1
|
Ruiz de la Bastida A, Langa S, Peirotén Á, Curiel JA, Fernández-González R, Maroto M, Arqués J, Gutiérrez-Adán A, Landete JM. Fermented Lignan-Enriched Soy Beverage Ameliorates the Metabolic Effects of a High-Fat Diet on Female Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5194-5207. [PMID: 39985458 DOI: 10.1021/acs.jafc.4c06947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2025]
Abstract
Fermented vegetable beverages have potential beneficial effects on the health associated with the production of bioactive flavonoids and lignans by selected bacterial strains. Here, we studied the effects of a soy beverage and a soy beverage fermented by Bifidobacterium pseudocatenulatum INIA P815, both supplemented with lignan extracts, in a female mouse model on a high-fat diet followed for 16 weeks. The high-fat diet induced an increase in adipose tissue and plasma cholesterol as well as modified the fecal microbiota. Mice groups receiving any of the beverages showed a reduction in the mean area of ovarian fat tissue adipocytes and exhibited bioactive flavonoids and lignans in plasma and tissues, accompanied by a higher antioxidant activity in plasma. The group of mice subjected to the fermented beverage also demonstrated a lower increase in plasma cholesterol levels, an increase in short-chain fatty acid production, and higher levels of daidzein, genistein, enterolignans, and herbacetin in the plasma and organs. Moreover, the fertility of the mice that received the fermented beverage was also enhanced, resulting in a higher percentage of blastocysts per female mouse. Therefore, the consumption of the beverage fermented by B. pseudocatenulatum INIA P815 could be favoring the health of mice by ameliorating, to some extent, the effects of a high-fat diet.
Collapse
Affiliation(s)
- Ana Ruiz de la Bastida
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid 28040, Spain
| | - Susana Langa
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid 28040, Spain
| | - Ángela Peirotén
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid 28040, Spain
| | - José Antonio Curiel
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid 28040, Spain
| | - Raúl Fernández-González
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid 28040, Spain
| | - María Maroto
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid 28040, Spain
| | - Juan Arqués
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid 28040, Spain
| | - Alfonso Gutiérrez-Adán
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid 28040, Spain
| | - José María Landete
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid 28040, Spain
| |
Collapse
|
2
|
Ion G, Bostan M, Hardman WE, Putt McFarland M, Bleotu C, Radu N, Diaconu CC, Mihaila M, Caramihai MD, Hotnog CM. Nutrients Lowering Obesity-Linked Chemokines Blamable for Metastasis. Int J Mol Sci 2025; 26:2275. [PMID: 40076892 PMCID: PMC11899810 DOI: 10.3390/ijms26052275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Food intake is an essential contributor to both health and disease. Nutrients contribute to a beneficial metabolic equilibrium at the cellular level, preventing or delaying disease onset. Dietary intake contributes to obesity, and obesity supports further cancer and metastasis. Metastasis, a multifactorial and multistep process, is supported by the systemic inflammation of obesity. Spreading of the cancer cells requires the presence of a plethora of recruiter and regulator molecules. Molecules such as chemokines are provided at high levels by obesity-associated fat depots. Chemokine up-regulation in adipose tissue of obese individuals has been associated with different types of cancers such as breast, prostate, colon, liver, and stomach. Chemokines support all metastasis steps from invasion/migration to intravasation, circulation, extravasation, and ending with colonization. The obesity pool of chemokines supporting these processes includes CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CCL11, CCL18, CCL19, CCL20, CXCL1, CXCL5, CXCL 8, CXCL10, and CXCL12. Keeping obesity under control can be beneficial in reducing the levels of pro-inflammatory chemokines and the risk of poor cancer outcome. Nutrients can help, support, and boost cancer treatment effects or jeopardize the treatment. Constituents with anti-inflammatory and anti-obesity properties such as polyphenols, organosulfur components, fatty acids, curcumin, and vitamin E have a proven beneficial effect in lowering obesity and its contribution to metastasis.
Collapse
Affiliation(s)
- Gabriela Ion
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (G.I.); (C.M.H.)
| | - Marinela Bostan
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (G.I.); (C.M.H.)
- Department of Immunology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
| | - Wanda Elaine Hardman
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA (M.P.M.)
| | - Margaret Putt McFarland
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA (M.P.M.)
| | - Coralia Bleotu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.B.); (C.C.D.)
- Research Institute of the University of Bucharest (ICUB), University of Bucharest, 060023 Bucharest, Romania
- The Academy of Romanian Scientist, 050711 Bucharest, Romania
| | - Nicoleta Radu
- Faculty of Biotechnology, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 011464 Bucharest, Romania;
- Biotechnology Department, National Institute for Chemistry and Petrochemistry R&D of Bucharest, 060021 Bucharest, Romania
| | - Carmen Cristina Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.B.); (C.C.D.)
| | - Mirela Mihaila
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (G.I.); (C.M.H.)
- Faculty of Pharmacy, Titu Maiorescu University, 040314 Bucharest, Romania
| | - Mihai Dan Caramihai
- Faculty of Automatic Control and Computer Science, National University of Science and Technology Politehnica Bucharest, 060042 Bucharest, Romania;
| | - Camelia Mia Hotnog
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (G.I.); (C.M.H.)
- Department of Biochemistry and Biophysics, Faculty of Midwives and Nursing, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
| |
Collapse
|
3
|
Xiang J, Mlambo R, Dube P, Machona O, Shaw I, Seid Y, He Y, Luo M, Hong T, He B, Zhou W, Tan S. The obesogenic side of Genistein. Front Endocrinol (Lausanne) 2023; 14:1308341. [PMID: 38098865 PMCID: PMC10720314 DOI: 10.3389/fendo.2023.1308341] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023] Open
Abstract
Genistein (GN) has been highly recommended for its medicinal properties like anticancer, antidiabetic, antihyperlipidemic, antiviral, and antioxidant activities among others. Recently, scientists realized that Genistein is an endocrine disruptor. It is an obesogen that interferes with the endocrine system causing obesity through many mechanisms like inducing adipocyte differentiation, lipid accumulation, and transformation of some stem cells into adipocytes (bone marrow mesenchymal stem cells for example) in vitro. Animal studies show that GN upregulates genes associated with adipogenesis like CCAAT/enhancer binding protein alpha (Cebpα), CCAAT/enhancer binding protein beta (Cebpβ), and PPARγ. In silico studies reveal a strong binding affinity for estrogen receptors. All these findings were contingent on concentration and tissues. It is beyond dispute that obesity is one of the most frustrating medical conditions under the sun. The pathophysiology of this disease was first attributed to a high-calorie diet and lack of physical activity. However, studies proved that these two factors are not enough to account for obesity in both children and adults. This mini review highlights how Genistein interaction with the peroxisome proliferator-activated receptor gamma protein can cause obesity.
Collapse
Affiliation(s)
- Jia Xiang
- Academician Workstation, Changsha Medical University, Changsha, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Ronald Mlambo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Progress Dube
- Simon Mazorodze School of Medical and Health Sciences, Great Zimbabwe University, Masvingo, Zimbabwe
| | - Oleen Machona
- Simon Mazorodze School of Medical and Health Sciences, Great Zimbabwe University, Masvingo, Zimbabwe
| | - Ibrahim Shaw
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Yimer Seid
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Yongju He
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, China
| | - Min Luo
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tingting Hong
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
| | - Binsheng He
- Academician Workstation, Changsha Medical University, Changsha, China
| | - Wenhu Zhou
- Academician Workstation, Changsha Medical University, Changsha, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Songwen Tan
- Academician Workstation, Changsha Medical University, Changsha, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Watanabe S, Haruyama R, Umezawa K, Tomioka I, Nakamura S, Katayama S, Mitani T. Genistein enhances NAD + biosynthesis by upregulating nicotinamide phosphoribosyltransferase in adipocytes. J Nutr Biochem 2023; 121:109433. [PMID: 37648097 DOI: 10.1016/j.jnutbio.2023.109433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
A decrease in the NAD+ level in adipocytes causes adipose-tissue dysfunction, leading to systemic glucose, and lipid metabolism failure. Therefore, it is necessary to develop small molecules and nutraceuticals that can increase NAD+ levels in adipocytes. Genistein, a nutraceutical derived from soybeans, has various physiological activities and improves glucose and lipid metabolism. In this study, we aimed to unravel the effects of genistein on the NAD+ level in adipocytes and the underlying molecular mechanisms. Genistein enhanced NAD+ biosynthesis by increasing the expression of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in NAD+ biosynthesis. A pull-down assay using genistein-immobilized beads revealed prohibitin 1 (PHB1) as a target protein of genistein. The knockdown of Phb1 suppressed the genistein-induced increase in NAMPT expression and NAD+ level in adipocytes. Genistein-bound PHB1 contributed to the stabilization of the transcription factor CCAAT/enhancer-binding protein β through the activation of extracellular signal-regulated kinase, resulting in increased NAMPT expression at the transcriptional level. Genistein induced the dephosphorylation of peroxisome proliferator-activated receptor at serine 273 and increased the level of the insulin-sensitizing adipokine adiponectin in adipocytes, whereas the knockdown of Nampt and Phb1 abolished these genistein-mediated effects. Our results proved the potential efficacy of genistein in increasing the NAD+ level and restoring metabolic function in adipocytes. Furthermore, we identified PHB1, localized to the plasma membrane, as a novel candidate target protein for increased expression of NAMPT in adipocytes. Overall, these findings will assist in developing NAD+-boosting nutraceuticals to alleviate metabolic dysfunctions in adipose tissues.
Collapse
Affiliation(s)
- Shun Watanabe
- Division of Food Science and Biotechnology, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Riki Haruyama
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
| | - Koji Umezawa
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Ikuo Tomioka
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan; Division of Biotechnology, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Soichiro Nakamura
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
| | - Shigeru Katayama
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Takakazu Mitani
- Division of Food Science and Biotechnology, Graduate School of Science and Technology, Shinshu University, Nagano, Japan; Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan.
| |
Collapse
|
5
|
Cheng Y, Tang Y, Tan Y, Li J, Zhang X. KCNK9 mediates the inhibitory effects of genistein on hepatic metastasis from colon cancer. Clinics (Sao Paulo) 2023; 78:100141. [PMID: 36905879 PMCID: PMC10019991 DOI: 10.1016/j.clinsp.2022.100141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 10/08/2022] [Accepted: 11/01/2022] [Indexed: 03/12/2023] Open
Abstract
OBJECTIVE The tyrosine-protein kinase inhibitor, genistein, can inhibit cell malignant transformation and has an antitumor effect on various types of cancer. It has been shown that both genistein and KNCK9 can inhibit colon cancer. This research aimed to investigate the suppressive effects of genistein on colon cancer cells and the association between the application of genistein and KCNK9 expression level. METHODS The Cancer Genome Atlas (TCGA) database was used to study the correlation between the KCNK9 expression level and the prognosis of colon cancer patients. HT29 and SW480 colon cancer cell lines were cultured to examine the inhibitory effects of KCNK9 and genistein on colon cancer in vitro, and a mouse model of colon cancer with liver metastasis was established to verify the inhibitory effect of genistein in vivo. RESULTS KCNK9 was overexpressed in colon cancer cells and was associated with a shorter Overall Survival (OS), a shorter Disease-Specific Survival (DFS), and a shorter Progression-Free Interval (PFI) of colon cancer patients. In vitro experiments showed that downregulation of KCNK9 or genistein application could suppress cell proliferation, migration, and invasion abilities, induce cell cycle quiescence, promote cell apoptosis, and reduce epithelial-mesenchymal transition of the colon cancer cell line. In vivo experiments revealed that silencing of KCNK9 or application of genistein could inhibit hepatic metastasis from colon cancer. Additionally, genistein could inhibit KCNK9 expression, thereby attenuating Wnt/β-catenin signaling pathway. CONCLUSION Genistein inhibited the occurrence and progression of colon cancer through Wnt/β-catenin signaling pathway that could be mediated by KCNK9.
Collapse
Affiliation(s)
- Yuan Cheng
- Department of Pharmacology Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Tang
- Department of Pharmacology Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiming Tan
- Department of Pharmacology Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juan Li
- Department of Pharmacology Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuping Zhang
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, China.
| |
Collapse
|
6
|
Deroyer C, Poulet C, Paulissen G, Ciregia F, Malaise O, Plener Z, Cobraiville G, Daniel C, Gillet P, Malaise MG, de Seny D. CEMIP (KIAA1199) regulates inflammation, hyperplasia and fibrosis in osteoarthritis synovial membrane. Cell Mol Life Sci 2022; 79:260. [PMID: 35474501 PMCID: PMC9042994 DOI: 10.1007/s00018-022-04282-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/24/2022] [Accepted: 04/01/2022] [Indexed: 11/26/2022]
Abstract
Osteoarthritis (OA) synovial membrane is mainly characterized by low-grade inflammation, hyperplasia with increased cell proliferation and fibrosis. We previously underscored a critical role for CEMIP in fibrosis of OA cartilage. However, its role in OA synovial membrane remains unknown. An in vitro model with fibroblast-like synoviocytes from OA patients and an in vivo model with collagenase-induced OA mice were used to evaluate CEMIP-silencing effects on inflammation, hyperplasia and fibrosis. Our results showed that i. CEMIP expression was increased in human and mouse inflamed synovial membrane; ii. CEMIP regulated the inflammatory response pathway and inflammatory cytokines production in vitro and in vivo; iii. CEMIP induced epithelial to mesenchymal transition pathway and fibrotic markers in vitro and in vivo; iv. CEMIP increased cell proliferation and synovial hyperplasia; v. CEMIP expression was increased by inflammatory cytokines and by TGF-β signaling; vi. anti-fibrotic drugs decreased CEMIP expression. All these findings highlighted the central role of CEMIP in OA synovial membrane development and underscored that targeting CEMIP could be a new therapeutic approach.
Collapse
Affiliation(s)
- Céline Deroyer
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium.
| | - Christophe Poulet
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Geneviève Paulissen
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Federica Ciregia
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Olivier Malaise
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Zelda Plener
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Gaël Cobraiville
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | | | - Philippe Gillet
- Department of Orthopaedic Surgery, CHULiège, 4000, Liège, Belgium
| | - Michel G Malaise
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Dominique de Seny
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| |
Collapse
|
7
|
Malaise O, Paulissen G, Deroyer C, Ciregia F, Poulet C, Neuville S, Plener Z, Daniel C, Gillet P, Lechanteur C, Brondello JM, de Seny D, Malaise M. Influence of Glucocorticoids on Cellular Senescence Hallmarks in Osteoarthritic Fibroblast-like Synoviocytes. J Clin Med 2021; 10:jcm10225331. [PMID: 34830613 PMCID: PMC8617749 DOI: 10.3390/jcm10225331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/25/2021] [Accepted: 11/13/2021] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis (OA) is recognized as being a cellular senescence-linked disease. Intra-articular injections of glucocorticoids (GC) are frequently used in knee OA to treat synovial effusion but face controversies about toxicity. We investigated the influence of GC on cellular senescence hallmarks and senescence induction in fibroblast-like synoviocytes (FLS) from OA patients and mesenchymal stem cells (MSC). Methods: Cellular senescence was assessed via the proliferation rate, β-galactosidase staining, DNA damage and CKI expression (p21, p16INK4A). Experimental senescence was induced by irradiation. Results: The GC prednisolone did not induce an apparent senescence phenotype in FLS, with even higher proliferation, no accumulation of β-galactosidase-positive cells nor DNA damage and reduction in p21mRNA, only showing the enhancement of p16INK4A. Prednisolone did not modify experimental senescence induction in FLS, with no modulation of any senescence parameters. Moreover, prednisolone did not induce a senescence phenotype in MSC: despite high β-galactosidase-positive cells, no reduction in proliferation, no DNA damage and no CKI enhancement was observed. Conclusions: We provide reassuring in vitro data about the use of GC regarding cellular senescence involvement in OA: the GC prednisolone did not induce a senescent phenotype in OA FLS (the proliferation ratio was even higher) and in MSC and did not worsen cellular senescence establishment.
Collapse
Affiliation(s)
- Olivier Malaise
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
- Correspondence: ; Tel.: +32-4-366-7863
| | - Geneviève Paulissen
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Céline Deroyer
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Federica Ciregia
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Christophe Poulet
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Sophie Neuville
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Zelda Plener
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Christophe Daniel
- Orthopedic Surgery Department, CHU de Liège, 4000 Liège, Belgium; (C.D.); (P.G.)
| | - Philippe Gillet
- Orthopedic Surgery Department, CHU de Liège, 4000 Liège, Belgium; (C.D.); (P.G.)
| | - Chantal Lechanteur
- Laboratory of Cell and Gene Therapy, Department of Hematology, CHU de Liège, 4000 Liège, Belgium;
| | - Jean-Marc Brondello
- Institute for Regenerative Medicine and Biotherapy, Univ Montpellier, INSERM UMR1183, 34298 Montpellier, France;
| | - Dominique de Seny
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Michel Malaise
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| |
Collapse
|
8
|
Borah AK, Sharma P, Singh A, Kalita KJ, Saha S, Chandra Borah J. Adipose and non-adipose perspectives of plant derived natural compounds for mitigation of obesity. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114410. [PMID: 34273447 DOI: 10.1016/j.jep.2021.114410] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/07/2021] [Accepted: 07/10/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Phyto-preparations and phyto-compounds, by their natural origin, easy availability, cost-effectiveness, and fruitful traditional uses based on accumulated experiences, have been extensively explored to mitigate the global burden of obesity. AIM OF THIS REVIEW The review aimed to analyse and critically summarize the prospect of future anti-obesity drug leads from the extant array of phytochemicals for mitigation of obesity, using adipose related targets (adipocyte formation, lipid metabolism, and thermogenesis) and non-adipose targets (hepatic lipid metabolism, appetite, satiety, and pancreatic lipase activity). Phytochemicals as inhibitors of adipocyte differentiation, modulators of lipid metabolism, and thermogenic activators of adipocytes are specifically discussed with their non-adipose anti-obesogenic targets. MATERIALS AND METHODS PubMed, Google Scholar, Scopus, and SciFinder were accessed to collect data on traditional medicinal plants, compounds derived from plants, their reported anti-obesity mechanisms, and therapeutic targets. The taxonomically accepted name of each plant in this review has been vetted from "The Plant List" (www.theplantlist.org) or MPNS (http://mpns.kew.org). RESULTS Available knowledge of a large number of phytochemicals, across a range of adipose and non-adipose targets, has been critically analysed and delineated by graphical and tabular depictions, towards mitigation of obesity. Neuro-endocrinal modulation in non-adipose targets brought into sharp dual focus, both non-adipose and adipose targets as the future of anti-obesity research. Numerous phytochemicals (Berberine, Xanthohumol, Ursolic acid, Guggulsterone, Tannic acid, etc.) have been found to be effectively reducing weight through lowered adipocyte formation, increased lipolysis, decreased lipogenesis, and enhanced thermogenesis. They have been affirmed as potential anti-obesity drugs of future because of their effectiveness yet having no threat to adipose or systemic insulin sensitivity. CONCLUSION Due to high molecular diversity and a greater ratio of benefit to risk, plant derived compounds hold high therapeutic potential to tackle obesity and associated risks. This review has been able to generate fresh perspectives on the anti-diabetic/anti-hyperglycemic/anti-obesity effect of phytochemicals. It has also brought into the focus that many phytochemicals demonstrating in vitro anti-obesogenic effects are yet to undergo in vivo investigation which could lead to potential phyto-molecules for dedicated anti-obesity action.
Collapse
Affiliation(s)
- Anuj Kumar Borah
- Dept. of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, 784028, Assam, India
| | - Pranamika Sharma
- Laboratory of Chemical Biology, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India
| | - Archana Singh
- Dept. of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, 784028, Assam, India
| | - Kangkan Jyoti Kalita
- Laboratory of Chemical Biology, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India
| | - Sougata Saha
- Dept. of Biotechnology, NIT Durgapur, West Bengal, 713209, India
| | - Jagat Chandra Borah
- Laboratory of Chemical Biology, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India.
| |
Collapse
|
9
|
Bernatoniene J, Kazlauskaite JA, Kopustinskiene DM. Pleiotropic Effects of Isoflavones in Inflammation and Chronic Degenerative Diseases. Int J Mol Sci 2021; 22:ijms22115656. [PMID: 34073381 PMCID: PMC8197878 DOI: 10.3390/ijms22115656] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Isoflavones are phytoestrogens of plant origin, mostly found in the members of the Fabaceae family, that exert beneficial effects in various degenerative disorders. Having high similarity to 17-β-estradiol, isoflavones can bind estrogen receptors, scavenge reactive oxygen species, activate various cellular signal transduction pathways and modulate growth and transcription factors, activities of enzymes, cytokines, and genes regulating cell proliferation and apoptosis. Due to their pleiotropic activities isoflavones might be considered as a natural alternative for the treatment of estrogen decrease-related conditions during menopause. This review will focus on the effects of isoflavones on inflammation and chronic degenerative diseases including cancer, metabolic, cardiovascular, neurodegenerative diseases, rheumatoid arthritis and adverse postmenopausal symptoms.
Collapse
Affiliation(s)
- Jurga Bernatoniene
- Department of Drug Technology and Social Pharmacy, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania; (J.A.K.); (D.M.K.)
- Correspondence:
| | - Jurga Andreja Kazlauskaite
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania; (J.A.K.); (D.M.K.)
| | - Dalia Marija Kopustinskiene
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania; (J.A.K.); (D.M.K.)
| |
Collapse
|
10
|
Ahmed QU, Ali AHM, Mukhtar S, Alsharif MA, Parveen H, Sabere ASM, Nawi MSM, Khatib A, Siddiqui MJ, Umar A, Alhassan AM. Medicinal Potential of Isoflavonoids: Polyphenols That May Cure Diabetes. Molecules 2020; 25:molecules25235491. [PMID: 33255206 PMCID: PMC7727648 DOI: 10.3390/molecules25235491] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 01/18/2023] Open
Abstract
In recent years, there is emerging evidence that isoflavonoids, either dietary or obtained from traditional medicinal plants, could play an important role as a supplementary drug in the management of type 2 diabetes mellitus (T2DM) due to their reported pronounced biological effects in relation to multiple metabolic factors associated with diabetes. Hence, in this regard, we have comprehensively reviewed the potential biological effects of isoflavonoids, particularly biochanin A, genistein, daidzein, glycitein, and formononetin on metabolic disorders and long-term complications induced by T2DM in order to understand whether they can be future candidates as a safe antidiabetic agent. Based on in-depth in vitro and in vivo studies evaluations, isoflavonoids have been found to activate gene expression through the stimulation of peroxisome proliferator-activated receptors (PPARs) (α, γ), modulate carbohydrate metabolism, regulate hyperglycemia, induce dyslipidemia, lessen insulin resistance, and modify adipocyte differentiation and tissue metabolism. Moreover, these natural compounds have also been found to attenuate oxidative stress through the oxidative signaling process and inflammatory mechanism. Hence, isoflavonoids have been envisioned to be able to prevent and slow down the progression of long-term diabetes complications including cardiovascular disease, nephropathy, neuropathy, and retinopathy. Further thoroughgoing investigations in human clinical studies are strongly recommended to obtain the optimum and specific dose and regimen required for supplementation with isoflavonoids and derivatives in diabetic patients.
Collapse
Affiliation(s)
- Qamar Uddin Ahmed
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang DM, Malaysia; (A.H.M.A); (A.S.M.S.); (M.S.M.N.); (A.K.); (M.J.S.)
- Pharmacognosy Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang DM, Malaysia
- Correspondence: (Q.U.A.); (S.M.)
| | - Abdul Hasib Mohd Ali
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang DM, Malaysia; (A.H.M.A); (A.S.M.S.); (M.S.M.N.); (A.K.); (M.J.S.)
| | - Sayeed Mukhtar
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia; (M.A.A.); (H.P.)
- Correspondence: (Q.U.A.); (S.M.)
| | - Meshari A. Alsharif
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia; (M.A.A.); (H.P.)
| | - Humaira Parveen
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia; (M.A.A.); (H.P.)
| | - Awis Sukarni Mohmad Sabere
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang DM, Malaysia; (A.H.M.A); (A.S.M.S.); (M.S.M.N.); (A.K.); (M.J.S.)
- Pharmacognosy Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang DM, Malaysia
| | - Mohamed Sufian Mohd. Nawi
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang DM, Malaysia; (A.H.M.A); (A.S.M.S.); (M.S.M.N.); (A.K.); (M.J.S.)
- Pharmacognosy Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang DM, Malaysia
| | - Alfi Khatib
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang DM, Malaysia; (A.H.M.A); (A.S.M.S.); (M.S.M.N.); (A.K.); (M.J.S.)
- Pharmacognosy Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang DM, Malaysia
| | - Mohammad Jamshed Siddiqui
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang DM, Malaysia; (A.H.M.A); (A.S.M.S.); (M.S.M.N.); (A.K.); (M.J.S.)
- Pharmacognosy Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang DM, Malaysia
| | - Abdulrashid Umar
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, P M B: 2436 Sokoto, Nigeria; (A.U.); (A.M.A.)
| | - Alhassan Muhammad Alhassan
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, P M B: 2436 Sokoto, Nigeria; (A.U.); (A.M.A.)
| |
Collapse
|
11
|
Kuryłowicz A, Cąkała-Jakimowicz M, Puzianowska-Kuźnicka M. Targeting Abdominal Obesity and Its Complications with Dietary Phytoestrogens. Nutrients 2020; 12:nu12020582. [PMID: 32102233 PMCID: PMC7071386 DOI: 10.3390/nu12020582] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/21/2022] Open
Abstract
In the assessment of the health risk of an obese individual, both the amount of adipose tissue and its distribution and metabolic activity are essential. In adults, the distribution of adipose tissue differs in a gender-dependent manner and is regulated by sex steroids, especially estrogens. Estrogens affect adipocyte differentiation but are also involved in the regulation of the lipid metabolism, insulin resistance, and inflammatory activity of the adipose tissue. Their deficiency results in unfavorable changes in body composition and increases the risk of metabolic complications, which can be partially reversed by hormone replacement therapy. Therefore, the idea of the supplementation of estrogen-like compounds to counteract obesity and related complications is compelling. Phytoestrogens are natural plant-derived dietary compounds that resemble human estrogens in their chemical structure and biological activity. Supplementation with phytoestrogens may confer a range of beneficial effects. However, results of studies on the influence of phytoestrogens on body composition and prevalence of obesity are inconsistent. In this review, we present data from in vitro, animal, and human studies regarding the role of phytoestrogens in adipose tissue development and function in the context of their potential application in the prevention of visceral obesity and related complications.
Collapse
Affiliation(s)
- Alina Kuryłowicz
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland; (M.C.-J.); (M.P.-K.)
- Correspondence: ; Tel.: +48226086591; Fax: +48226086410
| | - Marta Cąkała-Jakimowicz
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland; (M.C.-J.); (M.P.-K.)
| | - Monika Puzianowska-Kuźnicka
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland; (M.C.-J.); (M.P.-K.)
- Department of Geriatrics and Gerontology, Medical Centre of Postgraduate Education, 61/63 Kleczewska Street, 01-826, Warsaw, Poland
| |
Collapse
|
12
|
Osteoarthritis Is a Low-Grade Inflammatory Disease: Obesity's Involvement and Herbal Treatment. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:2037484. [PMID: 31781260 PMCID: PMC6874989 DOI: 10.1155/2019/2037484] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/30/2019] [Accepted: 10/03/2019] [Indexed: 12/26/2022]
Abstract
Osteoarthritis (OA) is considered a major cause of disability around the globe. This handicapping disease causes important cartilage and bone alteration that is associated with serious pains and loss of joint function. Despite its frequent association with obesity, the aetiology of OA is not fully understood. In this review, the different aspects of OA and its correlation with obesity were analysed. Through examining different mechanisms by which obesity may trigger and/or exacerbate OA, we point out some relevant signalling pathways that may evolve as candidates for pharmacological drug development. As such, we also suggest a review of different herbal medicines (HMs) and their main compounds, which specifically interfere with the identified pathways. We have shown that obesity's involvement in OA is not only limited to the mechanical weight exerted on the joints (mechanical hypothesis), but also induces an inflammatory state by different mechanisms, including increased leptin expression, compromised gut mucosa, and/or gut microbiota disruption. The main signalling pathways involved in OA inflammation, which are associated with obesity, are protein tyrosine phosphatase 1B (PTP1B) and TLR4 or DAP12. Moreover, we also underline the contamination of plant extracts with LPS as an important factor to consider when studying HM's effects on articular cells. By summarizing recent publications, this review aims at highlighting newly established aspects of obesity involvement in OA other than the mechanical one.
Collapse
|
13
|
15-Deoxy-Δ-12, 14-prostaglandin J2 acts cooperatively with prednisolone to reduce TGF-β-induced pro-fibrotic pathways in human osteoarthritis fibroblasts. Biochem Pharmacol 2019; 165:66-78. [DOI: 10.1016/j.bcp.2019.03.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 03/28/2019] [Indexed: 12/20/2022]
|
14
|
Deroyer C, Charlier E, Neuville S, Malaise O, Gillet P, Kurth W, Chariot A, Malaise M, de Seny D. CEMIP (KIAA1199) induces a fibrosis-like process in osteoarthritic chondrocytes. Cell Death Dis 2019; 10:103. [PMID: 30718510 PMCID: PMC6362103 DOI: 10.1038/s41419-019-1377-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/16/2018] [Accepted: 01/18/2019] [Indexed: 12/21/2022]
Abstract
CEMIP (for "Cell migration-inducing protein" also called KIAA1199 and Hybid for "Hyaluronan-binding protein") expression is increased in cancers and described as a regulator of cell survival, growth and invasion. In rheumatoid arthritis, CEMIP is referred to as an angiogenic marker and participates in hyaluronic acid degradation. In this study, CEMIP expression is investigated in healthy and osteoarthritis (OA) cartilage from human and mouse. Its role in OA physiopathology is deciphered, specifically in chondrocytes proliferation and dedifferentiation and in the extracellular matrix remodeling. To this end, CEMIP, αSMA and types I and III collagen expressions were assessed in human OA and non-OA cartilage. CEMIP expression was also investigated in a mouse OA model. CEMIP expression was studied in vitro using a chondrocyte dedifferentiation model. High-throughput RNA sequencing was performed on chondrocytes after CEMIP silencing. Results showed that CEMIP was overexpressed in human and murine OA cartilage and along chondrocytes dedifferentiation. Most of genes deregulated in CEMIP-depleted cells were involved in cartilage turnover (e.g., collagens), mesenchymal transition and fibrosis. CEMIP regulated β-catenin protein level. Moreover, CEMIP was essential for chondrocytes proliferation and promoted αSMA expression, a fibrosis marker, and TGFβ signaling towards the p-Smad2/3 (Alk5/PAI-1) pathway. Interestingly, CEMIP was induced by the pSmad1/5 (Alk1) pathway. αSMA and type III collagen expressions were overexpressed in human OA cartilage and along chondrocytes dedifferentiation. Finally, CEMIP was co-expressed in situ with αSMA in all OA cartilage layers. In conclusion, CEMIP was sharply overexpressed in human and mouse OA cartilage and along chondrocytes dedifferentiation. CEMIP-regulated transdifferentiation of chondrocytes into "chondro-myo-fibroblasts" expressing α-SMA and type III collagen, two fibrosis markers. Moreover, these "chondro-myo-fibroblasts" were found in OA cartilage but not in healthy cartilage.
Collapse
Affiliation(s)
- Céline Deroyer
- Laboratory of Rheumatology, GIGA I3, CHU de Liege, University of Liege, Liege, Belgium.
| | - Edith Charlier
- Laboratory of Rheumatology, GIGA I3, CHU de Liege, University of Liege, Liege, Belgium
| | - Sophie Neuville
- Laboratory of Rheumatology, GIGA I3, CHU de Liege, University of Liege, Liege, Belgium
| | - Olivier Malaise
- Laboratory of Rheumatology, GIGA I3, CHU de Liege, University of Liege, Liege, Belgium
| | | | | | - Alain Chariot
- Laboratory of Medical Chemistry, GIGA Molecular Biology of Diseases, University of Liege, Liege, Belgium.,Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Liege, Belgium
| | - Michel Malaise
- Laboratory of Rheumatology, GIGA I3, CHU de Liege, University of Liege, Liege, Belgium
| | - Dominique de Seny
- Laboratory of Rheumatology, GIGA I3, CHU de Liege, University of Liege, Liege, Belgium
| |
Collapse
|
15
|
Davis JE, Hastings D. Transcriptional Regulation of TCF/LEF and PPARγ by Daidzein and Genistein in 3T3-L1 Preadipocytes. J Med Food 2018; 21:761-768. [DOI: 10.1089/jmf.2017.0136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Jeremy E. Davis
- Department of Animal Science, Food and Nutrition, Southern Illinois University, Carbondale, Illinois, USA
| | - Darcie Hastings
- Department of Animal Science, Food and Nutrition, Southern Illinois University, Carbondale, Illinois, USA
| |
Collapse
|
16
|
BAY 11-7085 induces glucocorticoid receptor activation and autophagy that collaborate with apoptosis to induce human synovial fibroblast cell death. Oncotarget 2018; 7:23370-82. [PMID: 26993765 PMCID: PMC5029633 DOI: 10.18632/oncotarget.8042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 02/28/2016] [Indexed: 11/29/2022] Open
Abstract
Inhibition of proapoptotic pathways in synovial fibroblasts is one of the major causes of synovial proliferation and hyperplasia in rheumatic diseases. We have shown previously that NF-κB inhibitor BAY 11-7085, through inactivation of PPAR-γ, induces apoptosis in human synovial fibroblasts. In this work we showed that BAY 11-7085 induced autophagy that preceded BAY 11-7085-induced apoptosis. Of interest, BAY 11-7085 induced Serine 211 phosphorylation and degradation of glucocorticoid receptor (GR). Glucocorticoid prednisolone induced both activation and degradation of GR, as well as autophagy in synovial fibroblasts. BAY 11-7085-induced cell death was significantly decreased with glucocorticoid inhibitor mifepristone and with inhibitors of autophagy. Both BAY 11-7085-induced autophagy and GR activation were down regulated with PPAR-γ agonist, 15d-PGJ2 and MEK/ERK inhibitor UO126. Inhibition of autophagy markedly decreased endogenous and BAY 11-7085-induced ERK phosphorylation, suggesting a positive feed back loop between ERK activation and autophagy in synovial fibroblasts. Co-transfection of MEK1 with PPAR-γ1 in HEK293 cells caused known inhibitory phosphorylation of PPAR-γ1 (Serine 112) and enhanced GR degradation, in the absence or presence of prednisolone. Furthermore, GR was both phosphorylated on Serine 211 and down regulated in synovial fibroblasts during serum starvation induced autophagy. These results showed that GR activation and PPAR-γ inactivation mediated BAY 11-7085-induced autophagy.
Collapse
|
17
|
Aryaeian N, Sedehi SK, Arablou T. Polyphenols and their effects on diabetes management: A review. Med J Islam Repub Iran 2017; 31:134. [PMID: 29951434 PMCID: PMC6014790 DOI: 10.14196/mjiri.31.134] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Indexed: 12/15/2022] Open
Abstract
Background: Type 2 diabetes is a growing public health problem and is associated with increased morbidity and mortality. The worldwide prevalence of type 2 diabetes is rising. Polyphenols, such as flavonoids, phenolic acid, and stilbens, are a large and heterogeneous group of phytochemicals in plant-based foods. In this review, we aimed at assessing the studies on polyphenols and diabetes management. Methods: A literature search in the PubMed, EMBASE, Scopus, and ISI Web of Science databases was conducted to identify relevant studies published from 1986 to Jan 2017. Results: Several animal models and a limited number of human studies have revealed that polyphenols decrease hyperglycemia and improve acute insulin secretion and insulin sensitivity. The possible mechanisms include decrease in glucose absorption in the intestine, inhibition of carbohydrates digestion, stimulation of insulin secretion, modulation of glucose release from the liver, activation of insulin receptors and glucose uptake in insulin-sensitive tissues, modulation of intracellular signaling pathways, and gene expression. Conclusion: Growing evidence indicates that various dietary polyphenols may influence blood glucose at different levels and may also help control and prevent diabetes complication. However, we still need more clinical trials to determine the effects of polyphenols- rich foods, their effective dose, and mechanisms of their effects in managing diabetes.
Collapse
Affiliation(s)
- Naheed Aryaeian
- Research Center for Environmental Health Technology, Iran University of Medical Sciences and Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Khorshidi Sedehi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Tahereh Arablou
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Wang S, Wang Y, Pan MH, Ho CT. Anti-obesity molecular mechanism of soy isoflavones: weaving the way to new therapeutic routes. Food Funct 2017; 8:3831-3846. [PMID: 29043346 DOI: 10.1039/c7fo01094j] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obesity is ringing alarm bells globally. Advances in food science and nutrition research have been devoted to identifying food components that exert anti-obesity effects, as well as investigating the molecular mechanisms by which they modulate the progression of obesity. Soy foods have attracted much interest as high-protein components of the human diet and as unique sources of isoflavones. As they have similar chemical structures to endogenous estrogens, isoflavones are believed to interact with intracellular estrogen receptors, which results in reductions in the accumulation of lipids and the distribution of adipose tissue. Both in vitro and in vivo studies have revealed other signaling pathways in which isoflavones are involved in the inhibition of adipogenesis and lipogenesis by interacting with various transcription factors and upstream signaling molecules. Although the biological mechanisms that cause the biphasic effects of isoflavones and various controversial results remain unknown, it is noteworthy that isoflavones exhibit pleiotropic effects in the human body to regulate metabolism and balance, which may potentially prevent and treat obesity.
Collapse
Affiliation(s)
- Siyu Wang
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA.
| | | | | | | |
Collapse
|
19
|
Genistein: Its role in metabolic diseases and cancer. Crit Rev Oncol Hematol 2017; 119:13-22. [PMID: 29065980 DOI: 10.1016/j.critrevonc.2017.09.004] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/23/2017] [Accepted: 09/11/2017] [Indexed: 12/12/2022] Open
Abstract
Genistein is an isoflavone present in soy and is known to have multiple molecular effects, such as the inhibition of inflammation, promotion of apoptosis, and modulation of steroidal hormone receptors and metabolic pathways. Since these molecular effects impact carcinogenesis, cancer propagation, obesity, osteoporosis, and metabolic syndromes, genistein plays an important role in preventing and treating common disorders. The role of genistein has not been adequately evaluated in all these clinical settings. This review summarizes some of the known molecular effects of genistein and its potential role in health maintenance and treatment.
Collapse
|
20
|
Insights on Molecular Mechanisms of Chondrocytes Death in Osteoarthritis. Int J Mol Sci 2016; 17:ijms17122146. [PMID: 27999417 PMCID: PMC5187946 DOI: 10.3390/ijms17122146] [Citation(s) in RCA: 253] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/05/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a joint pathology characterized by progressive cartilage degradation. Medical care is mainly based on alleviating pain symptoms. Compelling studies report the presence of empty lacunae and hypocellularity in cartilage with aging and OA progression, suggesting that chondrocyte cell death occurs and participates to OA development. However, the relative contribution of apoptosis per se in OA pathogenesis appears complex to evaluate. Indeed, depending on technical approaches, OA stages, cartilage layers, animal models, as well as in vivo or in vitro experiments, the percentage of apoptosis and cell death types can vary. Apoptosis, chondroptosis, necrosis, and autophagic cell death are described in this review. The question of cell death causality in OA progression is also addressed, as well as the molecular pathways leading to cell death in response to the following inducers: Fas, Interleukin-1β (IL-1β), Tumor Necrosis factor-α (TNF-α), leptin, nitric oxide (NO) donors, and mechanical stresses. Furthermore, the protective role of autophagy in chondrocytes is highlighted, as well as its decline during OA progression, enhancing chondrocyte cell death; the transition being mainly controlled by HIF-1α/HIF-2α imbalance. Finally, we have considered whether interfering in chondrocyte apoptosis or promoting autophagy could constitute therapeutic strategies to impede OA progression.
Collapse
|
21
|
Glucocorticoid-induced leucine zipper (GILZ) is involved in glucocorticoid-induced and mineralocorticoid-induced leptin production by osteoarthritis synovial fibroblasts. Arthritis Res Ther 2016; 18:219. [PMID: 27716396 PMCID: PMC5050640 DOI: 10.1186/s13075-016-1119-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 09/13/2016] [Indexed: 01/09/2023] Open
Abstract
Background Glucocorticoid-induced leucine zipper (GILZ) is a mediator of the anti-inflammatory activities of glucocorticoids. However, GILZ deletion does not impair the anti-inflammatory activities of exogenous glucocorticoids in mice arthritis models and GILZ could also mediate some glucocorticoid-related adverse events. Osteoarthritis (OA) is a metabolic disorder that is partly attributed to adipokines such as leptin, and we previously observed that glucocorticoids induced leptin secretion in OA synovial fibroblasts. The purpose of this study was to position GILZ in OA through its involvement in the anti-inflammatory activities of glucocorticoids and/or in the metabolic pathway of leptin induction. The influences of mineralocorticoids on GILZ and leptin expression were also investigated. Methods Human synovial fibroblasts were isolated from OA patients during knee replacement surgery. Then, the cells were treated with a glucocorticoid (prednisolone), a mineralocorticoid (aldosterone), a glucocorticoid receptor (GR) antagonist (mifepristone), a selective glucocorticoid receptor agonist (Compound A), mineralocorticoid receptor (MR) antagonists (eplerenone and spironolactone), TNF-α or transforming growth factor (TGF)-β. Cells were transfected with shRNA lentiviruses for the silencing of GILZ and GR. The leptin, IL-6, IL-8 and matrix metalloproteinase (MMP)-1 levels were measured by ELISA. Leptin, the leptin receptor (Ob-R), GR and GILZ expression levels were analyzed by western blotting and/or RT-qPCR. Results (1) The glucocorticoid prednisolone and the mineralocorticoid aldosterone induced GILZ expression dose-dependently in OA synovial fibroblasts, through GR but not MR. Similar effects on leptin and Ob-R were observed: leptin secretion and Ob-R expression were also induced by prednisolone and aldosterone through GR; (2) GILZ silencing experiments demonstrated that GILZ was involved in the glucocorticoid-induced and mineralocorticoid-induced leptin secretion and Ob-R expression in OA synovial fibroblasts; and (3) GILZ inhibition did not alter the production of pro-inflammatory cytokines by OA synovial fibroblast or the anti-inflammatory properties of glucocorticoids. Conclusions The absence of GILZ prevents corticoid-induced leptin and Ob-R expression without affecting the anti-inflammatory properties of glucocorticoids in OA synovial fibroblasts. Mineralocorticoids also induce leptin and Ob-R expression through GILZ.
Collapse
|
22
|
Zhang LY, Xue HG, Chen JY, Chai W, Ni M. Genistein induces adipogenic differentiation in human bone marrow mesenchymal stem cells and suppresses their osteogenic potential by upregulating PPARγ. Exp Ther Med 2016; 11:1853-1858. [PMID: 27168816 PMCID: PMC4840518 DOI: 10.3892/etm.2016.3120] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 01/11/2016] [Indexed: 01/10/2023] Open
Abstract
Genistein is a soy isoflavone that exists in the form of an aglycone. It is the primary active component in soy isoflavone and has a number of biological activities (anti-inflammatory and anti-oxidative). However, the specific effect of genistein on human bone marrow mesenchymal stem cells (BMSCs) remains unclear. In the present study, the mechanism underlying the effect of genistein on the suppression of BMSC adipogenic differentiation and the enhancement of osteogenic potential was investigated using an MTT assay. It was observed that genistein significantly increased BMSC cell proliferation in a time- and dose-dependent manner (P<0.01). In addition, reverse transcription-quantitative polymerase chain reaction revealed that genistein significantly inhibited the expression of runt-related transcription factor 2 (Runx2), type I collagen (Col I) and osteocalcin (OC; P<0.01). Furthermore, 20 µm genistein significantly inhibited the activity of alkaline phosphatase (ALP) and increased the activity of triglycerides (TGs) increased (P<0.01) as determined by an enzyme-linked immunosorbent assay. Finally, western blotting revealed that BMSC pretreatment with 20 µm genistein significantly increased peroxisome proliferator-activated receptor γ (PPARγ) protein expression (P<0.01). This suggests that the downregulation of PPARγ may significantly reduce the effect of genistein on cell proliferation, suppress the expression of Runx2, Col I and OC mRNA, and reduce ALP and promote TG activity in BMSCs. Thus, the results of the present study conclude that genistein induces adipogenic differentiation in human BMSCs and suppresses their osteogenic potential by upregulating the expression of PPARγ. In conclusion, genistein may be a promising candidate drug for treatment against osteogenesis.
Collapse
Affiliation(s)
- Li-Yan Zhang
- Department of Orthopedics, General Hospital of Chinese People's Liberation Army, Beijing 100853, P.R. China; First Department of Orthopedics, The Affiliated Hospital of Beihua University, Jilin, Jilin 132001, P.R. China
| | - Hao-Gang Xue
- First Department of Orthopedics, The Affiliated Hospital of Beihua University, Jilin, Jilin 132001, P.R. China
| | - Ji-Ying Chen
- Department of Orthopedics, General Hospital of Chinese People's Liberation Army, Beijing 100853, P.R. China
| | - Wei Chai
- Department of Orthopedics, General Hospital of Chinese People's Liberation Army, Beijing 100853, P.R. China
| | - Ming Ni
- Department of Orthopedics, General Hospital of Chinese People's Liberation Army, Beijing 100853, P.R. China
| |
Collapse
|
23
|
Younger J, Kapphahn K, Brennan K, Sullivan SD, Stefanick ML. Association of Leptin with Body Pain in Women. J Womens Health (Larchmt) 2016; 25:752-60. [PMID: 27028709 PMCID: PMC4939369 DOI: 10.1089/jwh.2015.5509] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Leptin, an appetite-regulatory hormone, is also known to act as a proinflammatory adipokine. One of the effects of increased systemic leptin concentrations may be greater sensitivity to pain. We report the results of two studies examining the association between leptin and pain: a small pilot longitudinal study, followed by a large cross-sectional study. In Study 1, three women with physician-diagnosed fibromyalgia provided blood draws daily for 25 consecutive days, as well as daily self-reported musculoskeletal pain. Daily fluctuations in serum leptin were positively associated with pain across all three participants (F (1,63) = 12.8, p < 0.001), with leptin predicting ∼49% of the pain variance. In Study 2, the relationship between leptin and body pain was examined in a retrospective cross-sectional analysis of 5676 generally healthy postmenopausal women from the Women's Health Initiative. Leptin levels obtained from single blood draws were tested for a relationship with self-reported body pain. Body mass index (BMI) was also included as a predictor of pain. Both leptin and BMI were found to be independently associated with self-reported pain (p = 0.001 and p < 0.001, respectively), with higher leptin levels and greater BMI each being associated with greater pain. Leptin appears to be a predictor of body pain both within- and between-individuals and may be a driver of generalized pain states such as fibromyalgia.
Collapse
Affiliation(s)
- Jarred Younger
- 1 University of Alabama at Birmingham , Birmingham, Alabama
| | | | | | | | - Marcia L Stefanick
- 5 Department of Medicine, Stanford Prevention Research Center, Stanford University , Stanford, California
| |
Collapse
|
24
|
Charlier E, Malaise O, Zeddou M, Neuville S, Cobraiville G, Deroyer C, Sanchez C, Gillet P, Kurth W, de Seny D, Relic B, Malaise MG. Restriction of spontaneous and prednisolone-induced leptin production to dedifferentiated state in human hip OA chondrocytes: role of Smad1 and β-catenin activation. Osteoarthritis Cartilage 2016; 24:315-24. [PMID: 26318657 DOI: 10.1016/j.joca.2015.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aetiology of OA is not fully understood although several adipokines such as leptin are known mediators of disease progression. Since leptin levels were increased in synovial fluid compared to serum in OA patients, it was suggested that joint cells themselves could produce leptin. However, exact mechanisms underlying leptin production by chondrocytes are poorly understood. Nevertheless, prednisolone, although displaying powerful anti-inflammatory properties has been recently reported to be potent stimulator of leptin and its receptor in OA synovial fibroblasts. Therefore, we investigated, in vitro, spontaneous and prednisolone-induced leptin production in OA chondrocytes, focusing on transforming growth factor-β (TGFβ) and Wnt/β-catenin pathways. DESIGN We used an in vitro dedifferentiation model, comparing human freshly isolated hip OA chondrocytes cultivated in monolayer during 1 day (type II, COL2A1 +; type X, COL10A1 + and type I collagen, COL1A1 -) or 14 days (COL2A1 -; COL10A1 - and COL1A1+). RESULTS Leptin expression was not detected in day1 OA chondrocytes whereas day14 OA chondrocytes produced leptin, significantly increased with prednisolone. Activin receptor-like kinase 1 (ALK1)/ALK5 ratio was shifted during dedifferentiation, from high ALK5 and phospho (p)-Smad2 expression at day1 to high ALK1, endoglin and p-Smad1/5 expression at day14. Moreover, inactive glycogen synthase kinase 3 (GSK3) and active β-catenin were only found in dedifferentiated OA chondrocytes. Smad1 and β-catenin but not endoglin stable lentiviral silencing led to a significant decrease in leptin production by dedifferentiated OA chondrocytes. CONCLUSIONS Only dedifferentiated OA chondrocytes produced leptin. Prednisolone markedly enhanced leptin production, which involved Smad1 and β-catenin activation.
Collapse
Affiliation(s)
- E Charlier
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium.
| | - O Malaise
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - M Zeddou
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - S Neuville
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - G Cobraiville
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - C Deroyer
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - C Sanchez
- Bone and Cartilage Research Unit, Arthropole Liège, University of Liège, Belgium
| | - P Gillet
- Orthopedic Surgery Unit, CHU of Liège, Belgium
| | - W Kurth
- Orthopedic Surgery Unit, CHU of Liège, Belgium
| | - D de Seny
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - B Relic
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - M G Malaise
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| |
Collapse
|
25
|
Shin HW, Jang ES, Moon BS, Lee JJ, Lee DE, Lee CH, Shin CS. Anti-obesity effects of gochujang products prepared using rice koji and soybean meju in rats. Journal of Food Science and Technology 2016; 53:1004-13. [PMID: 27162380 DOI: 10.1007/s13197-015-2162-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 12/09/2015] [Accepted: 12/22/2015] [Indexed: 01/18/2023]
Abstract
The Korean traditional hot sauce gochujang has been reported to have biological activities. Different kinds of gochujang products were prepared based on combinations of a fungal rice koji with two kinds of bacterial soybean mejus. Diets that included gochujang products were fed to rats and anti-obesity effects were investigated. Gochujang products reduced body weight gains, epididymal fat weights, and triglyceride levels in the serum and the liver. Effects were exerted by the diet that included the non-fermented gochujang mixture, increased using a fungal rice koji, and further enhanced using a bacterial soybean meju. Dietary effects were apparently induced via inhibition of the lipogenic enzymes fatty acid synthase, malic enzyme, and lipoprotein lipase by gochujang products in epididymal adipose tissues, and inhibition of glucose-6-phosphate dehydrogenase in the liver. High levels of capsaicin and genistein in gochujang products are considered to contribute to anti-obesity effects.
Collapse
Affiliation(s)
- H W Shin
- CJ CheilJedang Corp., Food Research Institute, Suwon-si, 443-270 South Korea ; Department of Biotechnology, Yonsei University, Seoul, 120-749 South Korea
| | - E S Jang
- CJ CheilJedang Corp., Food Research Institute, Suwon-si, 443-270 South Korea
| | - B S Moon
- CJ CheilJedang Corp., Food Research Institute, Suwon-si, 443-270 South Korea
| | - J J Lee
- Department of Food and Nutrition, Chosun University, Gwangju, 500-759 South Korea
| | - D E Lee
- Department of Bioscience and Biotechnology, Konkuk Univeristy, Seoul, 143-701 South Korea
| | - C H Lee
- Department of Bioscience and Biotechnology, Konkuk Univeristy, Seoul, 143-701 South Korea
| | - C S Shin
- Department of Biotechnology, Yonsei University, Seoul, 120-749 South Korea
| |
Collapse
|
26
|
Naaijkens BA, Krijnen PAJ, Meinster E, ter Horst EN, Vo K, Musters RJP, Kamp O, Niessen HWM, Juffermans LJM, van Dijk A. Acute myocardial infarction does not affect functional characteristics of adipose-derived stem cells in rats, but reduces the number of stem cells in adipose tissue. Cell Tissue Res 2015. [PMID: 26202892 PMCID: PMC4675794 DOI: 10.1007/s00441-015-2239-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In most pre-clinical animal studies investigating stem cell therapy in acute myocardial infarction (AMI), the administered stem cells are isolated from healthy donors. In clinical practice, however, patients who suffer from AMI will receive autologous cells, for example using adipose-derived stem cells (ASC). During AMI, inflammation is induced and we hypothesized that this might affect characteristics of ASC. To investigate this, ASC were isolated from rat adipose tissue 1 day (1D group, n = 5) or 7 days (7D group, n = 6) post-AMI, and were compared with ASC from healthy control rats (Control group, n = 6) and sham-operated rats (Sham 1D group, n = 5). We found that significantly fewer ASC were present 1 day post-AMI in the stromal vascular fraction (SVF), determined by a colony-forming-unit assay (p < 0.001 vs. Control and 7D). These data were confirmed by flow cytometry, showing fewer CD90-positive cells in SVF of the 1D group. When cultured, no differences were found in proliferation rate and cell size between the groups in the first three passages. Also, no difference in the differentiation capacity of ASC was found. In conclusion, it was shown that significantly fewer stem cells were present in the SVF 1 day post-AMI; however, the stem cells that were present showed no functional differences.
Collapse
Affiliation(s)
- B A Naaijkens
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands. .,Interuniversity Cardiology Institute of the Netherlands (ICIN), Utrecht, Netherlands. .,Institute of Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, Netherlands.
| | - P A J Krijnen
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands.,Institute of Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, Netherlands
| | - E Meinster
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
| | - E N ter Horst
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands.,Interuniversity Cardiology Institute of the Netherlands (ICIN), Utrecht, Netherlands.,Institute of Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, Netherlands
| | - K Vo
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
| | - R J P Musters
- Department of Physiology, VU University Medical Center, Amsterdam, Netherlands.,Institute of Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, Netherlands
| | - O Kamp
- Department of Cardiology, VU University Medical Center, Amsterdam, Netherlands.,Interuniversity Cardiology Institute of the Netherlands (ICIN), Utrecht, Netherlands
| | - H W M Niessen
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands.,Department of Cardiac Surgery, VU University Medical Center, Amsterdam, Netherlands.,Institute of Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, Netherlands
| | - L J M Juffermans
- Department of Physiology, VU University Medical Center, Amsterdam, Netherlands.,Department of Cardiology, VU University Medical Center, Amsterdam, Netherlands.,Interuniversity Cardiology Institute of the Netherlands (ICIN), Utrecht, Netherlands.,Institute of Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, Netherlands
| | - A van Dijk
- Department of Pathology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands.,Institute of Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
27
|
Lee CM, Yoon MS, Kim YC. Effects of Pueraria lobata Root Ethanol Extract on Adipogenesis and Lipogenesis During 3T3-L1 Differentiation into Adipocytes. Toxicol Res 2015; 31:191-201. [PMID: 26191386 PMCID: PMC4505350 DOI: 10.5487/tr.2015.31.2.191] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/10/2015] [Accepted: 06/14/2015] [Indexed: 01/18/2023] Open
Abstract
We evaluated the inhibitory effect of Pueraria lobata root ethanol extract (PLREE) on lipid accumulation during 3T3-L1 differentiation to adipocytes by measuring the intracellular expression of adipogenic, lipogenic, and lipolytic markers and lipid accumulation. The total polyphenol and flavonoid content of PLREE were 47 and 29 mg/g, respectively. The electron donating capacity of PLREE at 1,000 μg/mL was 48.8%. Treatment of 3T3-L1 preadipocytes with 100, 250, or 500 μg/mL PLREE for 8 days dose-dependently promoted the differentiation of 3T3-L1 cells. In contrast, the lipid content of PLREE-treated cells was significantly reduced by 7.8% (p < 0.05), 35.6% (p < 0.001), and 42.2% (p < 0.001) following treatment with 100, 250, and 500 μg/mL PLREE, respectively, as compared to differentiated control cells. PLREE upregulated peroxisome proliferator-activated receptor γ mRNA and protein, and sterol regulator element-binding protein-1c mRNA levels, but did not affect CCAAT/enhancer binding-protein β and α mRNA levels. PLREE also downregulated acetyl-CoA carboxylase mRNA and protein, fatty acid synthase (FAS) protein, and leptin mRNA levels, but did not affect FAS mRNA expression. PLREE upregulated adipose triglyceride lipase mRNA and protein expression, and hormone-sensitive lipase (HSL) protein expression, but did not affect HSL mRNA expression. In conclusion, we found that PLREE enhanced adipogenesis, but reduced lipogenesis, resulting in decreased lipid accumulation in 3T3-L1 cells.
Collapse
Affiliation(s)
- Chae Myoung Lee
- Division of Beauty Cooperation, Keimyung College University, Daegu, Korea
| | - Mi Sook Yoon
- Division of Beauty Cooperation, Keimyung College University, Daegu, Korea
| | - Young Chul Kim
- Major in Public Health, Faculty of Food & Health Sciences, Keimyung University, Daegu, Korea
| |
Collapse
|
28
|
Sundahl N, Bridelance J, Libert C, De Bosscher K, Beck IM. Selective glucocorticoid receptor modulation: New directions with non-steroidal scaffolds. Pharmacol Ther 2015; 152:28-41. [PMID: 25958032 DOI: 10.1016/j.pharmthera.2015.05.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 04/23/2015] [Indexed: 12/22/2022]
Abstract
Glucocorticoids remain the frontline treatment for inflammatory disorders, yet represent a double-edged sword with beneficial therapeutic actions alongside adverse effects, mainly in metabolic regulation. Considerable efforts were made to improve this balance by attempting to amplify therapeutic beneficial anti-inflammatory actions and to minimize adverse metabolic actions. Most attention has focused on the development of novel compounds favoring the transrepressing actions of the glucocorticoid receptor, assumed to be important for anti-inflammatory actions, over the transactivating actions, assumed to underpin the undesirable actions. These compounds are classified as selective glucocorticoid receptor agonists (SEGRAs) or selective glucocorticoid receptor modulators (SEGRMs). The latter class is able to modulate the activity of a GR agonist and/or may not classically bind the glucocorticoid receptor ligand-binding pocket. SEGRAs and SEGRMs are collectively denominated SEGRAMs (selective glucocorticoid receptor agonists and modulators). Although this transrepression vs transactivation concept proved to be too simplistic, the developed SEGRAMs were helpful in elucidating various molecular actions of the glucocorticoid receptor, but have also raised many novel questions. We discuss lessons learned from recent mechanistic studies of selective glucocorticoid receptor modulators. This is approached by analyzing recent experimental insights in comparison with knowledge obtained using mutant GR research, thus clarifying the current view on the SEGRAM field. These insights also contribute to our understanding of the processes controlling glucocorticoid-mediated side effects as well as glucocorticoid resistance. Our perspective on non-steroidal SEGRAs and SEGRMs considers remaining opportunities to address research gaps in order to harness the potential for more safe and effective glucocorticoid receptor therapies.
Collapse
Affiliation(s)
- Nora Sundahl
- Laboratory of Experimental Cancer Research (LECR), Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Gent, Belgium
| | - Jolien Bridelance
- Laboratory of Experimental Cancer Research (LECR), Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Gent, Belgium
| | - Claude Libert
- Department for Molecular Biomedical Research, VIB, Gent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Gent, Belgium
| | - Karolien De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab (NRL), VIB Department of Medical Protein Research, Ghent University, Gent, Belgium.
| | - Ilse M Beck
- Laboratory of Experimental Cancer Research (LECR), Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Gent, Belgium
| |
Collapse
|
29
|
de Seny D, Cobraiville G, Charlier E, Neuville S, Lutteri L, Le Goff C, Malaise D, Malaise O, Chapelle JP, Relic B, Malaise MG. Apolipoprotein-A1 as a damage-associated molecular patterns protein in osteoarthritis: ex vivo and in vitro pro-inflammatory properties. PLoS One 2015; 10:e0122904. [PMID: 25849372 PMCID: PMC4388661 DOI: 10.1371/journal.pone.0122904] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/24/2015] [Indexed: 12/16/2022] Open
Abstract
Osteoarthritis (OA) is associated with a local inflammatory process. Dyslipidemia is known to be an underlying cause for the development of OA. Therefore, lipid and inflammatory levels were quantified ex vivo in blood and synovial fluid of OA patients (n=29) and compared to those of rheumatoid arthritis (RA) patients (n=27) or healthy volunteers (HV) (n=35). The role of apolipoprotein A-I (ApoA1) was investigated in vitro on inflammatory parameters using human joint cells isolated from cartilage and synovial membrane obtained from OA patients after joint replacement. Cells were stimulated with ApoA1 in the presence or not of serum amyloid A (SAA) protein and/or lipoproteins (LDL and HDL) at physiological concentration observed in OA synovial fluid. In our ex vivo study, ApoA1, LDL-C and total cholesterol levels were strongly correlated to each other inside the OA joint cavity whereas same levels were not or weakly correlated to their corresponding serum levels. In OA synovial fluid, ApoA1 was not as strongly correlated to HDL as observed in OA serum or in RA synovial fluid, suggesting a dissociative level between ApoA1 and HDL in OA synovial fluid. In vitro, ApoA1 induced IL-6, MMP-1 and MMP-3 expression by primary chondrocytes and fibroblast-like synoviocytes through TLR4 receptor. HDL and LDL attenuated joint inflammatory response induced by ApoA1 and SAA in a ratio dependent manner. In conclusion, a dysregulated lipidic profile in the synovial fluid of OA patients was observed and was correlated with inflammatory parameters in the OA joint cavity. Pro-inflammatory properties of ApoA1 were confirmed in vitro.
Collapse
Affiliation(s)
- Dominique de Seny
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Gaël Cobraiville
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Edith Charlier
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Sophie Neuville
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Laurence Lutteri
- Laboratory of Clinical Chemistry, CHU Hospital of Liege, 4000 Liège, Belgium
| | - Caroline Le Goff
- Laboratory of Clinical Chemistry, CHU Hospital of Liege, 4000 Liège, Belgium
| | - Denis Malaise
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Olivier Malaise
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Jean-Paul Chapelle
- Laboratory of Clinical Chemistry, CHU Hospital of Liege, 4000 Liège, Belgium
| | - Biserka Relic
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Michel G. Malaise
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| |
Collapse
|
30
|
Affiliation(s)
- Salvatore Chirumbolo
- Laboratory of Physiopathology of Obesity, Department of Medicine, University of Verona, LURM Est Policlinico GB Rossi, Piazzale Al Scuro 10, 37134, Verona, Italy,
| |
Collapse
|
31
|
Malaise O, Relic B, Quesada-Calvo F, Charlier E, Zeddou M, Neuville S, Gillet P, Louis E, de Seny D, Malaise MG. Selective glucocorticoid receptor modulator compound A, in contrast to prednisolone, does not induce leptin or the leptin receptor in human osteoarthritis synovial fibroblasts. Rheumatology (Oxford) 2014; 54:1087-92. [PMID: 25389358 DOI: 10.1093/rheumatology/keu428] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Glucocorticoids are powerful anti-inflammatory compounds that also induce the expression of leptin and leptin receptor (Ob-R) in synovial fibroblasts through TGF-βsignalling and Smad1/5 phosphorylation. Compound A (CpdA), a selective glucocorticoid receptor agonist, reduces inflammation in murine arthritis models and does not induce diabetes or osteoporosis, thus offering an improved risk:benefit ratio in comparison with glucocorticoids. Due to the detrimental role of leptin in OA pathogenesis, we sought to determine whether CpdA also induced leptin and Ob-R protein expression as observed with prednisolone. METHODS Human synovial fibroblasts and chondrocytes were isolated from the synovium and cartilage of OA patients after joint surgery. The cells were treated with prednisolone, TGF-β1, TNF-α and/or CpdA. Levels of leptin, IL-6, IL-8, MMP-1 and MMP-3 were measured by ELISA and expression levels of Ob-R phospho-Smad1/5, phospho-Smad2, α-tubulin and glyceraldehyde 3-phosphate dehydrogenase were analysed by western blotting. RESULTS CpdA, unlike prednisolone, did not induce leptin secretion or Ob-R protein expression in OA synovial fibroblasts. Moreover, CpdA decreased endogenous Ob-R expression and down-regulated prednisolone-induced leptin secretion and Ob-R expression. Mechanistically, CpdA, unlike prednisolone, did not induce Smad1/5 phosphorylation. CpdA, similarly to prednisolone, down-regulated endogenous and TNF-α-induced IL-6, IL-8, MMP-1 and MMP-3 protein secretion. The dissociative effect of CpdA was confirmed using chondrocytes with no induction of leptin secretion, but with a significant decrease in IL-6, IL-8, MMP-1 and MMP-3 protein secretion. CONCLUSION CpdA, unlike prednisolone, did not induce leptin or Ob-R in human OA synovial fibroblasts, thereby demonstrating an improved risk:benefit ratio.
Collapse
Affiliation(s)
- Olivier Malaise
- Laboratory of Rheumatology and Laboratory of Gastroenterology, GIGA Research, CHU, University of Liège and Department of Orthopedic Surgery, CHU of Liège, CHU, Sart- Tilman B35, Liège, Belgium
| | - Biserka Relic
- Laboratory of Rheumatology and Laboratory of Gastroenterology, GIGA Research, CHU, University of Liège and Department of Orthopedic Surgery, CHU of Liège, CHU, Sart- Tilman B35, Liège, Belgium
| | - Florence Quesada-Calvo
- Laboratory of Rheumatology and Laboratory of Gastroenterology, GIGA Research, CHU, University of Liège and Department of Orthopedic Surgery, CHU of Liège, CHU, Sart- Tilman B35, Liège, Belgium
| | - Edith Charlier
- Laboratory of Rheumatology and Laboratory of Gastroenterology, GIGA Research, CHU, University of Liège and Department of Orthopedic Surgery, CHU of Liège, CHU, Sart- Tilman B35, Liège, Belgium
| | - Mustapha Zeddou
- Laboratory of Rheumatology and Laboratory of Gastroenterology, GIGA Research, CHU, University of Liège and Department of Orthopedic Surgery, CHU of Liège, CHU, Sart- Tilman B35, Liège, Belgium
| | - Sophie Neuville
- Laboratory of Rheumatology and Laboratory of Gastroenterology, GIGA Research, CHU, University of Liège and Department of Orthopedic Surgery, CHU of Liège, CHU, Sart- Tilman B35, Liège, Belgium
| | - Philippe Gillet
- Laboratory of Rheumatology and Laboratory of Gastroenterology, GIGA Research, CHU, University of Liège and Department of Orthopedic Surgery, CHU of Liège, CHU, Sart- Tilman B35, Liège, Belgium
| | - Edouard Louis
- Laboratory of Rheumatology and Laboratory of Gastroenterology, GIGA Research, CHU, University of Liège and Department of Orthopedic Surgery, CHU of Liège, CHU, Sart- Tilman B35, Liège, Belgium
| | - Dominique de Seny
- Laboratory of Rheumatology and Laboratory of Gastroenterology, GIGA Research, CHU, University of Liège and Department of Orthopedic Surgery, CHU of Liège, CHU, Sart- Tilman B35, Liège, Belgium
| | - Michel G Malaise
- Laboratory of Rheumatology and Laboratory of Gastroenterology, GIGA Research, CHU, University of Liège and Department of Orthopedic Surgery, CHU of Liège, CHU, Sart- Tilman B35, Liège, Belgium
| |
Collapse
|
32
|
Li R, El Zowalaty AE, Chen W, Dudley EA, Ye X. Segregated responses of mammary gland development and vaginal opening to prepubertal genistein exposure in Bscl2(-/-) female mice with lipodystrophy. Reprod Toxicol 2014; 54:76-83. [PMID: 25462787 DOI: 10.1016/j.reprotox.2014.10.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 09/12/2014] [Accepted: 10/19/2014] [Indexed: 10/24/2022]
Abstract
Berardinelli-Seip congenital lipodystrophy 2-deficient (Bscl2(-/-)) mice recapitulate human BSCL2 disease with lipodystrophy. Bscl2-encoded seipin is detected in adipocytes and epithelium of mammary gland. Postnatal mammary gland growth spurt and vaginal opening signify pubertal onset in female mice. Bscl2(-/-) females have longer and dilated mammary gland ducts at 5-week old and delayed vaginal opening. Prepubertal exposure to 500ppm genistein diet increases mammary gland area and accelerates vaginal opening in both control and Bscl2(-/-) females. However, genistein treatment increases ductal length in control but not Bscl2(-/-) females. Neither prepubertal genistein treatment nor Bscl2-deficiency affects phospho-estrogen receptor α or progesterone receptor expression patterns in 5-week old mammary gland. Interestingly, Bscl2-deficiency specifically reduces estrogen receptor β expression in mammary gland ductal epithelium. In summary, Bscl2(-/-) females have accelerated postnatal mammary ductal development but delayed vaginal opening; they display segregated responses in mammary gland development and vaginal opening to prepubertal genistein treatment.
Collapse
Affiliation(s)
- Rong Li
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; Interdisciplinary Toxicology Program, University of Georgia, Athens, GA 30602, USA.
| | - Ahmed E El Zowalaty
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; Interdisciplinary Toxicology Program, University of Georgia, Athens, GA 30602, USA.
| | - Weiqin Chen
- Department of Physiology, Georgia Regents University, Augusta, GA 30912, USA.
| | - Elizabeth A Dudley
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; Interdisciplinary Toxicology Program, University of Georgia, Athens, GA 30602, USA.
| | - Xiaoqin Ye
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; Interdisciplinary Toxicology Program, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
33
|
de Seny D, Cobraiville G, Charlier E, Neuville S, Esser N, Malaise D, Malaise O, Calvo FQ, Relic B, Malaise MG. Acute-phase serum amyloid a in osteoarthritis: regulatory mechanism and proinflammatory properties. PLoS One 2013; 8:e66769. [PMID: 23776697 PMCID: PMC3680431 DOI: 10.1371/journal.pone.0066769] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 05/11/2013] [Indexed: 01/01/2023] Open
Abstract
Objective To determine if serum amyloid A (A-SAA) could be detected in human osteoarthritic (OA) joints and further clarify if high A-SAA level in joints result from a local production or from a diffusion process from abnormally elevated plasma concentration. Regulatory mechanism of A-SAA expression and its pro-inflammatory properties were also investigated. Methods A-SAA levels in serum and synovial fluid of OA (n = 29) and rheumatoid arthritis (RA) (n = 27) patients were measured and compared to matched-healthy volunteers (HV) (n = 35). In vitro cell cultures were performed on primary joint cells provided from osteoarthritis patients. Regulatory mechanisms were studied using Western-blotting, ELISA and lentiviral transfections. Results A-SAA was statistically increased in OA plasma patients compared to HV. Moreover, A-SAA level in OA plasma and synovial fluid increased with the Kellgren & Lauwrence grade. For all OA and RA patients, A-SAA plasma level was higher and highly correlated with its corresponding level in the synovial fluid, therefore supporting that A-SAA was mainly due to the passive diffusion process from blood into the joint cavity. However, A-SAA expression was also observed in vitro under corticosteroid treatment and/or under IL-1beta stimuli. A-SAA expression was down-regulated by PPAR-γ agonists (genistein and rosiglitazone) and up-regulated by TGF-β1 through Alk1 (Smad1/5) pathway. RhSAA induced proinflammatory cytokines (IL-6, IL-8, GRO-α and MCP-1) and metalloproteinases (MMP-1, MMP-3 and MMP-13) expression in FLS and chondrocytes, which expression was downregulated by TAK242, a specific TLR4 inhibitor. Conclusion Systemic or local A-SAA expression inside OA joint cavity may play a key role in inflammatory process seen in osteoarthritis, which could be counteracted by TLR4 inhibition.
Collapse
Affiliation(s)
- Dominique de Seny
- Laboratory of Rheumatology, GIGA Research, University of Liège, CHU Liège, Liège, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Dietary relevant mixtures of phytoestrogens inhibit adipocyte differentiation in vitro. Food Chem Toxicol 2013; 55:265-71. [DOI: 10.1016/j.fct.2012.12.060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 12/20/2012] [Accepted: 12/22/2012] [Indexed: 11/19/2022]
|
35
|
Behloul N, Wu G. Genistein: A promising therapeutic agent for obesity and diabetes treatment. Eur J Pharmacol 2013. [DOI: 10.1016/j.ejphar.2012.11.013] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
36
|
Gencel VB, Benjamin MM, Bahou SN, Khalil RA. Vascular effects of phytoestrogens and alternative menopausal hormone therapy in cardiovascular disease. Mini Rev Med Chem 2012; 12:149-74. [PMID: 22070687 DOI: 10.2174/138955712798995020] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 07/27/2011] [Accepted: 08/05/2011] [Indexed: 01/10/2023]
Abstract
Phytoestrogens are estrogenic compounds of plant origin classified into different groups including isoflavones, lignans, coumestans and stilbenes. Isoflavones such as genistein and daidzein are the most studied and most potent phytoestrogens, and are found mainly in soy based foods. The effects of phytoestrogens are partly mediated via estrogen receptors (ERs): ERα, ERβ and possibly GPER. The interaction of phytoestrogens with ERs is thought to induce both genomic and non-genomic effects in many tissues including the vasculature. Some phytoestrogens such as genistein have additional non-ER-mediated effects involving signaling pathways such as tyrosine kinase. Experimental studies have shown beneficial effects of phytoestrogens on endothelial cells, vascular smooth muscle, and extracellular matrix. Phytoestrogens may also affect other pathophysiologic vascular processes such as lipid profile, angiogenesis, inflammation, tissue damage by reactive oxygen species, and these effects could delay the progression of atherosclerosis. As recent clinical trials showed no vascular benefits or even increased risk of cardiovascular disease (CVD) and CV events with conventional menopausal hormone therapy (MHT), phytoestrogens are being considered as alternatives to pharmacologic MHT. Epidemiological studies in the Far East population suggest that dietary intake of phytoestrogens may contribute to the decreased incidence of postmenopausal CVD and thromboembolic events. Also, the WHO-CARDIAC study supported that consumption of high soybean diet is associated with lower mortalities from coronary artery disease. However, as with estrogen, there has been some discrepancy between the experimental studies demonstrating the vascular benefits of phytoestrogens and the data from clinical trials. This is likely because the phytoestrogens clinical trials have been limited in many aspects including the number of participants enrolled, the clinical end points investigated, and the lack of long-term follow-up. Further investigation of the cellular mechanisms underlying the vascular effects of phytoestrogens and careful evaluation of the epidemiological evidence and clinical trials of their potential vascular benefits would put forward the use of phytoestrogens as an alternative MHT for the relief of menopausal symptoms and amelioration of postmenopausal CVD.
Collapse
Affiliation(s)
- V B Gencel
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
37
|
Leiherer A, Mündlein A, Drexel H. Phytochemicals and their impact on adipose tissue inflammation and diabetes. Vascul Pharmacol 2012; 58:3-20. [PMID: 22982056 DOI: 10.1016/j.vph.2012.09.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 09/03/2012] [Accepted: 09/04/2012] [Indexed: 01/17/2023]
Abstract
Type 2 diabetes mellitus is an inflammatory disease and the mechanisms that underlie this disease, although still incompletely understood, take place in the adipose tissue of obese subjects. Concurrently, the prevalence of obesity caused by Western diet's excessive energy intake and the lack of exercise escalates, and is believed to be causative for the chronic inflammatory state in adipose tissue. Overnutrition itself as an overload of energy may induce the adipocytes to secrete chemokines activating and attracting immune cells to adipose tissue. But also inflammation-mediating food ingredients like saturated fatty acids are believed to directly initiate the inflammatory cascade. In addition, hypoxia in adipose tissue as a direct consequence of obesity, and its effect on gene expression in adipocytes and surrounding cells in fat tissue of obese subjects appears to play a central role in this inflammatory response too. In contrast, revisiting diet all over the world, there are also some natural food products and beverages which are associated with curative effects on human health. Several natural compounds known as spices such as curcumin, capsaicin, and gingerol, or secondary plant metabolites catechin, resveratrol, genistein, and quercetin have been reported to provide an improved health status to their consumers, especially with regard to diabetes, and therefore have been investigated for their anti-inflammatory effect. In this review, we will give an overview about these phytochemicals and their role to interfere with inflammatory cascades in adipose tissue and their potential for fighting against inflammatory diseases like diabetes as investigated in vivo.
Collapse
Affiliation(s)
- Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment, Feldkirch, Austria
| | | | | |
Collapse
|
38
|
Guo W, Li Y, Liang W, Wong S, Apovian C, Kirkland JL, Corkey BE. Beta-mecaptoethanol suppresses inflammation and induces adipogenic differentiation in 3T3-F442A murine preadipocytes. PLoS One 2012; 7:e40958. [PMID: 22911724 PMCID: PMC3402440 DOI: 10.1371/journal.pone.0040958] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 06/15/2012] [Indexed: 12/25/2022] Open
Abstract
Preadipocytes are present in adipose tissues throughout adult life that can proliferate and differentiate into mature adipocytes in response to environmental cues. Abnormal increase in adipocyte number or size leads to fat tissue expansion. However, it is now recognized that adipocyte hypertrophy is a greater risk factor for metabolic syndrome whereas fat tissue that continues to produce newer and smaller fat cells through preadipocyte differentiation is “metabolically healthy”. Because adipocyte hypertrophy is often associated with increased oxidant stress and low grade inflammation, both are linked to disturbed cellular redox, we tested how preadipocyte differentiation may be regulated by beta-mercaptoethanol (BME), a pharmacological redox regulator and radical scavenger, using murine 3T3-F442A preadipocytes as the cell model. Effects of BME on adipogenesis were measured by microphotography, real-time PCR, and Western analysis. Our data demonstrated that preadipocyte differentiation could be regulated by extracellular BME. At an optimal concentration, BME enhanced expression of adipogenic gene markers and lipid accumulation. This effect was associated with BME-mediated down-regulation of inflammatory cytokine expression during early differentiation. BME also attenuated TNFalpha-induced activation of NFkappaB in differentiating preadipocytes and partially restored TNFalpha-mediated suppression on adipogenesis. Using a non-adipogenic HEK293 cell line transfected with luciferase reporter genes, we demonstrated that BME reduced basal and TNFalpha-induced NFkappaB activity and increased basal and ciglitazone-induced PPARgamma activity; both may contribute to the pro-adipogenic effect of BME in differentiating F442A preadipocytes.
Collapse
Affiliation(s)
- Wen Guo
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | |
Collapse
|
39
|
Skelhorne-Gross G, Reid AL, Apostoli AJ, Di Lena MA, Rubino RE, Peterson NT, Schneider M, SenGupta SK, Gonzalez FJ, Nicol CJB. Stromal adipocyte PPARγ protects against breast tumorigenesis. Carcinogenesis 2012; 33:1412-20. [PMID: 22581835 DOI: 10.1093/carcin/bgs173] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR)γ regulates the expression of genes essential for fat storage, primarily through its activity in adipocytes. It also has a role in carcinogenesis. PPARγ normally stops the in vivo progression of 7,12-dimethylbenz[a]anthracene (DMBA)-mediated breast tumours as revealed with PPARγ haploinsufficient mice. Since many cell types associated with the mammary gland express PPARγ, each with unique signal patterns, this study aimed to define which tissues are required for PPARγ-dependent antitumour effects. Accordingly, adipocyte-specific PPARγ knockout (PPARγ-A KO) mice and their wild-type (PPARγ-WT) controls were generated, and treated with DMBA for 6 weeks to initiate breast tumorigenesis. On week 7, mice were randomized to continue on normal chow diet or one supplemented with rosiglitazone (ROSI), and followed for 25 weeks for tumour outcomes. In PPARγ-A KO versus PPARγ-WT mice, malignant mammary tumour incidence was significantly higher and mammary tumour latency was decreased. DMBA + ROSI treatment reduced average mammary tumour volumes by 50%. Gene expression analyses of mammary glands by quantitative real-time polymerase chain reaction and immunofluorescence indicated that untreated PPARγ-A KOs had significantly decreased BRCA1 expression in mammary stromal adipocytes. Compared with PPARγ-WT mice, serum leptin levels in PPARγ-A KOs were also significantly higher throughout the study. Together, these data are the first to suggest that in vivo PPARγ expression in mammary stromal adipocytes attenuates breast tumorigenesis through BRCA1 upregulation and decreased leptin secretion. This study supports a protective effect of activating PPARγ as a novel chemopreventive therapy for breast cancer.
Collapse
Affiliation(s)
- Graham Skelhorne-Gross
- Department of Pathology and Molecular Medicine, Richardson Laboratories, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kontny E, Plebanczyk M, Lisowska B, Olszewska M, Maldyk P, Maslinski W. Comparison of rheumatoid articular adipose and synovial tissue reactivity to proinflammatory stimuli: contribution to adipocytokine network. Ann Rheum Dis 2012; 71:262-7. [PMID: 21989538 DOI: 10.1136/annrheumdis-2011-200123] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVES (1) To compare spontaneous and stimuli-induced adipocytokine secretion by articular adipose tissue (AAT) and synovial membrane (SM) explants obtained from patients with rheumatoid arthritis (RA). (2) To investigate the biological activity of AAT and SM released factors. METHODS Tissues were obtained from patients undergoing joint replacement surgery. Tissue explants were treated with proinflammatory cytokines relevant to RA pathogenesis (interleukin 1β (IL-1β), tumour necrosis factor (TNF), interferon γ, IL-15, IL-17, IL-23). Selected adipocytokine (TNF, IL-6, IL-8, IL-1β, IL-1Ra, adiponectin, leptin) concentrations were measured in culture supernatants using ELISA. The biological activity of tissue-conditioned media was evaluated by measuring production of selected factors (IL-6, IL-8, Dickkopf-1, osteoprotegerin) by fibroblast-like synoviocytes (FLS). RESULTS Spontaneous cytokine release from AAT was ≤12% of that produced by SM, while leptin was secreted in similar amounts. AAT was highly reactive to proinflammatory cytokines (IL-1β>TNF). AAT treated with IL-1β released four times more leptin, similar amounts of IL-6 and IL-8 and about 20% of TNF, as compared with SM. Upon activation, the IL-1 receptor antagonist (IL-1Ra)/IL-1β ratio was higher in AAT than in SM cultures. Irrespective of activation status, SM produced twice as much adiponectin as AAT. Conditioned media from AAT and SM cultures similarly upregulated IL-6, IL-8, Dickkopf-1 and osteoprotegerin production by rheumatoid FLS. CONCLUSION Rheumatoid AAT is highly reactive tissue which upon stimulation secretes considerable amounts of proinflammatory (IL-6, IL-8, TNF) and anti-inflammatory (IL-1Ra) cytokines and classical adipokines. This tissue releases biologically active factors that intensify pathogenic activities of rheumatoid FLS. Thus, AAT should be considered an important contributor to the pathological processes taking place in the RA joint.
Collapse
Affiliation(s)
- Ewa Kontny
- Department of Pathophysiology and Immunology, Institute of Rheumatology, Warsaw, Poland.
| | | | | | | | | | | |
Collapse
|
41
|
Zeddou M, Relic B, Malaise O, Charlier E, Desoroux A, Beguin Y, de Seny D, Malaise MG. Differential signalling through ALK-1 and ALK-5 regulates leptin expression in mesenchymal stem cells. Stem Cells Dev 2011; 21:1948-55. [PMID: 22087763 DOI: 10.1089/scd.2011.0321] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Leptin plays a central role in maintaining energy balance, with multiple other systemic effects. Despite leptin importance in peripheral regulation of mesenchymal stem cells (MSC) differentiation, little is known about its expression mechanism. Leptin is often described as adipokine, while it is expressed by other cell types. We have recently shown an in vitro leptin expression, enhanced by glucocorticoids in synovial fibroblasts (SVF). Here, we investigated leptin expression in MSC from bone marrow (BM-MSC) and umbilical cord matrix (UMSC). Results showed that BM-MSC, but not UMSC, expressed leptin that was strongly enhanced by glucocorticoids. Transforming growth factor β1 (TGF-β1) markedly inhibited the endogenous- and glucocorticoid-induced leptin expression in BM-MSC. Since TGF-β1 was shown to signal via ALK-5-Smad2/3 and/or ALK-1-Smad1/5 pathways, we analyzed the expression of proteins from both pathways. In BM-MSC, TGF-β1 increased phosphorylated Smad2 (p-Smad2) expression, while ALK-5 inhibitor (SB431542) induced leptin expression and significantly restored TGF-β1-induced leptin inhibition. In addition, both prednisolone and SB431542 increased p-Smad1/5 expression. These results suggested the ALK-5-Smad2 pathway as an inhibitor of leptin expression, while ALK-1-Smad1/5 as an activator. Indeed, Smad1 expression silencing induced leptin expression inhibition. Furthermore, prednisolone enhanced the expression of TGF-βRII while decreasing p-Smad2 in BM-MSC and SVF but not in UMSC. In vitro differentiation revealed differential osteogenic potential in SVF, BM-MSC, and UMSC that was correlated to their leptin expression potential. Our results suggest that ALK-1/ALK-5 balance regulates leptin expression in MSC. It also underlines UMSC as leptin nonproducer MSC for cell therapy protocols where leptin expression is not suitable.
Collapse
Affiliation(s)
- Mustapha Zeddou
- Laboratory of Rheumatology, GIGA-I3, GIGA Research Centre, University and CHU of Liège, Liège, Belgium
| | | | | | | | | | | | | | | |
Collapse
|