1
|
Boccanegra B, Cappellari O, Mantuano P, Trisciuzzi D, Mele A, Tulimiero L, De Bellis M, Cirmi S, Sanarica F, Cerchiara AG, Conte E, Meanti R, Rizzi L, Bresciani E, Denoyelle S, Fehrentz JA, Cruciani G, Nicolotti O, Liantonio A, Torsello A, De Luca A. Growth hormone secretagogues modulate inflammation and fibrosis in mdx mouse model of Duchenne muscular dystrophy. Front Immunol 2023; 14:1119888. [PMID: 37122711 PMCID: PMC10130389 DOI: 10.3389/fimmu.2023.1119888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Growth hormone secretagogues (GHSs) exert multiple actions, being able to activate GHS-receptor 1a, control inflammation and metabolism, to enhance GH/insulin-like growth factor-1 (IGF-1)-mediated myogenesis, and to inhibit angiotensin-converting enzyme. These mechanisms are of interest for potentially targeting multiple steps of pathogenic cascade in Duchenne muscular dystrophy (DMD). Methods Here, we aimed to provide preclinical evidence for potential benefits of GHSs in DMD, via a multidisciplinary in vivo and ex vivo comparison in mdx mice, of two ad hoc synthesized compounds (EP80317 and JMV2894), with a wide but different profile. 4-week-old mdx mice were treated for 8 weeks with EP80317 or JMV2894 (320 µg/kg/d, s.c.). Results In vivo, both GHSs increased mice forelimb force (recovery score, RS towards WT: 20% for EP80317 and 32% for JMV2894 at week 8). In parallel, GHSs also reduced diaphragm (DIA) and gastrocnemius (GC) ultrasound echodensity, a fibrosis-related parameter (RS: ranging between 26% and 75%). Ex vivo, both drugs ameliorated DIA isometric force and calcium-related indices (e.g., RS: 40% for tetanic force). Histological analysis highlighted a relevant reduction of fibrosis in GC and DIA muscles of treated mice, paralleled by a decrease in gene expression of TGF-β1 and Col1a1. Also, decreased levels of pro-inflammatory genes (IL-6, CD68), accompanied by an increment in Sirt-1, PGC-1α and MEF2c expression, were observed in response to treatments, suggesting an overall improvement of myofiber metabolism. No detectable transcript levels of GHS receptor-1a, nor an increase of circulating IGF-1 were found, suggesting the presence of a novel receptor-independent mechanism in skeletal muscle. Preliminary docking studies revealed a potential binding capability of JMV2894 on metalloproteases involved in extracellular matrix remodeling and cytokine production, such as ADAMTS-5 and MMP-9, overactivated in DMD. Discussion Our results support the interest of GHSs as modulators of pathology progression in mdx mice, disclosing a direct anti-fibrotic action that may prove beneficial to contrast pathological remodeling.
Collapse
Affiliation(s)
- Brigida Boccanegra
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Ornella Cappellari
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Paola Mantuano
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Daniela Trisciuzzi
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Antonietta Mele
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Lisamaura Tulimiero
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Michela De Bellis
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Santa Cirmi
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Francesca Sanarica
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | | | - Elena Conte
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Ramona Meanti
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Severine Denoyelle
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier, France
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Orazio Nicolotti
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Antonella Liantonio
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Annamaria De Luca
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
2
|
Chen W, Huang C, Shi Y, Li N, Wang E, Hu R, Li G, Yang F, Zhuang Y, Liu P, Hu G, Gao X, Guo X. Investigation of the Crosstalk between GRP78/PERK/ATF-4 Signaling Pathway and Renal Apoptosis Induced by Nephropathogenic Infectious Bronchitis Virus Infection. J Virol 2022; 96:e0142921. [PMID: 34669445 PMCID: PMC8791289 DOI: 10.1128/jvi.01429-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/16/2021] [Indexed: 11/20/2022] Open
Abstract
This study aims to explore the crosstalk between GRP78/PERK/ATF-4 signaling pathway and renal apoptosis induced by nephropathogenic infectious bronchitis virus (NIBV). Hy-Line brown chickens were divided into two groups (Con, n = 100 and Dis, n = 200). At 28 days of age, each chicken in the Dis group was intranasally injected with SX9 strain (10-5/0.2 ml). Venous blood and kidney tissues were collected at 1, 5, 11, 18 and 28 days postinfection. Our results showed that NIBV infection upregulated the levels of creatinine, uric acid, and calcium (Ca2+) levels. Histopathological examination revealed severe hemorrhage and inflammatory cell infiltration near the renal tubules. Meanwhile, NIBV virus particles and apoptotic bodies were observed by ultramicro electron microscope. In addition, RT-qPCR and Western blot showed that NIBV upregulated the expression of GRP78, PERK, eIF2α, ATF-4, CHOP, Caspase-3, Caspase-9, P53, Bax, and on the contrary, downregulated the expression of Bcl-2. Furthermore, immunofluorescence localization analysis showed that the positive expression of Bcl-2 protein was significantly decreased. Correlation analysis indicated that endoplasmic reticulum (ER) stress gene expression, apoptosis gene expression, and renal injury were potentially related. Taken together, NIBV infection can induce renal ER stress and apoptosis by activating of GRP78/PERK/ATF-4 signaling pathway, leading to kidney damage. IMPORTANCE Nephropathogenic infectious bronchitis virus (NIBV) induced renal endoplasmic reticulum stress in chickens. NIBV infection induced kidney apoptosis in chickens. GRP78/PERK/ATF-4 signaling pathway is potentially related to renal apoptosis induced by NIBV.
Collapse
Affiliation(s)
- Wei Chen
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Cheng Huang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Yan Shi
- School of Computer and Information Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Ning Li
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Enqi Wang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Ruiming Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Guyue Li
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Fan Yang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Yu Zhuang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Ping Liu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Xiaona Gao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| |
Collapse
|
3
|
Napolitano T, Avolio F, Silvano S, Forcisi S, Pfeifer A, Vieira A, Navarro-Sanz S, Friano ME, Ayachi C, Garrido-Utrilla A, Atlija J, Hadzic B, Becam J, Sousa-De-Veiga A, Plaisant MD, Balaji S, Pisani DF, Mondin M, Schmitt-Kopplin P, Amri EZ, Collombat P. Gfi1 Loss Protects against Two Models of Induced Diabetes. Cells 2021; 10:cells10112805. [PMID: 34831029 PMCID: PMC8616283 DOI: 10.3390/cells10112805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/07/2021] [Accepted: 10/14/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Although several approaches have revealed much about individual factors that regulate pancreatic development, we have yet to fully understand their complicated interplay during pancreas morphogenesis. Gfi1 is transcription factor specifically expressed in pancreatic acinar cells, whose role in pancreas cells fate identity and specification is still elusive. Methods: In order to gain further insight into the function of this factor in the pancreas, we generated animals deficient for Gfi1 specifically in the pancreas. Gfi1 conditional knockout animals were phenotypically characterized by immunohistochemistry, RT-qPCR, and RNA scope. To assess the role of Gfi1 in the pathogenesis of diabetes, we challenged Gfi1-deficient mice with two models of induced hyperglycemia: long-term high-fat/high-sugar feeding and streptozotocin injections. Results: Interestingly, mutant mice did not show any obvious deleterious phenotype. However, in depth analyses demonstrated a significant decrease in pancreatic amylase expression, leading to a diminution in intestinal carbohydrates processing and thus glucose absorption. In fact, Gfi1-deficient mice were found resistant to diet-induced hyperglycemia, appearing normoglycemic even after long-term high-fat/high-sugar diet. Another feature observed in mutant acinar cells was the misexpression of ghrelin, a hormone previously suggested to exhibit anti-apoptotic effects on β-cells in vitro. Impressively, Gfi1 mutant mice were found to be resistant to the cytotoxic and diabetogenic effects of high-dose streptozotocin administrations, displaying a negligible loss of β-cells and an imperturbable normoglycemia. Conclusions: Together, these results demonstrate that Gfi1 could turn to be extremely valuable for the development of new therapies and could thus open new research avenues in the context of diabetes research.
Collapse
Affiliation(s)
- Tiziana Napolitano
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Fabio Avolio
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark;
| | - Serena Silvano
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Sara Forcisi
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, German Research Center for Environment Health, 85764 Neuherberg, Germany; (S.F.); (P.S.-K.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Anja Pfeifer
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Andhira Vieira
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | | | - Marika Elsa Friano
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Chaïma Ayachi
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Anna Garrido-Utrilla
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | | | - Biljana Hadzic
- Pediatric Oncology & Hematology Department, Centre Hospitalier Universitaire de Nice, Hopital Archet 2, 06202 Nice, France;
| | - Jérôme Becam
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Anette Sousa-De-Veiga
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Magali Dodille Plaisant
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | | | - Didier F. Pisani
- Medicine Faculty, Université Côte d’Azur, CNRS, LP2M, 06003 Nice, France;
| | - Magali Mondin
- Pôle Imagerie Photonique, Bordeaux Imaging Center, Université de Bordeaux, UMS 3420 CNRS-US4 Inserm, 33076 Bordeaux, France;
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, German Research Center for Environment Health, 85764 Neuherberg, Germany; (S.F.); (P.S.-K.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ez-Zoubir Amri
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
| | - Patrick Collombat
- Faculté des Sciences, Université Côte d’Azur, CNRS, Inserm, iBV, Parc Valrose, 06108 Nice, France; (T.N.); (S.S.); (A.P.); (A.V.); (M.E.F.); (C.A.); (A.G.-U.); (J.B.); (A.S.-D.-V.); (M.D.P.); (E.-Z.A.)
- Correspondence:
| |
Collapse
|
4
|
Yang Z, Li X, Zhang C, Sun N, Guo T, Lin J, Li F, Zhang J. Amniotic Membrane Extract Protects Islets From Serum-Deprivation Induced Impairments and Improves Islet Transplantation Outcome. Front Endocrinol (Lausanne) 2020; 11:587450. [PMID: 33363516 PMCID: PMC7753361 DOI: 10.3389/fendo.2020.587450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/04/2020] [Indexed: 11/25/2022] Open
Abstract
Islet culture prior to transplantation is a standard practice in many transplantation centers. Nevertheless, the abundant islet mass loss and function impairment during this serum-deprivation culture period restrain the success of islet transplantation. In the present study, we used a natural biomaterial derived product, amniotic membrane extract (AME), as medium supplementation of islet pretransplant cultivation to investigate its protective effect on islet survival and function and its underlying mechanisms, as well as the engraftment outcome of islets following AME treatment. Results showed that AME supplementation improved islet viability and function, and decreased islet apoptosis and islet loss during serum-deprived culture. This was associated with the increased phosphorylation of PI3K/Akt and MAPK/ERK signaling pathway. Moreover, transplantation of serum-deprivation stressed islets that were pre-treated with AME into diabetic mice revealed better blood glucose control and improved islet graft survival. In conclusion, AME could improve islet survival and function in vivo and in vitro, and was at least partially through increasing phosphorylation of PI3K/Akt and MAPK/ERK signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jialin Zhang
- Department of Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Kou H, Gui S, Dai Y, Guo Y, Wang H. Epigenetic repression of AT2 receptor is involved in β cell dysfunction and glucose intolerance of adult female offspring rats exposed to dexamethasone prenatally. Toxicol Appl Pharmacol 2020; 404:115187. [PMID: 32791177 DOI: 10.1016/j.taap.2020.115187] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/22/2020] [Accepted: 08/07/2020] [Indexed: 12/30/2022]
Abstract
Prenatal exposure to dexamethasone (PDE) impairs pancreatic β cell development and glucose homeostasis in offspring especially females. To explore the underlying intrauterine programming mechanism, pregnant Wistar rats were subcutaneously administered with dexamethasone (0, 0.2 and 0.8 mg/kg·d) from gestational days (GD) 9 to 20. Female offspring were collected on GD20 (fetus) and in postnatal week 28 (adult), respectively. PDE reduced the serum insulin levels, β cell mass, and pancreatic insulin expressions in fetuses and adults, causing glucose intolerance after maturity. The persistent suppression of pancreatic angiotensin II receptor type 2 (AT2R) expression before and after birth could be observed in the PDE females, which is accompanied with decreased histone 3 lysine 14 acetylation (H3K14ac) and H3K27ac levels in AT2R promoter. PDE increased the gene expressions of glucocorticoid receptor (GR) and histone deacetylase 2 (HDAC2) in fetal pancreas. Furthermore, dexamethasone inhibited insulin biosynthesis while activated GR and HDAC2 expression in the rat INS-1 cells. The AT2R expression was repressed by dexamethasone in vitro but only H3K27ac levels in AT2R promoter were lowered. Dexamethasone enhanced the interaction between GR and HDAC2 proteins as well as the binding of GR/HDAC2 complex to AT2R promoter. Moreover, overexpression of AT2R could restore the suppressed insulin biosynthesis induced by dexamethasone in vitro, and both GR antagonist and histone deacetylase abolished the decreased H3K27ac level and gene expression of AT2R. In conclusion, continuous epigenetic repression of AT2R before and after birth may be involved in β cell dysfunction and glucose intolerance of the PDE adult female offspring.
Collapse
Affiliation(s)
- Hao Kou
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan 40071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Shuxia Gui
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yongguo Dai
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yu Guo
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China.
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China.
| |
Collapse
|
6
|
Yao X, Li K, Liang C, Zhou Z, Wang J, Wang S, Liu L, Yu CL, Song ZB, Bao YL, Zheng LH, Sun Y, Wang G, Huang Y, Yi J, Sun L, Li Y. Tectorigenin enhances PDX1 expression and protects pancreatic β-cells by activating ERK and reducing ER stress. J Biol Chem 2020; 295:12975-12992. [PMID: 32690606 DOI: 10.1074/jbc.ra120.012849] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/16/2020] [Indexed: 11/06/2022] Open
Abstract
Pancreas/duodenum homeobox protein 1 (PDX1) is an important transcription factor that regulates islet β-cell proliferation, differentiation, and function. Reduced expression of PDX1 is thought to contribute to β-cell loss and dysfunction in diabetes. Thus, promoting PDX1 expression can be an effective strategy to preserve β-cell mass and function. Previously, we established a PDX1 promoter-dependent luciferase system to screen agents that can promote PDX1 expression. Natural compound tectorigenin (TG) was identified as a promising candidate that could enhance the activity of the promoter for the PDX1 gene. In this study, we first demonstrated that TG could promote the expression of PDX1 in β-cells via activating extracellular signal-related kinase (ERK), as indicated by increased phosphorylation of ERK; this effect was observed under either normal or glucotoxic/lipotoxic conditions. We then found that TG could suppress induced apoptosis and improved the viability of β-cells under glucotoxicity and lipotoxicity by activation of ERK and reduction of reactive oxygen species and endoplasmic reticulum (ER) stress. These effects held true in vivo as well: prophylactic or therapeutic use of TG could obviously inhibit ER stress and decrease islet β-cell apoptosis in the pancreas of mice given a high-fat/high-sucrose diet (HFHSD), thus dramatically maintaining or restoring β-cell mass and islet size, respectively. Accordingly, both prophylactic and therapeutic use of TG improved HFHSD-impaired glucose metabolism in mice, as evidenced by ameliorating hyperglycemia and glucose intolerance. Taken together, TG, as an agent promoting PDX1 expression exhibits strong protective effects on islet β-cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Xinlei Yao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China; Research Center of Agriculture and Medicine gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China
| | - Kun Li
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Chen Liang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Zilong Zhou
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Jiao Wang
- Research Center of Agriculture and Medicine gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China
| | - Shuyue Wang
- Research Center of Agriculture and Medicine gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China
| | - Lei Liu
- Research Center of Agriculture and Medicine gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China
| | - Chun-Lei Yu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Zhen-Bo Song
- Research Center of Agriculture and Medicine gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China
| | - Yong-Li Bao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Li-Hua Zheng
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Ying Sun
- Research Center of Agriculture and Medicine gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China
| | - Guannan Wang
- Research Center of Agriculture and Medicine gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China
| | - Yanxin Huang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Jingwen Yi
- Research Center of Agriculture and Medicine gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China
| | - Luguo Sun
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China.
| | - Yuxin Li
- Research Center of Agriculture and Medicine gene Engineering of Ministry of Education, Northeast Normal University, Changchun, China.
| |
Collapse
|
7
|
Kooptiwut S, Samon K, Semprasert N, Suksri K, Yenchitsomanus PT. Prunetin Protects Against Dexamethasone-Induced Pancreatic Β-Cell Apoptosis via Modulation of p53 Signaling Pathway. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20916328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Long-term administration of dexamethasone results in insulin resistance and pancreatic β-cell apoptosis. Prunetin (an O-methylated isoflavone, a type of flavonoid) is demonstrated to protect diabetes, but the molecular mechanism of this protection is still unclear. This study thus aims to investigate how prunetin protects against dexamethasone-induced pancreatic β-cell apoptosis. Rat insulinoma (INS-1) cells were cultured in medium with or without dexamethasone in the presence or absence of prunetin or pifithrin-α, a p53 inhibitor. Cell apoptosis was measured by Annexin V/propidium iodide staining. Dexamethasone significantly induced INS-1 apoptosis but dexamethasone plus prunetin significantly reduced INS-1 apoptosis. Dexamethasone-treated INS-1 upregulated p53 protein expression; the induction of p53 was also reduced in the presence of RU486, a glucocorticoid receptor (GR) inhibitor. This suggested that dexamethasone induced P53 via GR. Dexamethasone-treated INS-1 significantly increased p53, Bax, and Rb protein expressions, whereas treatments of dexamethasone plus prunetin or pifithrin-α significantly decreased these protein expressions. In addition, dexamethasone significantly decreased B-cell lymphoma 2 (Bcl2), while dexamethasone plus prunetin or pifithrin-α significantly increased Bcl2. Dexamethasone significantly increased caspase-3 activity while co-treatment of dexamethasone plus prunetin or pifithrin-α significantly decreased caspase-3 activity to the control level. Taken together, our results revealed that prunetin protected against dexamethasone-induced pancreatic β-cells apoptosis via modulation of the p53 signaling pathway.
Collapse
Affiliation(s)
- Suwattanee Kooptiwut
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanokwan Samon
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Namoiy Semprasert
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanchana Suksri
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pa-Thai Yenchitsomanus
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
8
|
Blanco AM, Cortés R, Bertucci JI, Soletto L, Sánchez E, Valenciano AI, Cerdá-Reverter JM, Delgado MJ. Brain transcriptome profile after CRISPR-induced ghrelin mutations in zebrafish. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1-21. [PMID: 31673996 DOI: 10.1007/s10695-019-00687-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/17/2019] [Indexed: 06/10/2023]
Abstract
Ghrelin (GRL) is a gut-brain hormone with a role in a wide variety of physiological functions in mammals and fish, which points out the ghrelinergic system as a key element for the appropriate biological functioning of the organism. However, many aspects of the multifunctional nature of GRL remain to be better explored, especially in fish. In this study, we used the CRISPR/Cas9 genome editing technique to generate F0 zebrafish in which the expression of grl is compromised. Then, we employed high-throughput mRNA sequencing (RNA-seq) to explore changes in the brain transcriptome landscape associated with the silencing of grl. The CRISPR/Cas9 technique successfully edited the genome of F0 zebrafish resulting in individuals with considerably lower levels of GRL mRNAs and protein and ghrelin O-acyl transferase (goat) mRNAs in the brain, intestine, and liver compared to wild-type (WT) zebrafish. Analysis of brain transcriptome revealed a total of 1360 differentially expressed genes (DEGs) between the grl knockdown (KD) and WT zebrafish, with 664 up- and 696 downregulated DEGs in the KD group. Functional enrichment analysis revealed that DEGs are highly enriched for terms related to morphogenesis, metabolism (especially of lipids), entrainment of circadian clocks, oxygen transport, apoptosis, and response to stimulus. The present study offers valuable information on the central genes and pathways implicated in functions of GRL, and points out the possible involvement of this peptide in some novel functions in fish, such as apoptosis and oxygen transport.
Collapse
Affiliation(s)
- Ayelén Melisa Blanco
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040, Madrid, Spain
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro de Investigación Mariña, Universidade de Vigo, Vigo, Spain
| | - Raúl Cortés
- Departamento de Fisiología de Peces y Biotecnología, Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas, Ribera de Cabanes, 12595, Torre de la Sal, Castellón, Spain
- Centro de Investigación en Recursos Naturales y Sustentabilidad, Universidad Bernardo O'Higgins, Fábrica, 1990, Santiago, Chile
| | | | - Lucia Soletto
- Departamento de Fisiología de Peces y Biotecnología, Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas, Ribera de Cabanes, 12595, Torre de la Sal, Castellón, Spain
| | - Elisa Sánchez
- Departamento de Fisiología de Peces y Biotecnología, Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas, Ribera de Cabanes, 12595, Torre de la Sal, Castellón, Spain
| | - Ana Isabel Valenciano
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040, Madrid, Spain
| | - José Miguel Cerdá-Reverter
- Departamento de Fisiología de Peces y Biotecnología, Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas, Ribera de Cabanes, 12595, Torre de la Sal, Castellón, Spain.
| | - María Jesús Delgado
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, José Antonio Nováis 12, 28040, Madrid, Spain.
| |
Collapse
|
9
|
Zhang J, Xie T. Ghrelin inhibits cisplatin-induced MDA-MB-231 breast cancer cell apoptosis via PI3K/Akt/mTOR signaling. Exp Ther Med 2019; 19:1633-1640. [PMID: 32104214 PMCID: PMC7027091 DOI: 10.3892/etm.2019.8398] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 02/21/2019] [Indexed: 02/07/2023] Open
Abstract
Ghrelin is a multi-functional peptide, its role on cancer cell apoptosis remains controversial. The present study examined the effects and mechanisms of ghrelin on cisplatin-induced apoptosis in human breast cancer cells. It was identified that ghrelin inhibited apoptosis in MDA-MB-231 cells in vitro and reversed the expression of B-cell lymphoma 2 (Bcl2) and Bcl2-associated X, and cleaved caspase-3 induced by cisplatin. Furthermore, ghrelin activated the phosphoinositide 3-kinases/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway after cisplatin treatment. The effects of ghrelin on the cisplatin-induced apoptosis and PI3K/Akt/mTOR signaling were reversed by the growth hormone secretagogue receptor small interfering RNA. The present study suggests that ghrelin may serve as a novel target for cisplatin resistance and a potential indicator of cisplatin sensitivity in breast cancer treatment.
Collapse
Affiliation(s)
- Jinyu Zhang
- Department of Oncology, Medical College of Heibei University, Baoding, Hebei 071000, P.R. China
| | - Tianhao Xie
- Department of General Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| |
Collapse
|
10
|
Zhang H, Zhu C, Sun Z, Yan X, Wang H, Xu H, Ma J, Zhang Y. Linderane protects pancreatic β cells from streptozotocin (STZ)-induced oxidative damage. Life Sci 2019; 233:116732. [PMID: 31394125 DOI: 10.1016/j.lfs.2019.116732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/03/2019] [Accepted: 08/04/2019] [Indexed: 12/25/2022]
Abstract
AIMS Linderane, an important bioactive compound in Linderae, improved glucose and lipid metabolism in ob/ob mice. However, the effect of linderane on streptozotocin (STZ)-induced oxidative damage in INS-1 cells remains unclear. MAIN METHODS INS-1 cells were pre-treated with different doses of linderane for 2 h and then treated with 3 mM STZ for 12 h. Cell viability was determined by MTT assay. Cell apoptosis was detected using an Annexin V-FITC Apoptosis Detection Kit. The level of intracellular ROS was determined using dichlorofluorescein-diacetate (DCFH-DA). The activities of insulin secretion, SOD, catalase (CAT) and GPx were measured using ELISA kits. The expression levels of bax, bcl-2, p38, p-p38, nuclear Nrf2 and HO-1 were measured using western blot. KEY FINDINGS The results showed that STZ-caused inhibitory effects on cell viability and insulin secretion were mitigated by linderane. Furthermore, linderane inhibited apoptosis and oxidative stress in STZ-induced INS-1 cells. Finally, linderane suppressed the activation of p38 MAPK pathway, as well as enhanced the activation of Nrf2 pathway in STZ-induced INS-1 cells. Activation of p38 MAPK pathway or inhibition of Nrf2 significantly reversed the protective effects of linderane against STZ-induced ROS production and cell apoptosis. SIGNIFICANCE The protective effects of linderane on STZ-induced INS-1 cells might be attributed to the inhibition of p38 MAPK and activation of Nrf2 pathway.
Collapse
Affiliation(s)
- Haijun Zhang
- The Second Department of Endocrinology, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang Province, China; The Second Department of Endocrinology, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161005, Heilongjiang Province, China.
| | - Chunping Zhu
- Department of Cardiac Function, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang Province, China; Department of Cardiac Function, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161005, Heilongjiang Province, China
| | - Zhe Sun
- The Second Department of Endocrinology, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang Province, China; The Second Department of Endocrinology, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161005, Heilongjiang Province, China
| | - Xiaoguang Yan
- The Second Department of Endocrinology, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang Province, China; The Second Department of Endocrinology, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161005, Heilongjiang Province, China
| | - Huihui Wang
- The Second Department of Endocrinology, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang Province, China; The Second Department of Endocrinology, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161005, Heilongjiang Province, China
| | - Haibo Xu
- The Second Department of Endocrinology, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang Province, China; The Second Department of Endocrinology, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161005, Heilongjiang Province, China
| | - Jiani Ma
- The Second Department of Endocrinology, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang Province, China; The Second Department of Endocrinology, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161005, Heilongjiang Province, China
| | - Yanrong Zhang
- The Second Department of Endocrinology, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang Province, China; The Second Department of Endocrinology, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar 161005, Heilongjiang Province, China
| |
Collapse
|
11
|
Tan Y, Nie W, Chen C, He X, Xu Y, Ma X, Zhang J, Tan M, Rong P, Wang W. Mesenchymal stem cells alleviate hypoxia-induced oxidative stress and enhance the pro-survival pathways in porcine islets. Exp Biol Med (Maywood) 2019; 244:781-788. [PMID: 31042075 DOI: 10.1177/1535370219844472] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
IMPACT STATEMENT The utilization of mesenchymal stem cells (MSCs) is a promising approach to serve as adjuvant therapy for islet transplantation. But the inability to translate promising preclinical results into sound therapeutic effects in human subjects indicates a lack of key knowledge of MSC-islet interactions that warrant further research. Hypoxia and oxidative stress are critical factors which lead to a tremendous loss of islet grafts. However, previous studies mainly focused on other aspects of MSC protection such as inducing revascularization, enhancing insulin secretion, and reducing islet apoptosis. In this study, we aim to investigate whether MSC can protect islet cells from hypoxic damage by inhibiting ROS production and the potential underlying pathways involved. We also explore the effects of MSC-derived exosomes and IL-6 on hypoxia-injured islets. Our data provide new molecular targets for developing MSC applications, and this may ultimately promote the efficiency of clinical islet transplantation.
Collapse
Affiliation(s)
- Yixiong Tan
- 1 Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China.,2 Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Wei Nie
- 1 Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China.,2 Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China.,3 Engineering and Technology Research Center for Xenotransplantation of Hunan Province, Changsha 410000, China
| | - Cheng Chen
- 1 Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China.,2 Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Xuesong He
- 1 Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China.,2 Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Yuzhi Xu
- 1 Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China.,2 Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Xiaoqian Ma
- 1 Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China.,2 Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China.,3 Engineering and Technology Research Center for Xenotransplantation of Hunan Province, Changsha 410000, China
| | - Juan Zhang
- 1 Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China.,2 Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Mengqun Tan
- 3 Engineering and Technology Research Center for Xenotransplantation of Hunan Province, Changsha 410000, China
| | - Pengfei Rong
- 1 Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China.,2 Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Wei Wang
- 1 Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China.,2 Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China.,3 Engineering and Technology Research Center for Xenotransplantation of Hunan Province, Changsha 410000, China
| |
Collapse
|
12
|
Lv W, Zhang J, Jiao A, Wang B, Chen B, Lin J. Resveratrol attenuates hIAPP amyloid formation and restores the insulin secretion ability in hIAPP-INS1 cell line via enhancing autophagy. Can J Physiol Pharmacol 2019; 97:82-89. [PMID: 30312115 DOI: 10.1139/cjpp-2016-0686] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
It has been proved that human islet amyloid polypeptide (hIAPP), the main constituent of islet amyloid deposition, is one of the important factors that can induce type 2 diabetes or graft failure after islet transplantation. As there is no research on whether resveratrol degrading the amyloid deposition by its special chemical structure or enhancing autophagy had been published, we decided to detect the function of resveratrol in degrading the amyloid deposition in pancreatic beta cells. We established stable hIAPP-INS1 cell line via transfecting INS1 cells by lentivirus that overexpresses hIAPP. Our research demonstrates that amyloid deposition existed in hIAPP-INS1 cell by the thioflavin S fluorescent staining, meanwhile the function of insulin secretion of hIAPP-INS1 cells was decreased significantly (p < 0.01). After treatment with resveratrol (20 μM) for 24 h, amyloid deposition in hIAPP-INS1 cells was decreased significantly, and the insulin secretion was restored significantly (p < 0.01). Once inhibited the autophagy of hIAPP-INS1 cells by 3-methyladenine for 24 h, resveratrol does not effectively remove hIAPP deposits again, and cannot improve the function of insulin secretion. These results provide a novel thought that resveratrol can degrade the amyloid deposition in type 2 diabetes and the graft after islet transplantation.
Collapse
Affiliation(s)
- Wu Lv
- Department of Hepatobiliary and Transplantation Surgery, The First Hospital of China Medical University, Shenyang, China
- Department of General Surgery, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Jialin Zhang
- Department of Hepatobiliary and Transplantation Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Ao Jiao
- Department of Hepatobiliary and Transplantation Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Bowen Wang
- Department of Hepatobiliary and Transplantation Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Baomin Chen
- Department of Hepatobiliary and Transplantation Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Jie Lin
- Department of General Surgery, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
13
|
Shi Y, Lv W, Jiao A, Zhang C, Zhang J. A Novel Pentapeptide Inhibitor Reduces Amyloid Deposit Formation by Direct Interaction with hIAPP. Int J Endocrinol 2019; 2019:9062032. [PMID: 30838043 PMCID: PMC6374824 DOI: 10.1155/2019/9062032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/14/2018] [Accepted: 11/26/2018] [Indexed: 11/17/2022] Open
Abstract
BACKGROUNDS The presence of amyloid deposits of human islet amyloid polypeptide (hIAPP) in islet β-cells has been associated with type 2 diabetes occurrence and islet graft failure. Self-assembly into oligomers and fibrils during the process of aggregation by hIAPP can lead to failure and depletion of β-cells. Studies have shown that some critical regions of hIAPP might contribute to the aggregation. Thus, many studies focused on finding the effective molecules, especially the short-peptide inhibitors, that bind to these regions and disrupt the aggregation of hIAPP. In the present study, a novel pentapeptide inhibitor Phe-Leu-Pro-Asn-Phe (FLPNF) was designed and its effectiveness on the inhibition of the formation of amyloid deposits was examined. METHODS The binding mode between FLPNF and hIAPP was performed using molecular docking. The effectiveness of FLPNF on inhibiting hIAPP amyloid aggregation was tested by Thioflavin T (ThT) staining. Furthermore, negative stain electron microscopy was used to observe hIAPP fibrils. A biolayer interferometry analysis was used to identify the interaction between FLPNF and hIAPP. In addition, the cytotoxicity toward INS-1 cells was tested by a cell proliferation assay. RESULTS FLPNF was predicted to have a compact conformation to bind at the site of hIAPP. FLPNF strongly inhibited the amyloid aggregation of hIAPP at a 10 : 1 molar ratio in vitro. Coincubation of FLPNF with hIAPP decreased the amount of hIAPP fibrils. Furthermore, a direct interaction between FLPNF and hIAPP was confirmed. FLPNF could also decrease the cytotoxic effect of hIAPP. CONCLUSIONS The novel pentapeptide inhibitor FLPNF was constructed and inhibited the aggregation through direct binding to hIAPP. It is considered a suitable inhibitor for hIAPP amyloid deposit formation.
Collapse
Affiliation(s)
- Yue Shi
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, The First Hospital of China Medical University, Shenyang 110001, China
- Department of Geriatric Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Wu Lv
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, The First Hospital of China Medical University, Shenyang 110001, China
- Department of General Surgery (VIP Ward), Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang 110001, China
| | - Ao Jiao
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| | - Chengshuo Zhang
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jialin Zhang
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
14
|
Ma J, Wang X, Lv T, Liu J, Ren Y, Zhang J, Zhang Y. Effects of Ghrelin on the Apoptosis of Rheumatoid Arthritis Fibroblast-Like Synoviocyte MH7A Cells. Biol Pharm Bull 2018; 42:158-163. [PMID: 30429411 DOI: 10.1248/bpb.b18-00285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ghrelin is a circulating peptide hormone, which involved in promoting feeding and regulating energy metabolism in human and rodents. Abnormal synovial hyperplasia is the most important pathologic hallmark of rheumatoid arthritis (RA), which is characterised by tumor-like expansion. Existing studies indicated that there may exist some relation between the decreased ghrelin and the abnormally proliferating synovial cells in RA. Therefore, the aim of this study is to explore the apoptotic effects of ghrelin on MH7A synovial cells in vitro. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to evaluate the effects of ghrelin on the viability of MH7A cells. Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick-end labeling (TUNEL) and flow cytometry were used to test the apoptotic effects of ghrelin. At last, Western blot and real-time PCR were performed to explore the expression of caspases-8, -9, and -3 after the treatment of ghrelin. MTT experiments showed that ghrelin could inhibit viability of MH7A cells. The results of flow cytometry and TUNEL showed that ghrelin could induce apoptosis of MH7A synovial cells. Western blot showed that expression of cleaved-caspases-8, -9, and -3 were increased in ghrelin stimulation group compared with the control group, while expression of pro-caspases-8, -9, and -3 had no significant difference. In mRNA levels, ghrelin can decrease pro-caspases-8, -9, and -3 mRNA expression, which confirmed the results of protein levels. Then these apoptotic effects were significantly reversed by [D-Lys3] GHRP-6 (ghrelin receptor antagonist). This study found that ghrelin can induce apoptosis of MH7A cells through caspase signaling pathways.
Collapse
Affiliation(s)
- Junxian Ma
- Department of Rheumatology and Immunology, Tangdu Hospital of the Fourth Military Medical University (Air Force Medical University)
| | - Xinbo Wang
- Department of Rheumatology and Immunology, Tangdu Hospital of the Fourth Military Medical University (Air Force Medical University)
| | - Tingting Lv
- Department of Rheumatology and Immunology, Tangdu Hospital of the Fourth Military Medical University (Air Force Medical University)
| | - Jie Liu
- Department of Rheumatology and Immunology, Tangdu Hospital of the Fourth Military Medical University (Air Force Medical University)
| | - Ying Ren
- Department of Rheumatology and Immunology, Tangdu Hospital of the Fourth Military Medical University (Air Force Medical University)
| | - Jinshan Zhang
- Department of Human Anatomy and Histology and Embryology, the Fourth Military Medical University (Air Force Medical University)
| | - Yan Zhang
- Department of Rheumatology and Immunology, Tangdu Hospital of the Fourth Military Medical University (Air Force Medical University)
| |
Collapse
|
15
|
Abdanipour A, Dadkhah M, Alipour M, Feizi H. Effect of Ghrelin on Caspase 3 and Bcl2 Gene Expression in H2O2 Treated Rat's Bone Marrow Stromal Cells. Adv Pharm Bull 2018; 8:429-435. [PMID: 30276139 PMCID: PMC6156489 DOI: 10.15171/apb.2018.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/14/2018] [Accepted: 07/19/2018] [Indexed: 12/18/2022] Open
Abstract
Purpose: The antiapoptotic effect of ghrelin in various cell lines including bone marrow stromal cells (BMSCs) has been proved. However, the real mechanism of this effect is not clear. Caspase3 and Bcl2 are well-known pro- and antiapoptotic regulatory genes in eukaryotes. The aim of the study was to find out the effect of ghrelin on Caspase 3 and Bcl2 change in BMSCs. Methods: Rat BMSCs were cultivated in DMEM. Passage 3 BMSCs were treated with ghrelin 100 μM for 48 h. Real-time PCR for Caspase 3 and Bcl2 was carried out from B (untreated BMSCs), BH (BMSCs treated with 125 µM H2O2), BGH (BMSCs treated with 100 µM ghrelin then 125 µM H2O2) and BG (BMSCs treated with 100 µM ghrelin) groups. For immunofluorescence, cells were incubated with anti Caspase 3 and Bcl2monoclonal antibodies. Primary antibodies were visualized using the FITC method. All data are presented as means ± SEM. Values of P<0.05 were considered statistically significant. Results: Ghrelin decreased mRNA expressions of Caspase-3 significantly as compared to the BH group (P<0.05). Also, Bcl-2 gene expression showed an increment in BG group as compare with BH and BGH groups (P<0.05). A high present of Bcl-2 positive cells were observed in the BGH group while Caspase-3 positive cells were significantly decreased in the BGH group compared with the BH group (P<0.05). Conclusion: Ghrelin probably enhances BMSCs viability through regulation of pro- and antiapoptotic genes Caspase 3 and Bcl2. However the signaling pathway of this effect should be elucidated in the future.
Collapse
Affiliation(s)
- Alireza Abdanipour
- Department of Anatomical Sciences, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Masoud Dadkhah
- Department of Physiology and Pharmacology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohsen Alipour
- Department of Physiology and Pharmacology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hadi Feizi
- Department of Physiology and Pharmacology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
16
|
Jiao A, Li F, Zhang C, Lv W, Chen B, Zhang J. Simulated Cholinergic Reinnervation of β (INS-1) Cells: Antidiabetic Utility of Heterotypic Pseudoislets Containing β Cell and Cholinergic Cell. Int J Endocrinol 2018; 2018:1505307. [PMID: 29755519 PMCID: PMC5884158 DOI: 10.1155/2018/1505307] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/02/2018] [Accepted: 01/17/2018] [Indexed: 12/30/2022] Open
Abstract
Cholinergic neurons can functionally support pancreatic islets in controlling blood sugar levels. However, in islet transplantation, the level of cholinergic reinnervation is significantly lower compared to orthotopic pancreatic islets. This abnormal reinnervation affects the survival and function of islet grafts. In this study, the cholinergic reinnervation of beta cells was simulated by 2D and 3D coculture of INS-1 and NG108-15 cells. In 2D culture conditions, 20 mM glucose induced a 1.24-fold increase (p < 0.0001) in insulin secretion from the coculture group, while in the 3D culture condition, a 1.78-fold increase (p < 0.0001) in insulin secretion from heterotypic pseudoislet group was observed. Glucose-stimulated insulin secretion (GSIS) from 2D INS-1 cells showed minimal changes when compared to 3D structures. E-cadherin expressed in INS-1 and NG108-15 cells was the key adhesion molecule for the formation of heterotypic pseudoislets. NG108-15 cells hardly affected the proliferation of INS-1 cells in vitro. Heterotypic pseudoislet transplantation recipient mice reverted to normoglycemic levels faster and had a greater blood glucose clearance compared to INS-1 pseudoislet recipient mice. In conclusion, cholinergic cells can promote insulin-secreting cells to function better in vitro and in vivo and E-cadherin plays an important role in the formation of heterotypic pseudoislets.
Collapse
Affiliation(s)
- Ao Jiao
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| | - Feng Li
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| | - Chengshuo Zhang
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| | - Wu Lv
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| | - Baomin Chen
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jialin Zhang
- Hepatobiliary Surgery Department and Unit of Organ Transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
17
|
Chen X, Jiang S, Huang S. Maduramicin-activated protein phosphatase 2A results in extracellular signal-regulated kinase 1/2 inhibition, leading to cytotoxicity in myocardial H9c2 cells. Toxicol Lett 2017; 284:96-102. [PMID: 29241734 DOI: 10.1016/j.toxlet.2017.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/08/2017] [Accepted: 12/09/2017] [Indexed: 11/26/2022]
Abstract
Maduramicin, a polyether ionophore antibiotic used as an anticoccidial agent in poultry industry, has been reported to be toxic to animals and humans if improperly used or by accident, resulting in heart failure, skeletal muscle degeneration and even death. However, the molecular mechanism underlying its cardiotoxicity remains elusive. Mitogen activated protein kinases (MAPKs) and protein phosphatases signaling pathways have been documented to be involved in the cell survival regulation. The present study was set to investigate the role of above pathways in maduramicin-induced myocardial cytotoxicity. Here we observed that maduramicin inhibited cell proliferation and reduced cell viability in H9c2 cells. Furthermore, we found that maduramicin suppressed extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation in a concentration-dependent manner. Ectopic expression of constitutively active MKK1 partially prevented the cytotoxicity of maduramicin. Moreover, we showed that maduramicin concentration-dependently activated protein phosphatase 2A (PP2A) by decreasing its phosphorylation and increasing its methylation. Inhibition of PP2A with okadaic acid attenuated maduramicin's toxicity. Overexpression of dominant negative PP2A partially rescued cells from maduramicin-inhibited ERK1/2 contributing to its cytotoxicity. The results indicate that maduramicin activates PP2A and consequently inhibits ERK1/2, leading to cytotoxicity in H9c2 cells. Our data suggest that manipulation of PP2A-ERK1/2 pathway may be a potential approach to prevent maduramicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xin Chen
- Postdoctoral Mobile Station of Biology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu Province, 210023, PR China; Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, PR China.
| | - Shanxiang Jiang
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, PR China.
| | | |
Collapse
|
18
|
Billert M, Skrzypski M, Sassek M, Szczepankiewicz D, Wojciechowicz T, Mergler S, Strowski MZ, Nowak KW. TRPV4 regulates insulin mRNA expression and INS-1E cell death via ERK1/2 and NO-dependent mechanisms. Cell Signal 2017; 35:242-249. [PMID: 28359774 DOI: 10.1016/j.cellsig.2017.03.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/21/2017] [Accepted: 03/24/2017] [Indexed: 12/19/2022]
Abstract
TRPV4 is a Ca2+-permeable, nonselective cation channel. Recently, TRPV4 was implicated in controlling peripheral insulin sensitivity, insulin secretion and apoptosis of pancreatic beta cells. Here, we characterize the role and potential mechanisms of TRPV4 in regulating insulin mRNA expression and cell death in insulin producing INS-1E cells and rat pancreatic islets. TRPV4 protein production was downregulated by siRNA. Intracellular calcium level was measured using Fluo-3 AM. Gene expression was studied by real-time PCR. Phosphorylation of extracellular signal-regulated kinase (ERK1 and ERK2) was detected by Western blot. Nitric oxide (NO) production was assessed by chemiluminescent reaction. Reactive oxygen species (ROS) level was analysed using a fluorogenic dye (DCFDA). Cell death was evaluated by determination of cytoplasmic histone-associated DNA fragments. Downregulation of TRPV4 neither affected insulin mRNA expression nor INS-1E cell growth. By contrast, pharmacological TRPV4 activation by 100nmol/l GSK1016790A increased Ca2+ levels in INS-1E cells and enhanced insulin mRNA expression after 1 and 3h, whereas a suppression of insulin mRNA expression was detected after 24h incubation. GSK1016790A increased ERK1/2 phosphorylation and NO production but not ROS production. Pharmacological blockade of ERK1/2 attenuated GSK1016790A-induced insulin mRNA expression. Inhibition of NO synthesis by l-NAME failed to affect insulin mRNA expression in GSK1016790A treated INS-1E cells. Furthermore, inhibition of NO production attenuated GSK1016790A-induced INS-1E cell death. In pancreatic islets, 100nmol/l GSK1016790A increased insulin mRNA levels after 3h without inducing cytotoxicity after 24h. In conclusion, TRPV4 differently regulates insulin mRNA expression in INS-1E cells via ERK1/2 and NO-dependent mechanisms.
Collapse
Affiliation(s)
- M Billert
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60-637 Poznań, Poland
| | - M Skrzypski
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60-637 Poznań, Poland.
| | - M Sassek
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60-637 Poznań, Poland
| | - D Szczepankiewicz
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60-637 Poznań, Poland
| | - T Wojciechowicz
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60-637 Poznań, Poland
| | - S Mergler
- Department of Ophthalmology, Charité University Medicine Berlin, Germany
| | - M Z Strowski
- Department of Hepatology and Gastroenterology, Interdisciplinary Centre of Metabolism, Endocrinology, Diabetes and Metabolism, Charité-University Medicine Berlin, 13353 Berlin, Germany; Department of Internal Medicine-Gastroenterology, Park-Klinik Weissensee, 13086 Berlin, Germany
| | - K W Nowak
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60-637 Poznań, Poland
| |
Collapse
|
19
|
Vinayagam R, Xu B. 7, 8-Dihydroxycoumarin (daphnetin) protects INS-1 pancreatic β-cells against streptozotocin-induced apoptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 24:119-126. [PMID: 28160851 DOI: 10.1016/j.phymed.2016.11.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/14/2016] [Accepted: 11/30/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND AND OBJECTIVE Daphnetin (7, 8-dihydroxycoumarin), a natural coumarin compound, is known to exhibit antioxidant and anti-inflammatrory effects. However, the underlying mechanisms of its anti-apoptotic and antidiabetic effects yet not been examined. Therefore, the present work studied the anti-apoptotic and anti-diabetic effects of daphnetin by in vitro experiments. METHODS The rat insulinoma (INS-1) cells were pre-treated with daphnetin at different concentrations (1, 10, 20 and 40µM) for 24h followed by exposition to streptozotocin (STZ) (3mM) for 12h. Effects of daphnetin and STZ on INS-1 cells were determined by MTT assay, glucose stimulated insulin secretion (GSIS) assay, lipid peroxidation, antioxidant status (SOD, CAT, GPx, and GST) Apoptosis staining (DAPI, Hoechst 33342, AO/EB and ROS) was performed by fluorescence microscopy, and Bcl-2, Bax and NF-κB protein expression was detected by Western blotting. RESULTS MTT assay indicated that the viability of INS-1 cells was significantly reduced with exposure to STZ for 12h as compared to control cells, while pre-treated with daphnetin for 24h resulted in a significant improvement of cell viability. The effects daphnetin treatment in INS-1 cells on insulin secretion was tested and results showed that the pre-treatment of daphnetin could improve GSIS. Further, daphnetin pre-treatment significantly reduced the levels of lipid peroxidation markers and also improved antioxidant enzymes' activities in STZ-induced INS-1 cells. Western blotting assay revealed that daphnetin could suppress apoptosis through up-regulation of anti-apoptotic Bcl-2 protein expression and the down-regulation of pro-apoptotic Bax and nuclear factor NF-κB protein levels. CONCLUSION The results showed that daphnetin might be used in treating diabetes due to its insulin stimulating property and subsequent regulation of apoptotic pathway.
Collapse
Affiliation(s)
- Ramachandran Vinayagam
- Food Science and Technology Program, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai, Guangdong 519085, China
| | - Baojun Xu
- Food Science and Technology Program, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai, Guangdong 519085, China.
| |
Collapse
|
20
|
Zeb1 Is a Potential Regulator of Six2 in the Proliferation, Apoptosis and Migration of Metanephric Mesenchyme Cells. Int J Mol Sci 2016; 17:ijms17081283. [PMID: 27509493 PMCID: PMC5000680 DOI: 10.3390/ijms17081283] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 07/21/2016] [Accepted: 07/27/2016] [Indexed: 01/10/2023] Open
Abstract
Nephron progenitor cells surround around the ureteric bud tips (UB) and inductively interact with the UB to originate nephrons, the basic units of renal function. This process is determined by the internal balance between self-renewal and consumption of the nephron progenitor cells, which is depending on the complicated regulation networks. It has been reported that Zeb1 regulates the proliferation of mesenchymal cells in mouse embryos. However, the role of Zeb1 in nephrons generation is not clear, especially in metanephric mesenchyme (MM). Here, we detected cell proliferation, apoptosis and migration in MM cells by EdU assay, flow cytometry assay and wound healing assay, respectively. Meanwhile, Western and RT-PCR were used to measure the expression level of Zeb1 and Six2 in MM cells and developing kidney. Besides, the dual-luciferase assay was conducted to study the molecular relationship between Zeb1 and Six2. We found that knock-down of Zeb1 decreased cell proliferation, migration and promoted cell apoptosis in MM cells and Zeb1 overexpression leaded to the opposite data. Western-blot and RT-PCR results showed that knock-down of Zeb1 decreased the expression of Six2 in MM cells and Zeb1 overexpression contributed to the opposite results. Similarly, Zeb1 promoted Six2 promoter reporter activity in luciferase assays. However, double knock-down of Zeb1 and Six2 did not enhance the apoptosis of MM cells compared with control cells. Nevertheless, double silence of Zeb1 and Six2 repressed cell proliferation. In addition, we also found that Zeb1 and Six2 had an identical pattern in distinct developing phases of embryonic kidney. These results indicated that there may exist a complicated regulation network between Six2 and Zeb1. Together, we demonstrate Zeb1 promotes proliferation and apoptosis and inhibits the migration of MM cells, in association with Six2.
Collapse
|