID: PMC10879963 DOI: 10.1039/d3em00353a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024]
Abstract
This work presents a case study in applying a systematic review framework (SYRINA) to the identification of chemicals as endocrine disruptors. The suitability and performance of the framework is tested with regard to the widely accepted World Health Organization definition of an endocrine disruptor (ED). The endocrine disrupting potential of triphenyl phosphate (TPP), a well-studied flame retardant reported to exhibit various endocrine related effects was assessed. We followed the 7 steps of the SYRINA framework, articulating the research objective via Populations, Exposures, Comparators, Outcomes (PECO) statements, performed literature search and screening, conducted study evaluation, performed data extraction and summarized and integrated the evidence. Overall, 66 studies, consisting of in vivo, in vitro and epidemiological data, were included. We concluded that triphenyl phosphate could be identified as an ED based on metabolic disruption and reproductive function. We found that the tools used in this case study and the optimizations performed on the framework were suitable to assess properties of EDs. A number of challenges and areas for methodological development in systematic appraisal of evidence relating to endocrine disrupting potential were identified; significant time and effort were needed for the analysis of in vitro mechanistic data in this case study, thus increasing the workload and time needed to perform the systematic review process. Further research and development of this framework with regards to grey literature (non-peer-reviewed literature) search, harmonization of study evaluation methods, more consistent evidence integration approaches and a pre-defined method to assess links between adverse effect and endocrine activity are recommended. It would also be advantageous to conduct more case studies for a chemical with less data than TPP.
Collapse
Affiliation(s)
- Thuy T Bui
- Department of Environmental Science, Stockholm University, Sweden.
| | | | - Khaled Abass
- Department of Environmental Health Sciences, College of Health Sciences, University of Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research (SIMR), University of Sharjah, United Arab Emirates
- Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Finland
| | | | | | - Mafalda Castro
- Section for Environmental Chemistry and Physics, University of Copenhagen, Denmark
| | - Laura Escrivá
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Spain
| | - Audrey Galizia
- United States Environmental Protection Agency, Center for Public Health and Environmental Assessment, USA
| | - Anda Gliga
- Institute of Environmental Medicine, Karolinska Institutet, Sweden
| | - Oskar Karlsson
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, Sweden
| | - Paul Whaley
- Lancaster Environment Centre, Lancaster University, UK
| | - Erin Yost
- United States Environmental Protection Agency, Center for Public Health and Environmental Assessment, USA
| | - Christina Rudén
- Department of Environmental Science, Stockholm University, Sweden.
| |
Collapse
|
4
|
Estrada-Soto S, Ornelas-Mendoza K, Navarrete-Vázquez G, Chávez-Silva F, Almanza-Pérez JC, Villalobos-Molina R, Ortiz-Barragán E, Loza-Rodríguez H, Rivera-Leyva JC, Flores-Flores A, Perea-Arango I, Rodríguez-Carpena JG, Ávila-Villarreal G. Insulin Sensitization by PPARγ and GLUT-4 Overexpression/Translocation Mediates the Antidiabetic Effect of Plantago australis. Pharmaceuticals (Basel) 2023; 16:ph16040535. [PMID: 37111292 PMCID: PMC10143998 DOI: 10.3390/ph16040535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Plantago australis Lam. Subsp. hirtella (Kunth) Rahn is a medicinal plant used as a diuretic, anti-inflammatory, antibacterial, throat cancer treatment and for the control of diabetes. P. australis was collected in the state of Morelos, México. The hydroalcoholic extract (HAEPa) of P. australis was obtained by maceration and concentrated in vacuo. Once dry, it was evaluated through an oral glucose tolerance test (OGTT) in normoglycemic mice and in a non-insulin-dependent diabetic mice model. The expression of PPARγ and GLUT-4 mRNA was determined by rt-PCR, and GLUT-4 translocation was confirmed by confocal microscopy. The toxicological studies were conducted in accordance with the guidelines suggested by the OECD, sections 423 and 407, with some modifications. HAEPa significantly decreased glycemia in OGTT curves, as well as in the experimental diabetes model compared to the vehicle group. In vitro tests showed that HAEPa induced an α-glucosidase inhibition and increased PPARγ and GLUT-4 expression in cell culture. The LD50 of HAEPa was greater than 2000 mg/kg, and sub-chronic toxicity studies revealed that 100 mg/kg/day for 28 days did not generate toxicity. Finally, LC-MS analysis led to the identification of verbascoside, caffeic acid and geniposidic acid, and phytochemical approaches allowed for the isolation of ursolic acid, which showed significant PPARγ overexpression and augmented GLUT-4 translocation. In conclusion, HAEPa induced significant antidiabetic action by insulin sensitization through PPARγ/GLUT-4 overexpression.
Collapse
Affiliation(s)
- Samuel Estrada-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico
| | - Kathia Ornelas-Mendoza
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico
| | | | - Fabiola Chávez-Silva
- Laboratorio de Farmacología, Depto. Ciencias de la Salud, D.C.B.S., Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico 09340, Mexico
| | - Julio Cesar Almanza-Pérez
- Laboratorio de Farmacología, Depto. Ciencias de la Salud, D.C.B.S., Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico 09340, Mexico
| | - Rafael Villalobos-Molina
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Erandi Ortiz-Barragán
- Laboratorio de Farmacología, Depto. Ciencias de la Salud, D.C.B.S., Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico 09340, Mexico
| | - Hilda Loza-Rodríguez
- Laboratorio de Microbiología Experimental, División de Tecnología Ambiental, Universidad Tecnológica de Nezahualcoyotl, Nezahualcoyotl 57000, Mexico
| | | | - Angélica Flores-Flores
- Departamento de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias, Ciudad de Mexico 14080, Mexico
| | - Irene Perea-Arango
- Centro de Investigación en Biotecnología, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico
| | - Javier-German Rodríguez-Carpena
- Centro Nayarita de Innovación y Transferencia de Tecnología "Unidad especializada en I+D+i en Calidad de Alimentos y Productos Naturales", Universidad Autónoma de Nayarit, Tepic 63000, Mexico
| | - Gabriela Ávila-Villarreal
- Centro Nayarita de Innovación y Transferencia de Tecnología "Unidad especializada en I+D+i en Calidad de Alimentos y Productos Naturales", Universidad Autónoma de Nayarit, Tepic 63000, Mexico
- Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic 63000, Mexico
| |
Collapse
|
5
|
Zahr T, Liu L, Chan M, Zhou Q, Cai B, He Y, Aaron N, Accili D, Sun L, Qiang L. PPARγ (Peroxisome Proliferator-Activated Receptor γ) Deacetylation Suppresses Aging-Associated Atherosclerosis and Hypercholesterolemia. Arterioscler Thromb Vasc Biol 2023; 43:30-44. [PMID: 36453279 PMCID: PMC9917767 DOI: 10.1161/atvbaha.122.318061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Atherosclerosis is a medical urgency manifesting at the onset of hypercholesterolemia and is associated with aging. Activation of PPARγ (peroxisome proliferator-activated receptor γ) counteracts metabolic dysfunction influenced by aging, and its deacetylation displays an atheroprotective property. Despite the marked increase of PPARγ acetylation during aging, it is unknown whether PPARγ acetylation is a pathogenic contributor to aging-associated atherosclerosis. METHODS Mice with constitutive deacetylation-mimetic PPARγ mutations on lysine residues K268 and K293 (2KR) in an LDL (low-density lipoprotein)-receptor knockout (Ldlr-/-) background (2KR:Ldlr-/-) were aged for 18 months on a standard laboratory diet to examine the cardiometabolic phenotype, which was confirmed in Western-type diet-fed 2KR:Ldlr+/- mice. Whole-liver RNA-sequencing and in vitro studies in bone marrow-derived macrophages were conducted to decipher the mechanism. RESULTS In contrast to severe atherosclerosis in WT:Ldlr-/- mice, aged 2KR:Ldlr-/- mice developed little to no plaque, which was underlain by a significantly improved plasma lipid profile, with particular reductions in circulating LDL. The protection from hypercholesterolemia was recapitulated in Western-type diet-fed 2KR:Ldlr+/- mice. Liver RNA-sequencing analysis revealed suppression of liver inflammation rather than changes in cholesterol metabolism. This anti-inflammatory effect of 2KR was attributed to polarized M2 activation of macrophages. Additionally, the upregulation of core circadian component Bmal1 (brain and muscle ARNT-like 1), perceived to be involved in anti-inflammatory immunity, was observed in the liver and bone marrow-derived macrophages. CONCLUSIONS PPARγ deacetylation in mice prevents the development of aging-associated atherosclerosis and hypercholesterolemia, in association with the anti-inflammatory phenotype of 2KR macrophages.
Collapse
Affiliation(s)
- Tarik Zahr
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, New York, USA
| | - Longhua Liu
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Michelle Chan
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Qiuzhong Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ying He
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Nicole Aaron
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, New York, USA
| | - Domenico Accili
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Medicine, Columbia University, New York, New York, USA
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Li Qiang
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| |
Collapse
|
6
|
Metre TV, Kodasi B, Bayannavar PK, Bheemayya L, Nadoni VB, Hoolageri SR, Shettar AK, Joshi SD, Kumbar VM, Kamble RR. Coumarin-4-yl‐1,2,3‐triazol‐4-yl-methyl-thiazolidine-2,4-diones: Synthesis, Glucose uptake activity and Cytotoxic Evaluation. Bioorg Chem 2022; 130:106235. [DOI: 10.1016/j.bioorg.2022.106235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/20/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
|
7
|
Wang J, Liu J, Luo M, Cui H, Zhang W, Zhao K, Dai H, Song F, Chen K, Yu Y, Zhou D, Li MJ, Yang H. Rational drug repositioning for coronavirus-associated diseases using directional mapping and side-effect inference. iScience 2022; 25:105348. [PMID: 36267550 PMCID: PMC9556799 DOI: 10.1016/j.isci.2022.105348] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/02/2022] [Accepted: 10/11/2022] [Indexed: 02/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogen of coronavirus disease 2019 (COVID-19), has infected hundreds of millions of people and caused millions of deaths. Looking for valid druggable targets with minimal side effects for the treatment of COVID-19 remains critical. After discovering host genes from multiscale omics data, we developed an end-to-end network method to investigate drug-host gene(s)-coronavirus (CoV) paths and the mechanism of action between the drug and the host factor in a directional network. We also inspected the potential side effect of the candidate drug on several common comorbidities. We established a catalog of host genes associated with three CoVs. Rule-based prioritization yielded 29 Food and Drug Administration (FDA)-approved drugs via accounting for the effects of drugs on CoVs, comorbidities, and drug-target confidence information. Seven drugs are currently undergoing clinical trials as COVID-19 treatment. This catalog of druggable host genes associated with CoVs and the prioritized repurposed drugs will provide a new sight in therapeutics discovery for severe COVID-19 patients.
Collapse
Affiliation(s)
- Jianhua Wang
- Department of Epidemiology and Biostatistics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China,Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jiaojiao Liu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Menghan Luo
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hui Cui
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Wenwen Zhang
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ke Zhao
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hongji Dai
- Department of Epidemiology and Biostatistics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Fangfang Song
- Department of Epidemiology and Biostatistics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Ying Yu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Dongming Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Corresponding author
| | - Mulin Jun Li
- Department of Epidemiology and Biostatistics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China,Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Corresponding author
| | - Hongxi Yang
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Corresponding author
| |
Collapse
|
8
|
Nobushi Y, Wada T, Koike Y, Kaneko H, Shimba S, Uchiyama T, Kishikawa Y. Inhibitory Effects of Hydrolysable Tannins on Lipid Accumulation in 3T3-L1 Cells. Biol Pharm Bull 2022; 45:1458-1465. [PMID: 36184503 DOI: 10.1248/bpb.b22-00227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Obesity is currently the most common cause of metabolic diseases including type 2 diabetes and hyperlipidemia. Obesity results from excess lipid accumulation in adipose tissue. Several studies have investigated the inhibitory effects of natural plant-derived products on adipocyte differentiation and lipid accumulation. In this study, we examined the effect of hydrolysable tannins composed of gallic acid and glucose on adipocyte differentiation in 3T3-L1 cells. 1,2,3,4,6-Penta-O-galloyl-β-D-glucose (PGG) (1), a representative gallotannin, inhibited lipid accumulation in 3T3-L1 cells, whereas ellagitannins (tellimagrandin I, eugeniin and casuarictin) did not. The expression of adipocyte differentiation-related genes, including peroxisome proliferator activator γ2 (Pparγ2), CCAAT/enhancer binding protein α (C/EBPα) and adipocyte fatty acid binding protein (aP2), was significantly suppressed in PGG (1)-treated 3T3-L1 cells beginning at day 2 after induction of differentiation. While PGG (1) did not directly reduce Pparγ2 expression, it reduced the expression of its target genes in mature adipocytes. In addition, PGG (1) treatment inhibited mitotic clonal expansion, one of earliest events of adipocyte differentiation. These findings indicate that PGG (1) has an inhibitory effect on adipocyte differentiation through the suppression of mitotic clonal expansion.
Collapse
|
9
|
Luo L, Liu M. Adiponectin: friend or foe in obesity and inflammation. MEDICAL REVIEW (2021) 2022; 2:349-362. [PMID: 37724325 PMCID: PMC10388816 DOI: 10.1515/mr-2022-0002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/27/2022] [Indexed: 09/20/2023]
Abstract
Adiponectin is an adipokine predominantly produced by fat cells, circulates and exerts insulin-sensitizing, cardioprotective and anti-inflammatory effects. Dysregulation of adiponectin and/or adiponectin signaling is implicated in a number of metabolic diseases such as obesity, insulin resistance, diabetes, and cardiovascular diseases. However, while the insulin-sensitizing and cardioprotective effects of adiponectin have been widely appreciated in the field, the obesogenic and anti-inflammatory effects of adiponectin are still of much debate. Understanding the physiological function of adiponectin is critical for adiponectin-based therapeutics for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Liping Luo
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
10
|
Fisk HL, Childs CE, Miles EA, Ayres R, Noakes PS, Paras-Chavez C, Antoun E, Lillycrop KA, Calder PC. Dysregulation of Subcutaneous White Adipose Tissue Inflammatory Environment Modelling in Non-Insulin Resistant Obesity and Responses to Omega-3 Fatty Acids – A Double Blind, Randomised Clinical Trial. Front Immunol 2022; 13:922654. [PMID: 35958557 PMCID: PMC9358040 DOI: 10.3389/fimmu.2022.922654] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/20/2022] [Indexed: 01/15/2023] Open
Abstract
Background Obesity is associated with enhanced lipid accumulation and the expansion of adipose tissue accompanied by hypoxia and inflammatory signalling. Investigation in human subcutaneous white adipose tissue (scWAT) in people living with obesity in which metabolic complications such as insulin resistance are yet to manifest is limited, and the mechanisms by which these processes are dysregulated are not well elucidated. Long chain omega-3 polyunsaturated fatty acids (LC n-3 PUFAs) have been shown to modulate the expression of genes associated with lipid accumulation and collagen deposition and reduce the number of inflammatory macrophages in adipose tissue from individuals with insulin resistance. Therefore, these lipids may have positive actions on obesity associated scWAT hypertrophy and inflammation. Methods To evaluate obesity-associated tissue remodelling and responses to LC n-3 PUFAs, abdominal scWAT biopsies were collected from normal weight individuals and those living with obesity prior to and following 12-week intervention with marine LC n-3 PUFAs (1.1 g EPA + 0.8 g DHA daily). RNA sequencing, qRT-PCR, and histochemical staining were used to assess remodelling- and inflammatory-associated gene expression, tissue morphology and macrophage infiltration. Results Obesity was associated with scWAT hypertrophy (P < 0.001), hypoxia, remodelling, and inflammatory macrophage infiltration (P = 0.023). Furthermore, we highlight the novel dysregulation of Wnt signalling in scWAT in non-insulin resistant obesity. LC n-3 PUFAs beneficially modulated the scWAT environment through downregulating the expression of genes associated with inflammatory and remodelling pathways (P <0.001), but there were altered outcomes in individuals living with obesity in comparison to normal weight individuals. Conclusion Our data identify dysregulation of Wnt signalling, hypoxia, and hypertrophy, and enhanced macrophage infiltration in scWAT in non-insulin resistant obesity. LC n-3 PUFAs modulate some of these processes, especially in normal weight individuals which may be preventative and limit the development of restrictive and inflammatory scWAT in the development of obesity. We conclude that a higher dose or longer duration of LC n-3 PUFA intervention may be needed to reduce obesity-associated scWAT inflammation and promote tissue homeostasis. Clinical Trial Registration www.isrctn.com, identifier ISRCTN96712688.
Collapse
Affiliation(s)
- Helena L Fisk
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Caroline E Childs
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Elizabeth A Miles
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Robert Ayres
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Paul S Noakes
- School of Medicine, The University of Notre Dame Australia, Freemantle, WA, Australia
| | | | - Elie Antoun
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Karen A Lillycrop
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Philip C Calder
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- National Institute for Health and Care Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton National Health Service (NHS) Foundation Trust and University of Southampton, Southampton, United Kingdom
| |
Collapse
|
11
|
Rhodanine scaffold: A review of antidiabetic potential and structure-activity relationships (SAR). MEDICINE IN DRUG DISCOVERY 2022. [DOI: 10.1016/j.medidd.2022.100131] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
12
|
Fisk HL, Childs CE, Miles EA, Ayres R, Noakes PS, Paras-Chavez C, Kuda O, Kopecký J, Antoun E, Lillycrop KA, Calder PC. Modification of subcutaneous white adipose tissue inflammation by omega-3 fatty acids is limited in human obesity-a double blind, randomised clinical trial. EBioMedicine 2022; 77:103909. [PMID: 35247847 PMCID: PMC8894262 DOI: 10.1016/j.ebiom.2022.103909] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/01/2022] [Accepted: 02/16/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Obesity is associated with enhanced inflammation. However, investigation in human subcutaneous white adipose tissue (scWAT) is limited and the mechanisms by which inflammation occurs have not been well elucidated. Marine long chain omega-3 polyunsaturated fatty acids (LC n-3 PUFAs) have anti-inflammatory actions and may reduce scWAT inflammation. METHODS Subcutaneous white adipose tissue (scWAT) biopsies were collected from individuals living with obesity (n=45) and normal weight individuals (n=39) prior to and following a 12-week intervention with either 3 g/day of a fish oil concentrate (providing 1.1 g eicosapentaenoic acid (EPA) + 0.8 g docosahexaenoic acid (DHA)) or 3 g/day of corn oil. ScWAT fatty acid, oxylipin, and transcriptome profiles were assessed by gas chromatography, ultra-pure liquid chromatography tandem mass spectrometry, RNA sequencing and qRT-PCR, respectively. FINDINGS Obesity was associated with greater scWAT inflammation demonstrated by lower concentrations of specialised pro-resolving mediators (SPMs) and hydroxy-DHA metabolites and an altered transcriptome with differential expression of genes involved in LC n-3 PUFA activation, oxylipin synthesis, inflammation, and immune response. Intervention with LC n-3 PUFAs increased their respective metabolites including the SPM precursor 14-hydroxy-DHA in normal weight individuals and decreased arachidonic acid derived metabolites and expression of genes involved in immune and inflammatory response with a greater effect in normal weight individuals. INTERPRETATION Downregulated expression of genes responsible for fatty acid activation and metabolism may contribute to an inflammatory oxylipin profile and limit the effects of LC n-3 PUFAs in obesity. There may be a need for personalised LC n-3 PUFA supplementation based on obesity status. FUNDING European Commission Seventh Framework Programme (Grant Number 244995) and Czech Academy of Sciences (Lumina quaeruntur LQ200111901).
Collapse
Affiliation(s)
- Helena L Fisk
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, IDS Building, MP887, Tremona Road, Southampton SO16 6YD, United Kingdom.
| | - Caroline E Childs
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, IDS Building, MP887, Tremona Road, Southampton SO16 6YD, United Kingdom
| | - Elizabeth A Miles
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, IDS Building, MP887, Tremona Road, Southampton SO16 6YD, United Kingdom
| | - Robert Ayres
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, IDS Building, MP887, Tremona Road, Southampton SO16 6YD, United Kingdom
| | - Paul S Noakes
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, IDS Building, MP887, Tremona Road, Southampton SO16 6YD, United Kingdom; Medical School, University of Notre Dame Australia, Fremantle, Australia
| | - Carolina Paras-Chavez
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, IDS Building, MP887, Tremona Road, Southampton SO16 6YD, United Kingdom
| | - Ondrej Kuda
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Kopecký
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Elie Antoun
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, IDS Building, MP887, Tremona Road, Southampton SO16 6YD, United Kingdom
| | - Karen A Lillycrop
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, IDS Building, MP887, Tremona Road, Southampton SO16 6YD, United Kingdom; School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, IDS Building, MP887, Tremona Road, Southampton SO16 6YD, United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, United Kingdom
| |
Collapse
|
13
|
Torres JL, Usategui-Martín R, Hernández-Cosido L, Bernardo E, Manzanedo-Bueno L, Hernández-García I, Mateos-Díaz AM, Rozo O, Matesanz N, Salete-Granado D, Chamorro AJ, Carbonell C, Garcia-Macia M, González-Sarmiento R, Sabio G, Muñoz-Bellvís L, Marcos M. PPAR-γ Gene Expression in Human Adipose Tissue Is Associated with Weight Loss After Sleeve Gastrectomy. J Gastrointest Surg 2022; 26:286-297. [PMID: 34882294 PMCID: PMC8821495 DOI: 10.1007/s11605-021-05216-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/25/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND The peroxisome proliferator-activated receptor (PPAR)-γ plays a key role in adipose tissue differentiation and fat metabolism. However, it is unclear which factors may regulate its expression and whether obese patients have changes in adipose tissue expression of PPAR-γor potential regulators such as miR-27. Thus, our aims were to analyze PPAR-γ and miR-27 expression in adipose tissue of obese patients, and to correlate their levels with clinical variables. SUBJECTS AND METHODS We included 43 morbidly obese subjects who underwent sleeve gastrectomy (31 of them completed 1-year follow-up) and 19 non-obese subjects. mRNA expression of PPAR-γ1 and PPAR-γ2, miR-27a, and miR-27b was measured by qPCR in visceral and subcutaneous adipose tissue. Clinical variables and serum adipokine and hormone levels were correlated with PPAR-γ and miR-27 expression. In addition, a systematic review of the literature regarding PPAR-γ expression in adipose tissue of obese patients was performed. RESULTS We found no differences in the expression of PPAR-γ and miR-27 in adipose tissue of obese patients vs. controls. The literature review revealed discrepant results regarding PPAR-γ expression in adipose tissue of obese patients. Of note, we described a significant negative correlation between pre-operative PPAR-γ1 expression in adipose tissue of obese patients and post-operative weight loss, potentially linked with insulin resistance markers. CONCLUSION PPAR-γ1 expression in adipose tissue is associated with weight loss after sleeve gastrectomy and may be used as a biomarker for response to surgery.
Collapse
Affiliation(s)
- Jorge-Luis Torres
- Department of Internal Medicine, University Hospital of Salamanca-SACYL-IBSAL, Salamanca, Spain ,Department of Internal Medicine, Complejo Asistencial de Zamora-SACYL, Zamora, Spain
| | - Ricardo Usategui-Martín
- IOBA, University of Valladolid, Valladolid, Spain ,Cooperative Health Network for Research (RETICS), Oftared, National Institute of Health Carlos III, ISCIII, Madrid, Spain
| | - Lourdes Hernández-Cosido
- Bariatric Surgery Unit, Department of General and Gastrointestinal Surgery, University Hospital of Salamanca, Salamanca, Spain ,University of Salamanca, Salamanca, Spain
| | - Edgar Bernardo
- Department of Vascular Biology and Inflammation, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | - Laura Manzanedo-Bueno
- Department of Internal Medicine, University Hospital of Salamanca-SACYL-IBSAL, Salamanca, Spain ,Department of Internal Medicine, Complejo Asistencial de Zamora-SACYL, Zamora, Spain
| | - Ignacio Hernández-García
- Department of Preventive Medicine and Public Health, Lozano Blesa University Clinical Hospital of Zaragoza, Zaragoza, Spain
| | - Ana-María Mateos-Díaz
- Department of Internal Medicine, University Hospital of Salamanca-SACYL-IBSAL, Salamanca, Spain
| | - Orlando Rozo
- Department of Surgery, Complejo Asistencial de Ávila-SACYL, Ávila, Spain
| | - Nuria Matesanz
- Department of Vascular Biology and Inflammation, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | | | - Antonio-Javier Chamorro
- Department of Internal Medicine, University Hospital of Salamanca-SACYL-IBSAL, Salamanca, Spain ,University of Salamanca, Salamanca, Spain
| | - Cristina Carbonell
- Department of Internal Medicine, University Hospital of Salamanca-SACYL-IBSAL, Salamanca, Spain ,University of Salamanca, Salamanca, Spain
| | - Marina Garcia-Macia
- Department of Internal Medicine, University Hospital of Salamanca-SACYL-IBSAL, Salamanca, Spain ,Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain ,Centro de Investigación Biomédica en Red Sobre Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Guadalupe Sabio
- Department of Vascular Biology and Inflammation, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | - Luis Muñoz-Bellvís
- Department of General and Gastrointestinal Surgery, Hospital Universitario de Salamanca, Biomedical Research Institute of Salamanca (IBSAL), Universidad de Salamanca, Salamanca, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca-SACYL-IBSAL, Salamanca, Spain ,University of Salamanca, Salamanca, Spain
| |
Collapse
|
14
|
Rato L, Sousa ACA. The Impact of Endocrine-Disrupting Chemicals in Male Fertility: Focus on the Action of Obesogens. J Xenobiot 2021; 11:163-196. [PMID: 34940512 PMCID: PMC8709303 DOI: 10.3390/jox11040012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/12/2021] [Accepted: 11/23/2021] [Indexed: 12/11/2022] Open
Abstract
The current scenario of male infertility is not yet fully elucidated; however, there is increasing evidence that it is associated with the widespread exposure to endocrine-disrupting chemicals (EDCs), and in particular to obesogens. These compounds interfere with hormones involved in the regulation of metabolism and are associated with weight gain, being also able to change the functioning of the male reproductive axis and, consequently, the testicular physiology and metabolism that are pivotal for spermatogenesis. The disruption of these tightly regulated metabolic pathways leads to adverse reproductive outcomes. The permanent exposure to obesogens has raised serious health concerns. Evidence suggests that obesogens are one of the leading causes of the marked decline of male fertility and key players in shaping the future health outcomes not only for those who are directly exposed but also for upcoming generations. In addition to the changes that lead to inefficient functioning of the male gametes, obesogens induce alterations that are “imprinted” on the genes of the male gametes, establishing a link between generations and contributing to the transmission of defects. Unveiling the molecular mechanisms by which obesogens induce toxicity that may end-up in epigenetic modifications is imperative. This review describes and discusses the suggested molecular targets and potential mechanisms for obesogenic–disrupting chemicals and the subsequent effects on male reproductive health.
Collapse
Affiliation(s)
- Luís Rato
- Health School of the Polytechnic Institute of Guarda, 6300-035 Guarda, Portugal
- Correspondence: (L.R.); (A.C.A.S.)
| | - Ana C. A. Sousa
- Department of Biology, School of Science and Technology, University of Évora, 7006-554 Évora, Portugal
- Comprehensive Health Research Centre (CHRC), University of Évora, 7000-671 Évora, Portugal
- Correspondence: (L.R.); (A.C.A.S.)
| |
Collapse
|
15
|
Dixit G, Prabhu A. The pleiotropic peroxisome proliferator activated receptors: Regulation and therapeutics. Exp Mol Pathol 2021; 124:104723. [PMID: 34822814 DOI: 10.1016/j.yexmp.2021.104723] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
The Peroxisome proliferator-activated receptors (PPARs) are key regulators of metabolic events in our body. Owing to their implication in maintenance of homeostasis, both PPAR agonists and antagonists assume therapeutic significance. Understanding the molecular mechanisms of each of the PPAR isotypes in the healthy body and during disease is crucial to exploiting their full therapeutic potential. This article is an attempt to present a rational analysis of the multifaceted therapeutic effects and underlying mechanisms of isotype-specific PPAR agonists, dual PPAR agonists, pan PPAR agonists as well as PPAR antagonists. A holistic understanding of the mechanistic dimensions of these key metabolic regulators will guide future efforts to identify novel molecules in the realm of metabolic, inflammatory and immunotherapeutic diseases.
Collapse
Affiliation(s)
- Gargi Dixit
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.
| |
Collapse
|
16
|
Kee Z, Ong SM, Heng CK, Ooi DSQ. Androgen-dependent tissue factor pathway inhibitor regulating protein: a review of its peripheral actions and association with cardiometabolic diseases. J Mol Med (Berl) 2021; 100:185-196. [PMID: 34797389 DOI: 10.1007/s00109-021-02160-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 02/07/2023]
Abstract
The first genome-wide association study on coronary artery disease (CAD) in the Han Chinese population identified C6orf105 as a susceptibility gene. The C6orf105 gene was later found to encode for a protein that regulates tissue factor pathway inhibitor (TFPI) expression in endothelial cells in an androgen-dependent manner, and the novel protein was thus termed androgen-dependent TFPI-regulating protein (ADTRP). Since the identification of ADTRP, there have been several studies associating genetic variants on the ADTRP gene with CAD risk, as well as research providing mechanistic insights on this novel protein and its functional role. ADTRP is a membrane protein, whose expression is upregulated by androgen, GATA-binding protein 2, oxidized low-density lipoprotein, peroxisome proliferator-activated receptors, and low-density lipoprotein receptors. ADTRP regulates multiple downstream targets involved in coagulation, inflammation, endothelial function, and vascular integrity. In addition, ADTRP functions as a fatty acid esters of hydroxy fatty acid (FAHFA)-specific hydrolase that is involved in energy metabolism. Current evidence suggests that ADTRP may play a role in the pathogenesis of atherosclerosis, CAD, obesity, and metabolic disorders. This review summarizes the current literature on ADTRP, with a focus on the peripheral actions of ADTRP, including expression, genetic variations, signaling pathways, and function. The evidence linking ADTRP and cardiometabolic diseases will also be discussed.
Collapse
Affiliation(s)
- Zizheng Kee
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block Level 12, 1E Kent Ridge Road, 119228, Singapore
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Kent Ridge, Singapore
| | - Sze Min Ong
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block Level 12, 1E Kent Ridge Road, 119228, Singapore
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Kent Ridge, Singapore
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block Level 12, 1E Kent Ridge Road, 119228, Singapore
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Kent Ridge, Singapore
| | - Delicia Shu Qin Ooi
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block Level 12, 1E Kent Ridge Road, 119228, Singapore.
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Kent Ridge, Singapore.
| |
Collapse
|
17
|
Dysregulation of endocannabinoid concentrations in human subcutaneous adipose tissue in obesity and modulation by omega-3 polyunsaturated fatty acids. Clin Sci (Lond) 2021; 135:185-200. [PMID: 33393630 DOI: 10.1042/cs20201060] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/01/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Obesity is believed to be associated with a dysregulated endocannabinoid system which may reflect enhanced inflammation. However, reports of this in human white adipose tissue (WAT) are limited and inconclusive. Marine long-chain omega-3 polyunsaturated fatty acids (LC n-3 PUFAs) have anti-inflammatory actions and therefore may improve obesity-associated adipose tissue inflammation. Therefore, fatty acid (FA) concentrations, endocannabinoid concentrations, and gene expression were assessed in subcutaneous WAT (scWAT) biopsies from healthy normal weight individuals (BMI 18.5-25 kg/m2) and individuals living with metabolically healthy obesity (BMI 30-40 kg/m2) prior to and following a 12-week intervention with 3 g fish oil/day (1.1 g eicosapentaenoic acid (EPA) + 0.8 g DHA) or 3 g corn oil/day (placebo). WAT from individuals living with metabolically healthy obesity had higher n-6 PUFAs and EPA, higher concentrations of two endocannabinoids (anandamide (AEA) and eicosapentaenoyl ethanolamide (EPEA)), higher expression of phospholipase A2 Group IID (PLA2G2D) and phospholipase A2 Group IVA (PLA2G4A), and lower expression of CNR1. In response to fish oil intervention, WAT EPA increased to a similar extent in both BMI groups, and WAT DHA increased by a greater extent in normal weight individuals. WAT EPEA and docosahexaenoyl ethanolamide (DHEA) increased in normal weight individuals only and WAT 2-arachidonyl glycerol (2-AG) decreased in individuals living with metabolically healthy obesity only. Altered WAT fatty acid, endocannabinoid, and gene expression profiles in metabolically healthy obesity at baseline may be linked. WAT incorporates n-3 PUFAs when their intake is increased which affects the endocannabinoid system; however, effects appear greater in normal weight individuals than in those living with metabolically healthy obesity.
Collapse
|
18
|
Woo M, Kim M. Insulin sensitization causes accelerated sinus nodal dysfunction through autophagic dysregulation in hypertensive mice. Transl Clin Pharmacol 2021; 29:92-106. [PMID: 34235122 PMCID: PMC8255547 DOI: 10.12793/tcp.2021.29.e9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 12/22/2022] Open
Abstract
Insulin sensitizers, while effective in glucose-lowering for diabetes control, are linked to an increased risk of heart disease through mechanisms that are not well understood. In this study, we investigated the molecular mechanisms underlying the effects of insulin sensitization on cardiac sinus node dysfunction. We used pharmacologic or genetic approaches to enhance insulin sensitivity, by treating with pioglitazone or rosiglitazone, or through phosphatase and tensin homolog (PTEN) deletion in cardiomyocytes respectively. We employed an angiotensin II (Ang II)-induced hypertensive animal model which causes sinus node dysfunction and accumulation of oxidized calcium/calmodulin-dependent protein kinase II (CaMKII), which also serves as a biomarker for this defect. While neither PTEN deficiency nor insulin sensitizers caused sinus node dysfunction in normotensive mice, both accelerated the onset of sinus node dysfunction and CaMKII oxidation in hypertensive mice. These abnormalities were accompanied by a significant defect in autophagy as revealed by unc-51 like autophagy activating kinase 1 (ULK1) signaling. Indeed, mice deficient in ulk1 in cardiomyocytes and the sinus node also showed early onset of slow atrial impulse conduction with frequent sinus pauses and upregulated CaMKII oxidation following Ang II infusion similar to that seen with PTEN deficiency, or treatment with insulin sensitizers. To further elucidate the role of autophagy in sinus node dysfunction, we treated mice with a peptide D-Tat-beclin1 that enhanced autophagy, which significantly abrogated the frequent sinus pauses and accumulation of oxidized CaMKII induced by insulin sensitizers treatment, or PTEN deficiency in hypertensive animals. Together, these findings provide clear evidence of the detrimental cardiac effects of insulin sensitization that occurs through failure of autophagy-mediated proteolytic clearance.
Collapse
Affiliation(s)
- Minna Woo
- Toronto General Research Institute and Division of Endocrinology and Metabolism, Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario M5S, Canada
| | - Minsuk Kim
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| |
Collapse
|
19
|
Dwivedi PSR, Rasal VP, Kotharkar E, Nare S, Khanal P. Gene set enrichment analysis of PPAR-γ regulators from Murraya odorata Blanco. J Diabetes Metab Disord 2021; 20:369-375. [PMID: 34178844 PMCID: PMC8212318 DOI: 10.1007/s40200-021-00754-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/24/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptor gamma (PPAR-γ) is reported to regulate insulin sensitivity and progression of Type 2 diabetes mellitus (T2DM). Hence the present study is aimed to identify PPAR-γ regulators from Murraya odorata Blanco and predict their role to manage T2DM. METHODS Multiple in-silico tools and databases like SwissTargetPrediction, ADVERPred, PubChem, and MolSoft, were used to retrieve the information related to bioactives, targets, druglikeness character, and probable side effects as applicable. Similarly, the Kyoto Encyclopedia of Genes and Genomes (KEGG) database was used to identify the regulated pathways. Further, the bioactives-protein-pathways network interaction was constructed using Cytoscape. Finally, molecular docking was performed using Autodock4. RESULTS Twenty-five bioactives were shortlisted in which six were predicted as PPAR-γ modulators. Among them, stigmasterol was predicted to possess the best binding affinity towards PPAR-γ and possessed no side effects. Similarly, n-hexadecanoic acid was predicted to modulate the highest number of proteins, and protein CD14 was targeted by the highest number of bioactives. Further, the PI3K-Akt pathway was predicted as the maximum modulated genes. CONCLUSIONS The anti-diabetic property of the Murraya odorata Blanco of fruit pulp may be due to the presence of n-hexadecanoic acid and stigmasterol; may also involve in the regulation of the PI3K-Akt pathway which needs further investigated by in-vitro and in-vivo protocols.
Collapse
Affiliation(s)
- Prarambh SR Dwivedi
- Department of Pharmacology and Toxicology, KLE College of Pharmacy Belagavi, KLE Academy of Higher Education and Research (KAHER), Belagavi, 590010 India
| | - V. P. Rasal
- Department of Pharmacology and Toxicology, KLE College of Pharmacy Belagavi, KLE Academy of Higher Education and Research (KAHER), Belagavi, 590010 India
| | - Ekta Kotharkar
- Department of Pharmacology and Toxicology, KLE College of Pharmacy Belagavi, KLE Academy of Higher Education and Research (KAHER), Belagavi, 590010 India
| | - Shailaja Nare
- Department of Pharmacology and Toxicology, KLE College of Pharmacy Belagavi, KLE Academy of Higher Education and Research (KAHER), Belagavi, 590010 India
| | - Pukar Khanal
- Department of Pharmacology and Toxicology, KLE College of Pharmacy Belagavi, KLE Academy of Higher Education and Research (KAHER), Belagavi, 590010 India
| |
Collapse
|
20
|
Lopez-Perez D, Redruello-Romero A, Garcia-Rubio J, Arana C, Garcia-Escudero LA, Tamayo F, Puentes-Pardo JD, Moreno-SanJuan S, Salmeron J, Blanco A, Galvez J, Leon J, Carazo Á. In Patients With Obesity, the Number of Adipose Tissue Mast Cells Is Significantly Lower in Subjects With Type 2 Diabetes. Front Immunol 2021; 12:664576. [PMID: 34093556 PMCID: PMC8177010 DOI: 10.3389/fimmu.2021.664576] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes (T2D) is a rising global health problem mainly caused by obesity and a sedentary lifestyle. In healthy individuals, white adipose tissue (WAT) has a relevant homeostatic role in glucose metabolism, energy storage, and endocrine signaling. Mast cells contribute to these functions promoting WAT angiogenesis and adipogenesis. In patients with T2D, inflammation dramatically impacts WAT functioning, which results in the recruitment of several leukocytes, including monocytes, that enhance this inflammation. Accordingly, the macrophages population rises as the WAT inflammation increases during the T2D status worsening. Since mast cell progenitors cannot arrive at WAT, the amount of WAT mast cells depends on how the new microenvironment affects progenitor and differentiated mast cells. Here, we employed a flow cytometry-based approach to analyze the number of mast cells from omental white adipose tissue (o-WAT) and subcutaneous white adipose tissue (s-WAT) in a cohort of 100 patients with obesity. Additionally, we measured the number of mast cell progenitors in a subcohort of 15 patients. The cohort was divided in three groups: non-T2D, pre-T2D, and T2D. Importantly, patients with T2D have a mild condition (HbA1c <7%). The number of mast cells and mast cell progenitors was lower in patients with T2D in both o-WAT and s-WAT in comparison to subjects from the pre-T2D and non-T2D groups. In the case of mast cells in o-WAT, there were statistically significant differences between non-T2D and T2D groups (p = 0.0031), together with pre-T2D and T2D groups (p=0.0097). However, in s-WAT, the differences are only between non-T2D and T2D groups (p=0.047). These differences have been obtained with patients with a mild T2D condition. Therefore, little changes in T2D status have a huge impact on the number of mast cells in WAT, especially in o-WAT. Due to the importance of mast cells in WAT physiology, their decrease can reduce the capacity of WAT, especially o-WAT, to store lipids and cause hypoxic cell deaths that will trigger inflammation.
Collapse
Affiliation(s)
- David Lopez-Perez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain.,Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Anaïs Redruello-Romero
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | | | - Carlos Arana
- Endocrinology and Nutrition Unit, Virgen de las Nieves University Hospital, Granada, Spain
| | - Luis A Garcia-Escudero
- Department of Statistics and Operative Research, Faculty of Sciences, University of Valladolid, Valladolid, Spain
| | | | - Jose D Puentes-Pardo
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain.,Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Sara Moreno-SanJuan
- Cytometry and Microscopy Research Service, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Javier Salmeron
- Gastroenterology Unit, San Cecilio University Hospital, Granada, Spain
| | - Armando Blanco
- Department of Computer Science and Artificial Intelligence, University of Granada, Granada, Spain
| | - Julio Galvez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain.,Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain.,Centro de Investigación Biomédica En Red para Enfermedades Hepáticas y Digestivas (CIBER-EHD), Center for Biomedical Research, University of Granada, Granada, Spain
| | - Josefa Leon
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Ángel Carazo
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain.,Clinical Management Unit of Digestive Disease, San Cecilio University Hospital, Granada, Spain
| |
Collapse
|
21
|
Noruddin NAA, Hamzah MF, Rosman Z, Salin NH, Shu-Chien AC, Muhammad TST. Natural Compound 3β,7β,25-trihydroxycucurbita-5,23(E)-dien-19-al from Momordica charantia Acts as PPARγ Ligand. Molecules 2021; 26:2682. [PMID: 34063700 PMCID: PMC8124227 DOI: 10.3390/molecules26092682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/22/2021] [Accepted: 04/25/2021] [Indexed: 12/11/2022] Open
Abstract
Momordica charantia is a popular vegetable associated with effective complementary and alternative diabetes management in some parts of the world. However, the molecular mechanism is less commonly investigated. In this study, we investigated the association between a major cucurbitane triterpenoid isolated from M. charantia, 3β,7β,25-trihydroxycucurbita-5,23(E)-dien-19-al (THCB) and peroxisome proliferator activated receptor gamma (PPARγ) activation and its related activities using cell culture and molecular biology techniques. In this study, we report on both M. charantia fruit crude extract and THCB in driving the luciferase activity of Peroxisome Proliferator Response Element, associated with PPARγ activation. Other than that, THCB also induced adipocyte differentiation at far less intensity as compared to the full agonist rosiglitazone. In conjunction, THCB treatment on adipocytes also resulted in upregulation of PPAR gamma target genes expression; AP2, adiponectin, LPL and CD34 at a lower magnitude compared to rosiglitazone's induction. THCB also induced glucose uptake into muscle cells and the mechanism is via Glut4 translocation to the cell membrane. In conclusion, THCB acts as one of the many components in M. charantia to induce hypoglycaemic effect by acting as PPARγ ligand and inducing glucose uptake activity in the muscles by means of Glut4 translocation.
Collapse
Affiliation(s)
- Nur Adelina Ahmad Noruddin
- National Institutes of Biotechnology Malaysia-Malaysian Institute of Pharmaceuticals and Nutraceuticals (NIBM-IPharm), Ministry of Science, Technology and Innovation, Blok 5A, Halaman Bukit Gambir 11700, Malaysia; (N.A.A.N.); (M.F.H.); (Z.R.); (N.H.S.)
| | - Mohamad Faiz Hamzah
- National Institutes of Biotechnology Malaysia-Malaysian Institute of Pharmaceuticals and Nutraceuticals (NIBM-IPharm), Ministry of Science, Technology and Innovation, Blok 5A, Halaman Bukit Gambir 11700, Malaysia; (N.A.A.N.); (M.F.H.); (Z.R.); (N.H.S.)
| | - Zulfadli Rosman
- National Institutes of Biotechnology Malaysia-Malaysian Institute of Pharmaceuticals and Nutraceuticals (NIBM-IPharm), Ministry of Science, Technology and Innovation, Blok 5A, Halaman Bukit Gambir 11700, Malaysia; (N.A.A.N.); (M.F.H.); (Z.R.); (N.H.S.)
| | - Nurul Hanim Salin
- National Institutes of Biotechnology Malaysia-Malaysian Institute of Pharmaceuticals and Nutraceuticals (NIBM-IPharm), Ministry of Science, Technology and Innovation, Blok 5A, Halaman Bukit Gambir 11700, Malaysia; (N.A.A.N.); (M.F.H.); (Z.R.); (N.H.S.)
| | - Alexander Chong Shu-Chien
- School of Biological Sciences, Universiti Sains Malaysia, Gelugor 11800, Malaysia;
- Centre for Chemical Biology, Universiti Sains Malaysia, Sains@USM, Blok B No. 10, Persiaran Bukit Jambul, Bayan Lepas 11900, Malaysia
| | | |
Collapse
|
22
|
Dixon ED, Nardo AD, Claudel T, Trauner M. The Role of Lipid Sensing Nuclear Receptors (PPARs and LXR) and Metabolic Lipases in Obesity, Diabetes and NAFLD. Genes (Basel) 2021; 12:genes12050645. [PMID: 33926085 PMCID: PMC8145571 DOI: 10.3390/genes12050645] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are metabolic disorders characterized by metabolic inflexibility with multiple pathological organ manifestations, including non-alcoholic fatty liver disease (NAFLD). Nuclear receptors are ligand-dependent transcription factors with a multifaceted role in controlling many metabolic activities, such as regulation of genes involved in lipid and glucose metabolism and modulation of inflammatory genes. The activity of nuclear receptors is key in maintaining metabolic flexibility. Their activity depends on the availability of endogenous ligands, like fatty acids or oxysterols, and their derivatives produced by the catabolic action of metabolic lipases, most of which are under the control of nuclear receptors. For example, adipose triglyceride lipase (ATGL) is activated by peroxisome proliferator-activated receptor γ (PPARγ) and conversely releases fatty acids as ligands for PPARα, therefore, demonstrating the interdependency of nuclear receptors and lipases. The diverse biological functions and importance of nuclear receptors in metabolic syndrome and NAFLD has led to substantial effort to target them therapeutically. This review summarizes recent findings on the roles of lipases and selected nuclear receptors, PPARs, and liver X receptor (LXR) in obesity, diabetes, and NAFLD.
Collapse
Affiliation(s)
| | | | | | - Michael Trauner
- Correspondence: ; Tel.: +43-140-4004-7410; Fax: +43-14-0400-4735
| |
Collapse
|
23
|
Saleh HA, Yousef MH, Abdelnaser A. The Anti-Inflammatory Properties of Phytochemicals and Their Effects on Epigenetic Mechanisms Involved in TLR4/NF-κB-Mediated Inflammation. Front Immunol 2021; 12:606069. [PMID: 33868227 PMCID: PMC8044831 DOI: 10.3389/fimmu.2021.606069] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Innate immune response induces positive inflammatory transducers and regulators in order to attack pathogens, while simultaneously negative signaling regulators are transcribed to maintain innate immune homeostasis and to avoid persistent inflammatory immune responses. The gene expression of many of these regulators is controlled by different epigenetic modifications. The remarkable impact of epigenetic changes in inducing or suppressing inflammatory signaling is being increasingly recognized. Several studies have highlighted the interplay of histone modification, DNA methylation, and post-transcriptional miRNA-mediated modifications in inflammatory diseases, and inflammation-mediated tumorigenesis. Targeting these epigenetic alterations affords the opportunity of attenuating different inflammatory dysregulations. In this regard, many studies have identified the significant anti-inflammatory properties of distinct naturally-derived phytochemicals, and revealed their regulatory capacity. In the current review, we demonstrate the signaling cascade during the immune response and the epigenetic modifications that take place during inflammation. Moreover, we also provide an updated overview of phytochemicals that target these mechanisms in macrophages and other experimental models, and go on to illustrate the effects of these phytochemicals in regulating epigenetic mechanisms and attenuating aberrant inflammation.
Collapse
Affiliation(s)
- Haidy A. Saleh
- Department of Chemistry, School of Sciences and Engineering, The American University in Cairo, Cairo, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Mohamed H. Yousef
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, Cairo, Egypt
| | - Anwar Abdelnaser
- Institute of Global Public Health, School of Sciences and Engineering, The American University in Cairo, Cairo, Egypt
| |
Collapse
|
24
|
Mohajer N, Du CY, Checkcinco C, Blumberg B. Obesogens: How They Are Identified and Molecular Mechanisms Underlying Their Action. Front Endocrinol (Lausanne) 2021; 12:780888. [PMID: 34899613 PMCID: PMC8655100 DOI: 10.3389/fendo.2021.780888] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 10/23/2021] [Indexed: 12/11/2022] Open
Abstract
Adult and childhood obesity have reached pandemic level proportions. The idea that caloric excess and insufficient levels of physical activity leads to obesity is a commonly accepted answer for unwanted weight gain. This paradigm offers an inconclusive explanation as the world continually moves towards an unhealthier and heavier existence irrespective of energy balance. Endocrine disrupting chemicals (EDCs) are chemicals that resemble natural hormones and disrupt endocrine function by interfering with the body's endogenous hormones. A subset of EDCs called obesogens have been found to cause metabolic disruptions such as increased fat storage, in vivo. Obesogens act on the metabolic system through multiple avenues and have been found to affect the homeostasis of a variety of systems such as the gut microbiome and adipose tissue functioning. Obesogenic compounds have been shown to cause metabolic disturbances later in life that can even pass into multiple future generations, post exposure. The rising rates of obesity and related metabolic disease are demanding increasing attention on chemical screening efforts and worldwide preventative strategies to keep the public and future generations safe. This review addresses the most current findings on known obesogens and their effects on the metabolic system, the mechanisms of action through which they act upon, and the screening efforts through which they were identified with. The interplay between obesogens, brown adipose tissue, and the gut microbiome are major topics that will be covered.
Collapse
Affiliation(s)
- Nicole Mohajer
- Deparment of Pharmaceutical Sciences, University of California, Irvine, CA, United States
| | - Chrislyn Y. Du
- Deparment of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Christian Checkcinco
- Deparment of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Bruce Blumberg
- Deparment of Pharmaceutical Sciences, University of California, Irvine, CA, United States
- Deparment of Developmental and Cell Biology, University of California, Irvine, CA, United States
- Deparment of Biomedical Engineering, University of California, Irvine, CA, United States
- *Correspondence: Bruce Blumberg,
| |
Collapse
|
25
|
R M, Mani S, Sali VK, Bhardwaj M, Vasanthi HR. Macrotyloma uniflorum a plant food alleviates the metabolic syndrome through modulation of adipokines and PPARs. J Food Biochem 2020; 45:e13595. [PMID: 33368458 DOI: 10.1111/jfbc.13595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/06/2020] [Accepted: 11/30/2020] [Indexed: 11/27/2022]
Abstract
A sedentary lifestyle combined with the intake of high-calorie diet has been the paramount cause of metabolic syndrome (MS) which is now a serious concern of public health worldwide as it involves the coexistence of hypertension, hyperlipidemia, glucose intolerance, and obesity. Hence, identifying a suitable strategy to overcome the worldwide menace of MS is imperative. Macrotyloma uniflorum a lesser known legume is highly nutritious and notable for its ethano-medicinal potential. Herein, the influence of M. uniflorum in high-fat dietinduced metabolic changes in a rodent model of metabolic syndrome was evaluated. Serum levels of glucose, total cholesterol, triglycerides, VLDL-c, and bodyweight were decreased, whereas HDL-c was increased in M. uniflorum-treated MS rats. The protein expression (AMPK-α, PPAR-α, and PPAR-γ) and gene expression (leptin, adiponectin, resistin, UCP2, NF-κB, and IL-6) results are impressive to highlight that M. uniflorum modulates the pathological conditions of MS and proves to be cardioprotective. Furthermore, the histopathological analysis confirmed the pathological changes and substantiates the influence of M. uniflorum to overcome MS. The HPLC and GC (MS) profiling reveals the presence of an array of polyphenols such as rutin (694.61 μg/g), catechin (500.12 μg/g), epicatechin (158.10 μg/g), gallic acid (17.98 μg/g), ferulic acid (10.911 μg/g), daidzein (6.51 μg/g), and PUFA, respectively, which probably exhibits the therapeutic effect on MS and associated complications by modulating lipid metabolism and adipogenesis. PRACTICAL APPLICATIONS: Metabolic disorders like CVD and diabetes are leading cause of mortality and morbidity worldwide. With emerging issues on adverse effects of modern drugs, the emphasis on "Food is Medicine and Medicine as Food" has taken dramatic dimensions in the healthcare sector. Therefore, nutraceuticals are in great demand in the developed world off late. Legumes, are potent elements in a balanced diet next to cereals. Exploring the medicinal properties of legumes could bring a revolution in public health and nutraceutical industries. This study scientifically validated the phytochemicals in M. uniflorum for its functional potential in the management of Metabolic Syndrome (MS). This study would help the nutraceutical industries to develop functional foods using M. uniflorum seeds to make porridges and soups or nutraceutical supplements with the bioflavonoids isolated from M. uniflorum for the management of metabolic disorders by mitigating hyperlipidemia, oxidative stress, and inflammation.
Collapse
Affiliation(s)
- Malarvizhi R
- Natural Products Research Laboratory, Department of Biotechnology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Sugumar Mani
- Natural Products Research Laboratory, Department of Biotechnology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Veeresh K Sali
- Natural Products Research Laboratory, Department of Biotechnology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Meenakshi Bhardwaj
- Natural Products Research Laboratory, Department of Biotechnology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Hannah R Vasanthi
- Natural Products Research Laboratory, Department of Biotechnology, School of Life Sciences, Pondicherry University, Puducherry, India
| |
Collapse
|
26
|
Peroxisome Proliferator-Activated Receptors as Molecular Links between Caloric Restriction and Circadian Rhythm. Nutrients 2020; 12:nu12113476. [PMID: 33198317 PMCID: PMC7696073 DOI: 10.3390/nu12113476] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
The circadian rhythm plays a chief role in the adaptation of all bodily processes to internal and environmental changes on the daily basis. Next to light/dark phases, feeding patterns constitute the most essential element entraining daily oscillations, and therefore, timely and appropriate restrictive diets have a great capacity to restore the circadian rhythm. One of the restrictive nutritional approaches, caloric restriction (CR) achieves stunning results in extending health span and life span via coordinated changes in multiple biological functions from the molecular, cellular, to the whole-body levels. The main molecular pathways affected by CR include mTOR, insulin signaling, AMPK, and sirtuins. Members of the family of nuclear receptors, the three peroxisome proliferator-activated receptors (PPARs), PPARα, PPARβ/δ, and PPARγ take part in the modulation of these pathways. In this non-systematic review, we describe the molecular interconnection between circadian rhythm, CR-associated pathways, and PPARs. Further, we identify a link between circadian rhythm and the outcomes of CR on the whole-body level including oxidative stress, inflammation, and aging. Since PPARs contribute to many changes triggered by CR, we discuss the potential involvement of PPARs in bridging CR and circadian rhythm.
Collapse
|
27
|
Guo F, Xu S, Zhu Y, Zheng X, Lu Y, Tu J, He Y, Jin L, Li Y. PPARγ Transcription Deficiency Exacerbates High-Fat Diet-Induced Adipocyte Hypertrophy and Insulin Resistance in Mice. Front Pharmacol 2020; 11:1285. [PMID: 32973516 PMCID: PMC7466717 DOI: 10.3389/fphar.2020.01285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/03/2020] [Indexed: 12/29/2022] Open
Abstract
Background The transcriptional factor peroxisome proliferator–activated receptor γ (PPARγ) is an important therapeutic target for the treatment of type 2 diabetes. However, the role of the PPARγ transcriptional activity remains ambiguous in its metabolic regulation. Methods Based on the crystal structure of PPARγ bound with the DNA target of PPARγ response element (PPRE), Arg134, Arg135, and Arg138, three crucial DNA binding sites for PPARγ, were mutated to alanine (3RA), respectively. In vitro AlphaScreen assay and cell-based reporter assay validated that PPARγ 3RA mutant cannot bind with PPRE and lost transcriptional activity, while can still bind ligand (rosiglitazone) and cofactors (SRC1, SRC2, and NCoR). By using CRISPR/Cas9, we created mice that were heterozygous for PPARγ-3RA (PPARγ3RA/+). The phenotypes of chow diet and high-fat diet fed PPARγ3RA/+ mice were investigated, and the molecular mechanism were analyzed by assessing the PPARγ transcriptional activity. Results Homozygous PPARγ-3RA mutant mice are embryonically lethal. The mRNA levels of PPARγ target genes were significantly decreased in PPARγ3RA/+ mice. PPARγ3RA/+ mice showed more severe adipocyte hypertrophy, insulin resistance, and hepatic steatosis than wild type mice when fed with high-fat diet. These phenotypes were ameliorated after the transcription activity of PPARγ was restored by rosiglitazone, a PPARγ agonist. Conclusion The current report presents a novel mouse model for investigating the role of PPARγ transcription in physiological functions. The data demonstrate that the transcriptional activity plays an indispensable role for PPARγ in metabolic regulation.
Collapse
Affiliation(s)
- Fusheng Guo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Shuangshuang Xu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yanlin Zhu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xing Zheng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yi Lu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jui Tu
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Ying He
- Laboratory Animal Center, Xiamen University, Xiamen, China
| | - Lihua Jin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China.,Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Yong Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
28
|
Peroxisome Proliferator-Activated Receptors and Caloric Restriction-Common Pathways Affecting Metabolism, Health, and Longevity. Cells 2020; 9:cells9071708. [PMID: 32708786 PMCID: PMC7407644 DOI: 10.3390/cells9071708] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Caloric restriction (CR) is a traditional but scientifically verified approach to promoting health and increasing lifespan. CR exerts its effects through multiple molecular pathways that trigger major metabolic adaptations. It influences key nutrient and energy-sensing pathways including mammalian target of rapamycin, Sirtuin 1, AMP-activated protein kinase, and insulin signaling, ultimately resulting in reductions in basic metabolic rate, inflammation, and oxidative stress, as well as increased autophagy and mitochondrial efficiency. CR shares multiple overlapping pathways with peroxisome proliferator-activated receptors (PPARs), particularly in energy metabolism and inflammation. Consequently, several lines of evidence suggest that PPARs might be indispensable for beneficial outcomes related to CR. In this review, we present the available evidence for the interconnection between CR and PPARs, highlighting their shared pathways and analyzing their interaction. We also discuss the possible contributions of PPARs to the effects of CR on whole organism outcomes.
Collapse
|
29
|
Prabhu DS, Rajeswari VD. PPAR-Gamma as putative gene target involved in Butein mediated anti-diabetic effect. Mol Biol Rep 2020; 47:5273-5283. [PMID: 32642916 DOI: 10.1007/s11033-020-05605-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/20/2020] [Indexed: 12/30/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder caused due to varied genetic and lifestyle factors. The search for a potential natural compound to enhance the treatment of diabetes is the need of the hour. Butein, a flavonoid, found sufficiently in Faba bean, is said to possess an anti-diabetic property. In-silico analysis, Butein is predicted as a potential anti-diabetic compound, due to its regulatory action on PPAR-Gamma. Based on this evidence, the Butein's anti-diabetic action is studied in diabetic induced rat models. The drug property of Butein is studied through in-silico analysis to determine the metabolic properties. In animal models, the biochemical analysis, histopathological and gene expression against PPAR-Gamma were studied comparatively. Butein being a hydrophobic compound, the bioavailability is said to be minimum. Hence, Butein formulation was made using biopolymer Chitosan for the synergistic anti-diabetic action. The Butein Chitosan formulation was optimized and characterized using analytical techniques. Further, the anti-diabetic activity of Butein and Butein Chitosan formulation was studied in diabetic induced rats. The obtained in-silico analysis results showed that Butein is the most favorable drug. Apparently, in the rat model, Butein and Butein Chitosan formulation effectively controlled the blood glucose levels without any side effects. The histopathological observations of the tissue samples showed nontoxic activity. Additionally, the gene expression analysis predicted the possible mechanism of anti-diabetic action exhibited through the down regulation of PPAR-Gamma. Whereas, the Butein Chitosan formulation failed, to show synergetic anti-diabetic activity as expected. This study is vital in introducing Butein as a safe anti-diabetic compound, which can be used in the treatment of T2DM.
Collapse
Affiliation(s)
- D Sathya Prabhu
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu, 632 014, India
| | - V Devi Rajeswari
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu, 632 014, India.
| |
Collapse
|
30
|
Ramaiahgari SC, Auerbach SS, Saddler TO, Rice JR, Dunlap PE, Sipes NS, DeVito MJ, Shah RR, Bushel PR, Merrick BA, Paules RS, Ferguson SS. The Power of Resolution: Contextualized Understanding of Biological Responses to Liver Injury Chemicals Using High-throughput Transcriptomics and Benchmark Concentration Modeling. Toxicol Sci 2020; 169:553-566. [PMID: 30850835 DOI: 10.1093/toxsci/kfz065] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Prediction of human response to chemical exposures is a major challenge in both pharmaceutical and toxicological research. Transcriptomics has been a powerful tool to explore chemical-biological interactions, however, limited throughput, high-costs, and complexity of transcriptomic interpretations have yielded numerous studies lacking sufficient experimental context for predictive application. To address these challenges, we have utilized a novel high-throughput transcriptomics (HTT) platform, TempO-Seq, to apply the interpretive power of concentration-response modeling with exposures to 24 reference compounds in both differentiated and non-differentiated human HepaRG cell cultures. Our goals were to (1) explore transcriptomic characteristics distinguishing liver injury compounds, (2) assess impacts of differentiation state of HepaRG cells on baseline and compound-induced responses (eg, metabolically-activated), and (3) identify and resolve reference biological-response pathways through benchmark concentration (BMC) modeling. Study data revealed the predictive utility of this approach to identify human liver injury compounds by their respective BMCs in relation to human internal exposure plasma concentrations, and effectively distinguished drug analogs with varied associations of human liver injury (eg, withdrawn therapeutics trovafloxacin and troglitazone). Impacts of cellular differentiation state (proliferated vs differentiated) were revealed on baseline drug metabolizing enzyme expression, hepatic receptor signaling, and responsiveness to metabolically-activated toxicants (eg, cyclophosphamide, benzo(a)pyrene, and aflatoxin B1). Finally, concentration-response modeling enabled efficient identification and resolution of plausibly-relevant biological-response pathways through their respective pathway-level BMCs. Taken together, these findings revealed HTT paired with differentiated in vitro liver models as an effective tool to model, explore, and interpret toxicological and pharmacological interactions.
Collapse
Affiliation(s)
- Sreenivasa C Ramaiahgari
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Scott S Auerbach
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Trey O Saddler
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Julie R Rice
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Paul E Dunlap
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Nisha S Sipes
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Michael J DeVito
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Ruchir R Shah
- Sciome, LLC, Research Triangle Park, Durham, North Carolina 27709
| | - Pierre R Bushel
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709.,Division of Intramural Research, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Bruce A Merrick
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Richard S Paules
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| | - Stephen S Ferguson
- *Biomolecular Screening Branch, Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, North Carolina 27709
| |
Collapse
|
31
|
Evans LW, Stratton MS, Ferguson BS. Dietary natural products as epigenetic modifiers in aging-associated inflammation and disease. Nat Prod Rep 2020; 37:653-676. [PMID: 31993614 PMCID: PMC7577396 DOI: 10.1039/c9np00057g] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Covering: up to 2020Chronic, low-grade inflammation is linked to aging and has been termed "inflammaging". Inflammaging is considered a key contributor to the development of metabolic dysfunction and a broad spectrum of diseases or disorders including declines in brain and heart function. Genome-wide association studies (GWAS) coupled with epigenome-wide association studies (EWAS) have shown the importance of diet in the development of chronic and age-related diseases. Moreover, dietary interventions e.g. caloric restriction can attenuate inflammation to delay and/or prevent these diseases. Common themes in these studies entail the use of phytochemicals (plant-derived compounds) or the production of short chain fatty acids (SCFAs) as epigenetic modifiers of DNA and histone proteins. Epigenetic modifications are dynamically regulated and as such, serve as potential therapeutic targets for the treatment or prevention of age-related disease. In this review, we will focus on the role for natural products that include phytochemicals and short chain fatty acids (SCFAs) as regulators of these epigenetic adaptations. Specifically, we discuss regulators of methylation, acetylation and acylation, in the protection from chronic inflammation driven metabolic dysfunction and deterioration of neurocognitive and cardiac function.
Collapse
Affiliation(s)
- Levi W Evans
- Department of Nutrition, University of Nevada, Reno, NV 89557, USA.
| | | | | |
Collapse
|
32
|
Long Noncoding RNA lnc-HC Regulates PPARγ-Mediated Hepatic Lipid Metabolism through miR-130b-3p. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:954-965. [PMID: 31770672 PMCID: PMC6881655 DOI: 10.1016/j.omtn.2019.10.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/12/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is due to the excessive lipid accumulation within hepatocytes. Metabolic nuclear receptors (MNRs) play great roles in lipid homeostasis. We have identified a novel long noncoding RNA (lncRNA), lnc-HC, which regulates hepatocytic cholesterol metabolism through reducing Cyp7a1 and Abca1 expression. Here, we further elucidate its roles in hepatic fatty acid and triglyceride (TG) metabolism through a novel lncRNA regulatory mechanism. The most prominent target of lnc-HC identified by in vitro study is PPARγ. Further studies revealed that lnc-HC negatively regulates PPARγ at both the mRNA and protein levels and suppresses hepatocytic lipid droplet formation. Importantly, the function of lnc-HC in regulating PPARγ expression depends on modulating miR-130b-3p expression from the transcriptional to the post-transcriptional level, not through lncRNA’s critical modulating patterns. In vivo, the reduction of lnc-HC expression significantly decreases miR-130b-3p expression, induces PPARγ expression, and increases TG concentration in rat livers with hyperlipidemia. These findings further help in understanding the regulatory pattern of lnc-HC in hepatic lipid metabolism and might present a possible therapeutic target for improving lipid homeostasis.
Collapse
|
33
|
Physiological and pathophysiological aspects of peroxisome proliferator-activated receptor regulation by fatty acids in poultry species. WORLD POULTRY SCI J 2019. [DOI: 10.1017/s0043933916000490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
34
|
Brierley DI, Harman JR, Giallourou N, Leishman E, Roashan AE, Mellows BA, Bradshaw HB, Swann JR, Patel K, Whalley BJ, Williams CM. Chemotherapy-induced cachexia dysregulates hypothalamic and systemic lipoamines and is attenuated by cannabigerol. J Cachexia Sarcopenia Muscle 2019; 10:844-859. [PMID: 31035309 PMCID: PMC6711413 DOI: 10.1002/jcsm.12426] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 02/19/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Muscle wasting, anorexia, and metabolic dysregulation are common side-effects of cytotoxic chemotherapy, having a dose-limiting effect on treatment efficacy, and compromising quality of life and mortality. Extracts of Cannabis sativa, and analogues of the major phytocannabinoid Δ9-tetrahydrocannabinol, have been used to ameliorate chemotherapy-induced appetite loss and nausea for decades. However, psychoactive side-effects limit their clinical utility, and they have little efficacy against weight loss. We recently established that the non-psychoactive phytocannabinoid cannabigerol (CBG) stimulates appetite in healthy rats, without neuromotor side-effects. The present study assessed whether CBG attenuates anorexia and/or other cachectic effects induced by the broad-spectrum chemotherapy agent cisplatin. METHODS An acute cachectic phenotype was induced in adult male Lister-hooded rats by 6 mg/kg (i.p.) cisplatin. In total 66 rats were randomly allocated to groups receiving vehicle only, cisplatin only, or cisplatin and 60 or 120 mg/kg CBG (po, b.i.d.). Feeding behavior, bodyweight and locomotor activity were recorded for 72 hours, at which point rats were sacrificed for post-mortem analyses. Myofibre atrophy, protein synthesis and autophagy dysregulation were assessed in skeletal muscle, plasma metabolic profiles were obtained by untargeted 1H-NMR metabonomics, and levels of endocannabinoid-like lipoamines quantified in plasma and hypothalami by targeted HPLC-MS/MS lipidomics. RESULTS CBG (120 mg/kg) modestly increased food intake, predominantly at 36-60hrs (p<0.05), and robustly attenuated cisplatin-induced weight loss from 6.3% to 2.6% at 72hrs (p<0.01). Cisplatin-induced skeletal muscle atrophy was associated with elevated plasma corticosterone (3.7 vs 13.1ng/ml, p<0.01), observed selectively in MHC type IIx (p<0.05) and IIb (p<0.0005) fibres, and was reversed by pharmacological rescue of dysregulated Akt/S6-mediated protein synthesis and autophagy processes. Plasma metabonomic analysis revealed cisplatin administration produced a wide-ranging aberrant metabolic phenotype (Q2Ŷ=0.5380, p=0.001), involving alterations to glucose, amino acid, choline and lipid metabolism, citrate cycle, gut microbiome function, and nephrotoxicity, which were partially normalized by CBG treatment (Q2Ŷ=0.2345, p=0.01). Lipidomic analysis of hypothalami and plasma revealed extensive cisplatin-induced dysregulation of central and peripheral lipoamines (29/79 and 11/26 screened, respectively), including reversible elevations in systemic N-acyl glycine concentrations which were negatively associated with the anti-cachectic effects of CBG treatment. CONCLUSIONS Endocannabinoid-like lipoamines may have hitherto unrecognized roles in the metabolic side-effects associated with chemotherapy, with the N-acyl glycine subfamily in particular identified as a potential therapeutic target and/or biomarker of anabolic interventions. CBG-based treatments may represent a novel therapeutic option for chemotherapy-induced cachexia, warranting investigation in tumour-bearing cachexia models.
Collapse
Affiliation(s)
- Daniel I. Brierley
- School of Psychology and Clinical Language SciencesUniversity of ReadingBerkshireUK
- School of PharmacyUniversity of ReadingBerkshireUK
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | - Joe R. Harman
- School of Biological SciencesUniversity of ReadingBerkshireUK
| | | | - Emma Leishman
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonINUSA
| | | | | | - Heather B. Bradshaw
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonINUSA
| | - Jonathan R. Swann
- Division of Computational and Systems MedicineImperial College LondonLondonUK
| | - Ketan Patel
- School of Biological SciencesUniversity of ReadingBerkshireUK
| | | | - Claire M. Williams
- School of Psychology and Clinical Language SciencesUniversity of ReadingBerkshireUK
| |
Collapse
|
35
|
El-Zahabi MA, Elbendary ER, Bamanie FH, Radwan MF, Ghareib SA, Eissa IH. Design, synthesis, molecular modeling and anti-hyperglycemic evaluation of phthalimide-sulfonylurea hybrids as PPARγ and SUR agonists. Bioorg Chem 2019; 91:103115. [PMID: 31310882 DOI: 10.1016/j.bioorg.2019.103115] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 06/29/2019] [Accepted: 07/09/2019] [Indexed: 10/26/2022]
Abstract
New series of phthalimide-sulfonylurea hybrids were prepared and examined for their in vivo anti-hyperglycemic activities in STZ-induced hyperglycemic rats using glibenclamide as a reference drug. Compounds 6c, 6d, 6g, 6h, 6j and 6k induced significant reduction in the blood glucose levels of diabetic rats ranging from 24.43 to 21.43%. Moreover, molecular docking and pharmacophore approaches were carried out to examine binding modes and fit values of the prepared compounds against PPARγ and SUR, respectively. Compounds 6c, 6d, 6j and 6m exhibited the highest binding free energies against PPARγ. Compounds 6c, 6j, 6k, 6l, and 6n showed the highest fit values against the generated pharmacophore model. Also, QSAR technique was carried out to estimate the proposed PPARγ binding affinities and insulin-secreting abilities. The synthesized compounds showed promising estimated activities. In-silico ADMET studies were performed to investigate pharmacokinetics of the synthesized compounds. They showed considerable human intestinal absorption with low BBB penetration.
Collapse
Affiliation(s)
- Mohamed Ayman El-Zahabi
- Medicinal Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| | - Eman R Elbendary
- Medicinal Chemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Faida H Bamanie
- Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed F Radwan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Salah A Ghareib
- Pharmacology Department, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ibrahim H Eissa
- Medicinal Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| |
Collapse
|
36
|
Jangam SS, Wankhede SB. Synthesis, Molecular Docking, and Biological Evaluation of the New Hybrids of 4-Thiazolidinone and 4(3H)-Quinazolinone Against Streptozotocin Induced Diabetic Rats. RUSS J GEN CHEM+ 2019. [DOI: 10.1134/s1070363219050256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
37
|
Seok H, Lee M, Shin E, Yun MR, Lee YH, Moon JH, Kim E, Lee PH, Lee BW, Kang ES, Lee HC, Cha BS. Low-dose pioglitazone can ameliorate learning and memory impairment in a mouse model of dementia by increasing LRP1 expression in the hippocampus. Sci Rep 2019; 9:4414. [PMID: 30867485 PMCID: PMC6416325 DOI: 10.1038/s41598-019-40736-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 02/21/2019] [Indexed: 01/07/2023] Open
Abstract
Amyloid-β (Aβ) accumulation in the brain is a pathological feature of Alzheimer's disease (AD) and enhancing Aβ clearance is a potential therapeutic strategy. Pioglitazone is a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist and is widely used to treat type 2 diabetes. We previously reported that low-dose pioglitazone increased the expression of low-density lipoprotein receptor-related protein 1 (LRP1), which upregulates the clearance of Aβ, using human brain microvascular endothelial cells. We investigated whether low-dose pioglitazone can rescue the pathological phenotype and memory impairment in senescence-accelerated mouse prone-8 (SAMP8) mice by increasing LRP1 levels. SAMP8 mice were treated with vehicle or pioglitazone in dosages of 2 or 5 mg/kg/day for 7 weeks. In the water maze test, 2 mg/kg/day of pioglitazone significantly attenuated the increased escape latency in SAMP8 mice (p = 0.026), while 5 mg/kg/day of treatment did not. Compared with vehicle treatment, the hippocampi of SAMP8 mice with 2 mg/kg/day of pioglitazone exhibited fewer Aβ deposits and reduced Aβ1-40 levels, along with elevated LRP1 expression (p = 0.005). Collectively, our results proposed that a new therapeutic application of the PPAR-γ agonist for AD treatment should be considered at a lower dose than the conventional dose used to treat diabetes.
Collapse
Affiliation(s)
- Hannah Seok
- Department of Internal Medicine, The Catholic University of Korea College of Medicine, Uijeongbu St. Mary's Hospital, Uijeongbu, Korea
| | - Minyoung Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Eugene Shin
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Mi Ra Yun
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Yong-Ho Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Hoon Moon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Korea
| | - Eosu Kim
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Byung-Wan Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Seok Kang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Chul Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Bong Soo Cha
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea. .,Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
38
|
Tiwari P, Saxena A, Gupta N, Medicherla KM, Suravajhala P, Mathur SK. Systems Genomics of Thigh Adipose Tissue From Asian Indian Type-2 Diabetics Revealed Distinct Protein Interaction Hubs. Front Genet 2019; 9:679. [PMID: 30671081 PMCID: PMC6331691 DOI: 10.3389/fgene.2018.00679] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 12/07/2018] [Indexed: 12/13/2022] Open
Abstract
We performed a systematic analysis of genes implicated in thigh subcutaneous adipose tissue of Asian Indian Type 2 Diabetes Mellitus (AIT2DM) and created a phenome-interactome network. This analysis was performed on 60 subjects specific to limb thigh fat by integrating phenotypic traits and similarity scores associated with AIT2DM. Using a phenotypic attribute, a contextual neighbor was identified across all the traits, viz. body mass index (BMI) statistics, adipocyte size, lipid parameters, homeostatic model assessment- insulin resistance (HOMA-IR), HOMA-ß. In this work, we have attempted to characterize transcription signatures using the phenome-interactome maps where each of the traits under study including the intermediary phenotypes has a distinct set of genes forming the hubs. Furthermore, we have identified various clinical, biochemical, and radiological parameters which show significant correlation with distinct hubs. We observed a number of novel pathways and genes including those that are non-coding RNAs implicated in AIT2DM.We showed that they appear to be associated with pathways, viz. tyrosine kinase JAK2, NOTCH thereby recruiting signaling molecules such as STAT5 and Src family kinases on the cell surface regulated them and our analyses comprising significant hubs suggest that thigh subcutaneous adipose tissue plays a role in pathophysiology of AIT2DM.
Collapse
Affiliation(s)
- Pradeep Tiwari
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Jaipur, India.,Department of Endocrinology, Sawai Man Singh Medical College and Hospital, Jaipur, India.,Department of Chemistry, School of Basic Sciences, Manipal University Jaipur, Jaipur, India
| | - Aditya Saxena
- Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University, Mathura, India
| | - Nidhi Gupta
- Department of Biotechnology, The IIS University, Jaipur, India
| | - Krishna Mohan Medicherla
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Jaipur, India
| | - Prashanth Suravajhala
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Jaipur, India
| | - Sandeep Kumar Mathur
- Department of Chemistry, School of Basic Sciences, Manipal University Jaipur, Jaipur, India
| |
Collapse
|
39
|
Feinberg T, Wieland LS, Miller LE, Munir K, Pollin TI, Shuldiner AR, Amoils S, Gallagher L, Bahr-Robertson M, D'Adamo CR. Polyherbal dietary supplementation for prediabetic adults: study protocol for a randomized controlled trial. Trials 2019; 20:24. [PMID: 30616613 PMCID: PMC6323847 DOI: 10.1186/s13063-018-3032-6] [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] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 11/01/2018] [Indexed: 12/15/2022] Open
Abstract
Background Prediabetes describes a state of hyperglycemia outside of normal limits that does not meet the criteria for diabetes diagnosis, is generally symptomless, and affects an estimated 38% of adults in the United States. Prediabetes typically precedes the diagnosis of type 2 diabetes, which accounts for increased morbidity and mortality. Although the use of dietary and herbal supplements is popular worldwide, and a variety of single herbal medicines have been examined for glycemic management, the potential of increasingly common polyherbal formulations to return glycemic parameters to normal ranges among adults with prediabetes remains largely unexplored. The purpose of this study is to evaluate the efficacy of a commercially available, polyherbal dietary supplement on glycemic and lipid parameters in prediabetic individuals. Methods In this multi-site, double-blinded, randomized controlled clinical trial, 40 participants with prediabetes will be randomized to either a daily oral polyherbal dietary supplement (GlucoSupreme™ Herbal; Designs for Health®, Suffield, CT, USA; containing cinnamon bark (Cinnamomum cassia), banaba leaf (Lagerstroemia speciosa standardized to 1% corosolic acid), kudzu root (Pueraria lobata standardized to 40% isoflavones), fenugreek seed (Trigonella foenum-graceum standardized to 60% saponins), gymnema leaf (Gymnema sylvestre standardized to 25% gymnemic acid), American ginseng root (Panax quinquefolius standardized to 5% ginsenosides), and berberine HCl derived from bark (Berberis aristata)) or placebo for 12 weeks. Short-, medium-, and comparatively long-term markers of glycemic control (blood glucose and fasting insulin, fructosamine, and glycated hemoglobin/A1c, respectively), and other glycemic parameters (GlycoMark, β-cell function, and insulin sensitivity/resistance) will be obtained. Lipid profile (total cholesterol, LDL, HDL, and triglycerides), inflammation (hs-CRP), progression to type 2 diabetes mellitus, as well as safety indices (ALT, AST) will be obtained. An intention-to-treat analysis will be used to assess changes in study outcomes. Discussion Treatment options for adults with prediabetes are currently limited. This study aims to evaluate the safety and efficacy of a commercially available dietary supplement in the popular, but as yet insufficiently studied, category of polyherbal formulas for the management of glycemic parameters and other biomarkers associated with prediabetes. Trial registration ClinicalTrials.gov, ID: NCT03388762. Retrospectively registered on 4 January 2018. Electronic supplementary material The online version of this article (10.1186/s13063-018-3032-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Termeh Feinberg
- University of Maryland School of Medicine Center for Integrative Medicine, Baltimore, MD, USA. .,Yale University School of Medicine Center for Medical Informatics, New Haven, CT, USA.
| | - L Susan Wieland
- University of Maryland School of Medicine Center for Integrative Medicine, Baltimore, MD, USA
| | | | - Kashif Munir
- University of Maryland School of Medicine Center for Diabetes and Endocrinology, Baltimore, MD, USA
| | - Toni I Pollin
- University of Maryland School of Medicine Department of Medicine, Baltimore, MD, USA
| | - Alan R Shuldiner
- University of Maryland School of Medicine Department of Medicine, Baltimore, MD, USA
| | - Steve Amoils
- Alliance Integrative Medicine, Cincinatti, OH, USA
| | | | - Mary Bahr-Robertson
- University of Maryland School of Medicine Center for Integrative Medicine, Baltimore, MD, USA
| | - Christopher R D'Adamo
- University of Maryland School of Medicine Center for Integrative Medicine, Baltimore, MD, USA
| |
Collapse
|
40
|
González FB, Villar SR, Toneatto J, Pacini MF, Márquez J, D’Attilio L, Bottasso OA, Piwien-Pilipuk G, Pérez AR. Immune response triggered by Trypanosoma cruzi infection strikes adipose tissue homeostasis altering lipid storage, enzyme profile and adipokine expression. Med Microbiol Immunol 2018; 208:651-666. [DOI: 10.1007/s00430-018-0572-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/30/2018] [Indexed: 12/11/2022]
|
41
|
Young AI, Wauthier FL, Donnelly P. Identifying loci affecting trait variability and detecting interactions in genome-wide association studies. Nat Genet 2018; 50:1608-1614. [PMID: 30323177 DOI: 10.1038/s41588-018-0225-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 08/03/2018] [Indexed: 11/09/2022]
Abstract
Identification of genetic variants with effects on trait variability can provide insights into the biological mechanisms that control variation and can identify potential interactions. We propose a two-degree-of-freedom test for jointly testing mean and variance effects to identify such variants. We implement the test in a linear mixed model, for which we provide an efficient algorithm and software. To focus on biologically interesting settings, we develop a test for dispersion effects, that is, variance effects not driven solely by mean effects when the trait distribution is non-normal. We apply our approach to body mass index in the subsample of the UK Biobank population with British ancestry (n ~408,000) and show that our approach can increase the power to detect associated loci. We identify and replicate novel associations with significant variance effects that cannot be explained by the non-normality of body mass index, and we provide suggestive evidence for a connection between leptin levels and body mass index variability.
Collapse
Affiliation(s)
- Alexander I Young
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK. .,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.
| | - Fabian L Wauthier
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.,Department of Statistics, University of Oxford, Oxford, UK
| | - Peter Donnelly
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK. .,Department of Statistics, University of Oxford, Oxford, UK.
| |
Collapse
|
42
|
Li J, Liu YP. The roles of PPARs in human diseases. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2018; 37:361-382. [PMID: 30036119 DOI: 10.1080/15257770.2018.1475673] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs), as members of nuclear hormone receptor superfamily, can be activated by binding natural or synthetic ligands. The use of related ligands has revealed many potential roles for PPARs in the pathogenesis of some human metabolic disorders and inflammatory-related disease. Based on the previous studies, this review primarily concluded the current progress of knowledge regarding the specific biological activity of PPARs in cancers, atherosclerosis, and type 2 diabetes mellitus, providing a foundation for the potential therapeutic use of PPAR ligands in human diseases.
Collapse
Affiliation(s)
- Jingjing Li
- a Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province , Sichuan Agricultural University , Chengdu , China
| | - Yi-Ping Liu
- a Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province , Sichuan Agricultural University , Chengdu , China
| |
Collapse
|
43
|
Prabhu DS, Rajeswari VD. In vitro and in silico analyses of Vicia faba L. on Peroxisome proliferator-activated receptor gamma. J Cell Biochem 2018; 119:7729-7737. [PMID: 29923224 DOI: 10.1002/jcb.27123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 05/07/2018] [Indexed: 01/08/2023]
Abstract
The agonists of peroxisome proliferator-activated receptor gamma (PPARγ) from natural victual products were used as antidiabetic agents. Faba bean (Vicia faba L.) is a consequential legume that was known to possess potential antidiabetic activity, whose mechanism of action was unknown. The current study was focused to ascertain gene expression of the nuclear receptor PPARγ by Faba bean pod extract in rat cell lines (RINm5F).The real-time polymerase chain reaction analysis demonstrated that Faba bean pod extract in concentrations of 160 µg/mL have shown 4.97-fold stimulation compared with control. The cells treated with 320 µg/mL has shown 5.89-fold upregulation, respectively. Furthermore, in silico docking analysis was carried out against PPARγ, using the bioactive compounds identified from Faba bean pod extracts, which were known reported compounds from the literature. The results suggest that gene expression of PPARγ was inhibited by the constituents in Faba bean. In silico analysis prognosticates, butein has a high binding energy (-8.6 kcal/mol) with an atomic contact energy of -214.10, followed by Apigenin and Quercetin against PPARγ. Similarly, the percentage of interaction was high for butein, followed by Apigenin and Quercetin than other compounds comparatively. Hence, the results conclude inhibition of PPARγ by the bioactive compounds from Faba bean, which may provide insights into developing future therapeutic molecules for diabetes mellitus.
Collapse
Affiliation(s)
- D Sathya Prabhu
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu, India
| | - V Devi Rajeswari
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu, India
| |
Collapse
|
44
|
Kim SH, Park HS, Hong MJ, Hur HJ, Kwon DY, Kim MS. Caffeic Acid Phenethyl Ester Improves Metabolic Syndrome by Activating PPAR-γ and Inducing Adipose Tissue Remodeling in Diet-Induced Obese Mice. Mol Nutr Food Res 2018; 62:e1700701. [DOI: 10.1002/mnfr.201700701] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 02/09/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Soon-Hee Kim
- Division of Nutrition and Metabolism; Korea Food Research Institute; Seongnam Republic of Korea
| | - Hee-Sook Park
- Division of Nutrition and Metabolism; Korea Food Research Institute; Seongnam Republic of Korea
| | - Moon Ju Hong
- Division of Nutrition and Metabolism; Korea Food Research Institute; Seongnam Republic of Korea
- Department of Food Biotechnology; University of Science and Technology; Seongnam Republic of Korea
| | - Haeng Jeon Hur
- Division of Nutrition and Metabolism; Korea Food Research Institute; Seongnam Republic of Korea
| | - Dae Young Kwon
- Division of Nutrition and Metabolism; Korea Food Research Institute; Seongnam Republic of Korea
| | - Myung-Sunny Kim
- Division of Nutrition and Metabolism; Korea Food Research Institute; Seongnam Republic of Korea
- Department of Food Biotechnology; University of Science and Technology; Seongnam Republic of Korea
| |
Collapse
|
45
|
Deepa P, Sowndhararajan K, Kim S, Park SJ. A role of Ficus species in the management of diabetes mellitus: A review. JOURNAL OF ETHNOPHARMACOLOGY 2018; 215:210-232. [PMID: 29305899 DOI: 10.1016/j.jep.2017.12.045] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/13/2017] [Accepted: 12/30/2017] [Indexed: 05/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Diabetes mellitus is one of the most common global health concerns, with a rapidly increasing incidence. A variety of medicinal plants, particularly those belonging to the genus Ficus (Moraceae), and their active compounds have been used to treat diabetes and related chronic disorders since ancient times. AIM OF THE STUDY The aim of this review is to provide information regarding traditional and scientific knowledge of Ficus species with antidiabetic activity to researchers. MATERIALS AND METHODS A literature search was conducted to obtain information about the antidiabetic properties of Ficus from the electronic databases. Common and scientific names of various Ficus species were used as keywords for the search, along with the terms antidiabetic, hypoglycemic and diabetes. RESULTS Among the assorted species of Ficus that were included in our search, F. benghalensis, F. carica, F. glomerata, F. glumosa, F. racemosa, and F. religiosa exhibited remarkable antidiabetic properties with various mechanisms of action. Moreover, Ficus species are versatile sources of bioactive metabolites such as flavonoids, phenolic acids, tannins, alkaloids, glycosides, coumarins, triterpenoids, sterols and vitamin E. These extracts and isolated compounds significantly have enhanced insulin secretion and subsequently reduced blood glucose level in various in vivo studies. CONCLUSION This review summarizes the antidiabetic potentials of the genus Ficus, including pharmacological studies with mechanisms of action as well as ethnobotanical uses. This review can help inform future scientific research towards the development of novel antidiabetic drugs.
Collapse
Affiliation(s)
- Ponnuvel Deepa
- School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, Gangwon-do, Republic of Korea.
| | - Kandhasamy Sowndhararajan
- School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, Gangwon-do, Republic of Korea.
| | - Songmun Kim
- School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, Gangwon-do, Republic of Korea.
| | - Se Jin Park
- School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, Gangwon-do, Republic of Korea.
| |
Collapse
|
46
|
Han HS, Jeon H, Kang SC. Phellopterin isolated from Angelica dahurica reduces blood glucose level in diabetic mice. Heliyon 2018; 4:e00577. [PMID: 29862342 PMCID: PMC5968131 DOI: 10.1016/j.heliyon.2018.e00577] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 02/05/2018] [Accepted: 03/13/2018] [Indexed: 01/14/2023] Open
Abstract
Insulin resistance is the critical condition for the development of metabolic syndromes including type II diabetes and heart disease. To investigate the active components of Angelica dahurica root which is known to increase insulin sensitivity, its methanol extract was subfractionated. The ethyl acetate (EtOAc) fraction of the Angelica dahurica root extract significantly promoted adipocyte differentiation in 3T3-L1 preadipocyte cells. Among the three compounds isolated from the EtOAc extract (bergapten (1), imperatorin (2) and phellopterin (3)), phellopterin (3) induced the highest adipocyte differentiation at 25 and 50 μg/mL. In addition, treatment with imperatorin (2) and phellopterin (3) increased the mRNA expression of peroxisome proliferator-activated receptors γ (PPARγ). In diabetic animal model induced by high-fat diets (HFD) and streptozotocin (STZ), administration of phellopterin ((3), 1 mg/kg and 2 mg/kg) significantly reduced the levels of blood glucose, triglycerides and total cholesterol. Taken together, these results indicate that phellopterin (3) enhances adipocytes differentiation in 3T3-L1 preadipocytes, phellopterin (3) significantly prevents HFD/STZ-induced type Ⅱ diabetes. The present study also provides phellopterin (3) may be a valuable therapeutic alternative for enhancing insulin sensitivity through promotion of adipocyte differentiation and by increasing mRNA expression levels of PPARγ, which is a major mediator of insulin sensitivity.
Collapse
Affiliation(s)
- Hyo Sang Han
- Department of Health Administration, JoongBu University, Geumsan, 32713, South Korea
| | - Hyelin Jeon
- Department of Oriental Medicine Biotechnology, KyungHee University, Yongin, 17104, South Korea
| | - Se Chan Kang
- Department of Oriental Medicine Biotechnology, KyungHee University, Yongin, 17104, South Korea
- Corresponding author.
| |
Collapse
|
47
|
Chávez-Silva F, Cerón-Romero L, Arias-Durán L, Navarrete-Vázquez G, Almanza-Pérez J, Román-Ramos R, Ramírez-Ávila G, Perea-Arango I, Villalobos-Molina R, Estrada-Soto S. Antidiabetic effect of Achillea millefollium through multitarget interactions: α-glucosidases inhibition, insulin sensitization and insulin secretagogue activities. JOURNAL OF ETHNOPHARMACOLOGY 2018; 212:1-7. [PMID: 29031783 DOI: 10.1016/j.jep.2017.10.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 10/07/2017] [Accepted: 10/07/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL IMPORTANCE Achillea millefolium L. (Asteraceae) is a perennial herb used in Mexican folk medicine for treatment of several pathologies, including inflammatory and spasmodic gastrointestinal disorders, hepatobiliary complaints, overactive cardiovascular, respiratory ailments and diabetes. AIM OF THE STUDY To evaluate the potential antidiabetic effect in vivo and to establish the potential mode of action through in vitro approaches of Achillea millefolium. MATERIALS AND METHODS The antidiabetic effect of hydroalcoholic extract of Achillea millefolium (HAEAm) was evaluated on the oral glucose tolerance tests, in normoglycemic and experimental Type 2 diabetic mice models. In addition, we evaluated the possible mode of action in in vitro assays to determine α-glucosidases inhibition, the insulin secretion and calcium mobilization in RINm5F cells and PPARγ and GLUT4 expression in 3T3-L1 cells. RESULTS HAEAm showed significant glucose diminution on oral glucose tolerance test and in acute experimental Type 2 diabetic assay with respect to the control (p < 0.05). In addition, HAEAm promoted the α-glucosidases inhibition by 55% at 1mg/ml respect to control. On the other hand, HAEAm increased the PPARγ (five-times) and GLUT4 (two-fold) relative expression than control (p < 0.05). Finally, HAEAm significantly increased the insulin secretion and [Ca2+]i compared with control. CONCLUSION The HAEAm possesses in vivo antidiabetic effect, having such effect through multitarget modes of action that involve antihyperglycemic (α-glucosidases inhibition), hypoglycemic (insulin secretion) and potential insulin sensitizer (PPARγ/GLUT4 overexpression) actions.
Collapse
Affiliation(s)
- Fabiola Chávez-Silva
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico
| | - Litzia Cerón-Romero
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico
| | - Luis Arias-Durán
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico
| | | | - Julio Almanza-Pérez
- Laboratorio de Farmacología, Depto. Ciencias de la Salud, D.C.B.S., Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Rubén Román-Ramos
- Laboratorio de Farmacología, Depto. Ciencias de la Salud, D.C.B.S., Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | | | - Irene Perea-Arango
- Centro de Investigación en Biotecnología, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico
| | - Rafael Villalobos-Molina
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México 54090, Mexico
| | - Samuel Estrada-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico.
| |
Collapse
|
48
|
Conway RF, Okarski KM, Szivek JA. A Purification Technique for Adipose-Derived Stromal Cell Cultures Leads to a More Regenerative Cell Population. J INVEST SURG 2018; 32:381-392. [PMID: 29388858 DOI: 10.1080/08941939.2017.1423420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
It has been demonstrated that adipose-derived stromal cells (ASCs) are a regenerative cell population with potential uses for bone and cartilage regeneration. However, the biomarker expression and heterogeneity of the population has not been thoroughly characterized. By analyzing biomarker expression, we aimed to understand the composition of ASC populations extracted using a common extraction technique in comparison to ASC populations given an additional purification step. Human adipose tissue samples were collected, and ASCs were extracted from these samples using a common, published extraction technique (primary extraction). These cells were cultured and half were given an additional purification. The primarily-extracted and purified cell populations were analyzed for biomarkers that correspond to specific cell types. The addition of the purification technique reduced the number of cells expressing hematopoietic and endothelial biomarkers and did not cause the yield of mesenchymal stem cell biomarker-expressing cells to decrease. Biomarkers corresponding to erythrocytes and lymphocytes were lost during the primary extraction, and biomarkers corresponding to most granulocytes and progenitor cells were lost during the additional purification. Biomarkers identifying dendritic cells, monocytes/macrophages, neutrophils, vascular endothelial cells, smooth muscle cells, and pericytes were upregulated in purified cell populations while those identifying fibroblasts and adipocytes were downregulated. Pluripotency biomarkers were more highly expressed in purified cell populations. These results demonstrate that the most commonly utilized adipose tissue recovery and ASC extraction technique leads to a heterogeneous cell population in which further purification of this population, as described in this manuscript, isolates a cell subset that has more regenerative potential.
Collapse
Affiliation(s)
- Renee F Conway
- a Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, College of Medicine, University of Arizona , Tucson , Arizona , USA
| | - Kevin M Okarski
- b Pinnacle Transplant Technologies , Phoenix , Arizona , USA
| | - John A Szivek
- a Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, College of Medicine, University of Arizona , Tucson , Arizona , USA
| |
Collapse
|
49
|
Sadeghabadi ZA, Nourbakhsh M, Alaee M, Larijani B, Razzaghy-Azar M. Peroxisome proliferator-activated receptor gamma expression in peripheral blood mononuclear cells and angiopoietin-like protein 4 levels in obese children and adolescents. J Endocrinol Invest 2018; 41:241-247. [PMID: 28733963 DOI: 10.1007/s40618-017-0730-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/10/2017] [Indexed: 01/20/2023]
Abstract
PURPOSE The peroxisome proliferator-activated receptor γ (PPARγ) is highly expressed in adipose tissue and functions as transcriptional regulator of metabolism and adipocyte differentiation. Angiopoietin-like protein 4 (ANGPTL4), a central player in various aspects of energy homoeostasis, is induced by PPARγ. The aim of this study was to evaluate ANGPTL4 plasma levels and PPARγ gene expression in peripheral blood mononuclear cells (PBMCs) of children and adolescents with obesity and their association with metabolic parameters. METHODS Seventy children and adolescents (35 obese and 35 age- and gender-matched control subjects), were selected. PBMCs were separated and their total RNA was extracted. After cDNA synthesis, PPARG gene expression was analyzed by real-time PCR. Relative differences in gene expression were calculated by ΔCt method using β-actin as a normalizer. Serum ANGPTL4 and insulin were measured using ELISA, and insulin resistance (IR) was calculated by the homeostatic model assessment of insulin resistance (HOMA-IR). Fasting plasma glucose (FPG), triglyceride, total cholesterol, LDL-C and HDL-C were also measured. RESULTS The expression of the PPARG gene as well as the plasma ANGPTL4 levels were significantly diminished in obese subjects as compared to control ones. However, they were not significantly different in obese children with IR compared to obese children without IR or in those with or without metabolic syndrome. A significant positive correlation was found between PPARγ and ANGPTL4 (r = 0.364, p = 0.002). PPARγ expression levels were also significantly correlated with FPG (r = -0.35, p = 0.003). CONCLUSION PPARγ is decreased in childhood obesity and may be responsible for diminished ANGPTL4 levels.
Collapse
Affiliation(s)
- Z A Sadeghabadi
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - M Nourbakhsh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - M Alaee
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - B Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - M Razzaghy-Azar
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- H. Aliasghar Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Ryu S, Kim DS, Lee MW, Lee JW, Sung KW, Koo HH, Yoo KH. Anti-leukemic effects of PPARγ ligands. Cancer Lett 2018; 418:10-19. [PMID: 29331412 DOI: 10.1016/j.canlet.2018.01.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 01/11/2023]
Abstract
The peroxisome proliferator-activated receptor (PPAR) γ, a subtype of PPARs, is a member of the nuclear receptor family. PPARγ and its ligands contribute to various types of diseases including cancer. Given that currently developed therapies against leukemia are not very effective or safe, PPARγ ligands have been shown to be a new class of compounds with the potential to treat hematologic malignancies, particularly leukemia. The capability of PPARγ ligands to induce apoptosis, inhibit proliferation, and promote differentiation of leukemia cells suggests it has significant potential as a drug against leukemia. However, the specific mechanisms and molecules involved are not well-understood, although a number of PPARγ ligands with anti-leukemic effects have been identified. This may explain why PPARγ ligands have not been widely evaluated in clinical trials. To fill the gaps in the lack of understanding of specific anti-leukemic processes of PPARγ ligands and further adapt these molecules as anti-leukemic agents, this review describes previous studies of the anti-leukemic effects of PPARγ ligands.
Collapse
Affiliation(s)
- Somi Ryu
- Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, South Korea.
| | - Dae Seong Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Myoung Woo Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, South Korea.
| |
Collapse
|