1
|
Wei S, Sun J, Li Y, Xu K, Wang M, Zhang Y. Losartan Attenuates Atherosclerosis in Uremic Mice by Regulating Treg/Th17 Balance via Mediating PTEN/PI3K/Akt Pathway. Nephron Clin Pract 2022; 146:528-538. [PMID: 35176745 DOI: 10.1159/000521770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/29/2021] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Uremia could accelerate atherosclerosis (AS) formation involving Treg/Th17 imbalance. Losartan regulates the imbalance between regulatory T cells (Treg cells) and T helper 17 cells (Th17 cells). However, their interactions in uremia accelerated AS (UAAS) remained poorly understood. METHODS UAAS mice model was established, and after losartan and VO-OHpic (VO, phosphatase and tensin homolog [PTEN] inhibitor) injection, biological indexes, and inflammatory cytokines (transforming growth factor-β1, TGF-β1; interleukin-10 [IL-10]; IL-17 and IL-6) levels were determined using enzyme-linked immunosorbent assay. Pathological changes on aorta were observed using hematoxylin-eosin staining. Percentages of Treg cells (CD4+CD25+Foxp3+) and Th17 cells (CD4+IL-17+) in total CD4+ T cells were determined using flow cytometry. PTEN expressions were measured using Western blot, quantitative real-time polymerase chain reaction, and immunohistochemistry staining as needed. RESULTS After UAAS mice model construction, biological indexes (urea, cholesterol, and triglycerides) levels were increased, and aortic atherosclerotic plaque was formed. In UAAS mice, in total CD4+ T cells, Treg cells percentage was decreased yet Th17 cells percentage was increased, and TGF-β1 and IL-10 levels were downregulated yet IL-17 and IL-6 levels were upregulated. An opposite effect was found after losartan treatment. PTEN was downregulated in UAAS mice, and suppressing PTEN reversed the alleviating effects of losartan in UAAS mice. CONCLUSION Losartan attenuated UAAS in mice by regulating Treg/Th17 cells balance via mediating PTEN/PI3K/Akt pathway, providing possible therapeutic method for UAAS in clinical practice.
Collapse
Affiliation(s)
- Shanzhai Wei
- Department of Nephrology, Shuyang Hospital of TCM, Suqian, China
| | - Jie Sun
- Department of Nephrology, Shuyang Hospital of TCM, Suqian, China
| | - Yibei Li
- Department of Nephrology, Shuyang Hospital of TCM, Suqian, China
| | - Kangchun Xu
- Department of Nephrology, Shuyang Hospital of TCM, Suqian, China
| | - Man Wang
- Department of Nephrology, Shuyang Hospital of TCM, Suqian, China
| | - Yilai Zhang
- Department of Nephrology, Shuyang Hospital of TCM, Suqian, China
| |
Collapse
|
2
|
Zonghai C, Tao L, Pengjiao M, Liang G, Rongchuan Z, Xinyan W, Wenyi N, Wei L, Yi W, Lang B. Mycobacterium tuberculosis ESAT6 modulates host innate immunity by downregulating miR-222-3p target PTEN. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166292. [PMID: 34710568 DOI: 10.1016/j.bbadis.2021.166292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/18/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022]
Abstract
Tuberculosis (TB) remains a major cause of mortality and morbidity worldwide, and it is instant to discover novel anti-TB drugs due to the rapidly growing drug-resistance TB. Mycobacterium tuberculosis (Mtb) secreted effector ESAT6 plays a critical role in modulation miRNAs to regulate host defense mechanisms during Mtb infection, it can be a possible target for new tuberculosis drugs. The non-tuberculous mycobacteria Mycobacterium smegmatis (M. smegmatis) and Mtb have high gene homology but no pathogenicity. We used ESAT6 to interfere with macrophages or mice infected by M. smegmatis and determined that it enhanced the survival rate of bacteria and regulated miR-222-3p target PTEN. Expression of miR-222-3p reduced and PTEN enhanced with the progression of macrophages infected by M. smegmatis with ESAT6 co-incubation. MiR-222-3p overexpression diminished M. smegmatis survival and upregulated proinflammatory cytokines. VO-Ohpic trihydrate (PTEN inhibitor) reduced M. smegmatis survival and upregulated proinflammatory cytokines in vivo and in vitro, and VO-Ohpic trihydrate reversed the tissue damage of mouse organs caused by ESAT6. These results uncover an ESAT6 dependent role for miR-222-3p and its target PTEN in regulating host immune responses to bacterial infection and may provide a potential site for the development of anti-tuberculosis drugs that specifically antagonize the virulence of ESAT6.
Collapse
Affiliation(s)
- Chen Zonghai
- Laboratory of Infection and Immunity, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Luo Tao
- Laboratory of Infection and Immunity, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Ma Pengjiao
- Laboratory of Infection and Immunity, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Ge Liang
- Laboratory of Infection and Immunity, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Zhao Rongchuan
- Laboratory of Infection and Immunity, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Wang Xinyan
- Laboratory of Infection and Immunity, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Ni Wenyi
- Laboratory of Infection and Immunity, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Liao Wei
- Laboratory of Infection and Immunity, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Wang Yi
- Laboratory of Infection and Immunity, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Bao Lang
- Laboratory of Infection and Immunity, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Liang T, Gao F, Chen J. Role of PTEN-less in cardiac injury, hypertrophy and regeneration. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:25. [PMID: 34337686 PMCID: PMC8326232 DOI: 10.1186/s13619-021-00087-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/18/2021] [Indexed: 12/20/2022]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Cardiomyocytes are capable of coordinated contractions, which are mainly responsible for pumping blood. When cardiac stress occurs, cardiomyocytes undergo transition from physiological homeostasis to hypertrophic growth, proliferation, or apoptosis. During these processes, many cellular factors and signaling pathways participate. PTEN is a ubiquitous dual-specificity phosphatase and functions by dephosphorylating target proteins or lipids, such as PIP3, a second messenger in the PI3K/AKT signaling pathway. Downregulation of PTEN expression or inhibiting its biologic activity improves heart function, promotes cardiomyocytes proliferation, reduces cardiac fibrosis as well as dilation, and inhibits apoptosis following ischemic stress such as myocardial infarction. Inactivation of PTEN exhibits a potentially beneficial therapeutic effects against cardiac diseases. In this review, we summarize various strategies for PTEN inactivation and highlight the roles of PTEN-less in regulating cardiomyocytes during cardiac development and stress responses.
Collapse
Affiliation(s)
- Tian Liang
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Feng Gao
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Jinghai Chen
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China. .,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
4
|
Cardiac Mitochondrial PTEN-L determines cell fate between apoptosis and survival during chronic alcohol consumption. Apoptosis 2021; 25:590-604. [PMID: 32591959 DOI: 10.1007/s10495-020-01616-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic alcohol consumption induces myocardial damage and a type of non-ischemic cardiomyopathy termed alcoholic cardiomyopathy, where mitochondrial ultrastructural damages and suppressed fusion activity promote cardiomyocyte apoptosis. The aim of the present study is to determine the role of mitochondrial fission proteins and/or other proteins that localise on cardiac mitochondria for apoptosis upon ethanol consumption. In vivo and in vitro chronic alcohol exposure increased mitochondrial Drp1 levels but knockdown of the same did not confer cardioprotection in H9c2 cells. These cells displayed downregulated expression of MFN2 and OPA1 for Bak-mediated cytochrome c release and apoptosis. Dysregulated PTEN/AKT cell survival signal in both ethanol treated and Drp1 knockdown cells augmented oxidative stress by promoting mitochondrial PTEN-L and MFN1 interaction. Inhibiting this interaction with VO-OHpic, a reversible PTEN inhibitor, prevented Bak insertion into the mitochondria and release of cytochrome c to cytoplasm. Thus, our study provides evidence that Drp1-mediated mitochondrial fission is dispensable for ethanol-induced cardiotoxicity and that stress signals induce mitochondrial PTEN-L accumulation for structural and functional dyshomeostasis. Our in vivo results also demonstrates the therapeutic potential of VO-OHpic for habitual alcoholics developing myocardial dysfunction.
Collapse
|
5
|
Zhu X, Li J, Wang H, Gasior FM, Lee C, Lin S, Zhu Z, Wang Y, Justice CN, O'Donnell JM, Vanden Hoek TL. TAT delivery of a PTEN peptide inhibitor has direct cardioprotective effects and improves outcomes in rodent models of cardiac arrest. Am J Physiol Heart Circ Physiol 2021; 320:H2034-H2043. [PMID: 33834871 DOI: 10.1152/ajpheart.00513.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have recently shown that pharmacologic inhibition of PTEN significantly increases cardiac arrest survival in a mouse model, however, this protection required pretreatment 30 min before the arrest. To improve the onset of PTEN inhibition during cardiac arrest treatment, we have designed a TAT fused cell-permeable peptide (TAT-PTEN9c) based on the C-terminal PDZ binding motif of PTEN for rapid tissue delivery and protection. Western blot analysis demonstrated that TAT-PTEN9c peptide significantly enhanced Akt activation in mouse cardiomyocytes in a concentration- and time-dependent manner. Mice were subjected to 8 min asystolic arrest followed by CPR, and 30 mice with successful CPR were then randomly assigned to receive either saline or TAT-PTEN9c treatment. Survival was significantly increased in TAT-PTEN9c-treated mice compared with that of saline control at 4 h after CPR. The treated mice had increased Akt phosphorylation at 30 min resuscitation with significantly decreased sorbitol content in heart or brain tissues and reduced release of taurine and glutamate in blood, suggesting improved glucose metabolism. In an isolated rat heart Langendorff model, direct effects of TAT-PTEN9c on cardiac function were measured for 20 min following 20 min global ischemia. Rate pressure product was reduced by >20% for both TAT vehicle and nontreatment groups following arrest. Cardiac contractile function was completely recovered with TAT-PTEN9c treatment given at the start of reperfusion. We conclude that TAT-PTEN9c enhances Akt activation and decreases glucose shunting to the polyol pathway in critical organs, thereby preventing osmotic injury and early cardiovascular collapse and death.NEW & NOTEWORTHY We have designed a cell-permeable peptide, TAT-PTEN9c, to improve cardiac arrest survival. It blocked endogenous PTEN binding to its adaptor and enhanced Akt signaling in mouse cardiomyocytes. It improved mouse survival after cardiac arrest, which is related to improved glucose metabolism and reduced glucose shunting to sorbitol in critical organs.
Collapse
Affiliation(s)
- Xiangdong Zhu
- Program in Advanced Resuscitation Medicine, Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Jing Li
- Program in Advanced Resuscitation Medicine, Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Huashan Wang
- Program in Advanced Resuscitation Medicine, Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | | | - Chunpei Lee
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Shaoxia Lin
- Program in Advanced Resuscitation Medicine, Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Zhiyi Zhu
- Program in Advanced Resuscitation Medicine, Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Youhua Wang
- Program in Advanced Resuscitation Medicine, Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Cody N Justice
- Program in Advanced Resuscitation Medicine, Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois.,Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - J Michael O'Donnell
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | | |
Collapse
|
6
|
Johnson TA, Singla DK. PTEN inhibitor VO-OHpic attenuates inflammatory M1 macrophages and cardiac remodeling in doxorubicin-induced cardiomyopathy. Am J Physiol Heart Circ Physiol 2018; 315:H1236-H1249. [PMID: 30095997 PMCID: PMC6297808 DOI: 10.1152/ajpheart.00121.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 07/30/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023]
Abstract
Doxorubicin (Doxo) is an effective agent commonly used in cancer therapeutics. Unfortunately, Doxo treatment can stimulate cardiomyopathy and subsequent heart failure, limiting the use of this drug. The role of phosphatase and tensin homolog (PTEN) in apoptosis has been documented in Doxo-induced cardiomyopathy (DIC) and heart failure models. However, whether direct inhibition of PTEN attenuates apoptosis, cardiac remodeling, and inflammatory M1 macrophages in the DIC model remains elusive. Therefore, the present study was designed to understand the effects of VO-OHpic (VO), a potent inhibitor of PTEN, in reducing apoptosis and cardiac remodeling. At day 56, echocardiography was performed, which showed that VO treatment significantly ( P < 0.05) improved heart function. Immunohistochemistry, TUNEL, and histological staining were used to determine apoptosis, proinflammatory M1 macrophages, anti-inflammatory M2 macrophages, and cardiac remodeling. Our data show a significant increase in apoptosis, hypertrophy, fibrosis, and proinflammatory M1 macrophages with Doxo treatment, whereas VO treatment significantly reduced apoptosis, adverse cardiac remodeling, and proinflammatory M1 macrophages significantly ( P < 0.05) compared with the Doxo-treated group. Western blot analysis confirmed the reduction of phosphorylated PTEN and increase in phosphorylated AKT protein expression in the Doxo + VO-treated group. Moreover, VO administration increased anti-inflammatory M2 macrophages. Collectively, our data suggest that VO treatment attenuates apoptosis and adverse cardiac remodeling, a process that is mediated through the PTEN/AKT pathway, resulting in improved heart function in DIC. NEW & NOTEWORTHY Doxorubicin-induced cardiomyopathy (DIC) is still a major issue in patients with cancer. These novel findings on the phosphatase and tensin homolog inhibitor VO-OHpic in DIC is the first report, as per the best of our knowledge, that VO-OHpic significantly decreases apoptosis, fibrosis, hypertrophy, adverse cardiac remodeling, and proinflammatory M1 macrophages and increases anti-inflammatory M2 macrophages along with significantly improved cardiac function. VO-OHpic could be a future therapeutic agent for patients with DIC.
Collapse
Affiliation(s)
- Taylor A Johnson
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida , Orlando, Florida
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida , Orlando, Florida
| |
Collapse
|
7
|
Mitchell J, Kim SJ, Koukos G, Seelmann A, Veit B, Shepard B, Blumer-Schuette S, Winter HS, Iliopoulos D, Pothoulakis C, Im E, Rhee SH. Colonic Inhibition of Phosphatase and Tensin Homolog Increases Colitogenic Bacteria, Causing Development of Colitis in Il10-/- Mice. Inflamm Bowel Dis 2018; 24:1718-1732. [PMID: 29788382 PMCID: PMC6231371 DOI: 10.1093/ibd/izy124] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Indexed: 12/11/2022]
Abstract
Background Phosphatase and tensin homolog (Pten) is capable of mediating microbe-induced immune responses in the gut. Thus, Pten deficiency in the intestine accelerates colitis development in Il10-/- mice. As some ambient pollutants inhibit Pten function and exposure to ambient pollutants may increase inflammatory bowel disease (IBD) incidence, it is of interest to examine how Pten inhibition could affect colitis development in genetically susceptible hosts. Methods With human colonic mucosa biopsies from pediatric ulcerative colitis and non-IBD control subjects, we assessed the mRNA levels of the PTEN gene and the gene involved in IL10 responses. The data from the human tissues were corroborated by treating Il10-/-, Il10rb-/-, and wild-type C57BL/6 mice with Pten-specific inhibitor VO-OHpic. We evaluated the severity of mouse colitis by investigating the tissue histology and cytokine production. The gut microbiome was investigated by analyzing the 16S ribosomal RNA gene sequence with mouse fecal samples. Results PTEN and IL10RB mRNA levels were reduced in the human colonic mucosa of pediatric ulcerative colitis compared with non-IBD subjects. Intracolonic treatment of the Pten inhibitor induced colitis in Il10-/- mice, characterized by reduced body weight, marked colonic damage, and increased production of inflammatory cytokines, whereas Il10rb-/- and wild-type C57BL/6 mice treated with the inhibitor did not develop colitis. Pten inhibitor treatment changed the fecal microbiome, with increased abundance of colitogenic bacteria Bacteroides and Akkermansia in Il10-/- mice. Conclusions Loss of Pten function increases the levels of colitogenic bacteria in the gut, thereby inducing deleterious colitis in an Il10-deficient condition.
Collapse
Affiliation(s)
- Jonathon Mitchell
- Department of Biological Sciences, Oakland University, Rochester, Michigan
| | - Su Jin Kim
- College of Pharmacy, Pusan National University, Busan, Korea
| | - Georgios Koukos
- Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, California
| | - Alexandra Seelmann
- Department of Biological Sciences, Oakland University, Rochester, Michigan
| | - Brendan Veit
- Department of Biological Sciences, Oakland University, Rochester, Michigan
| | - Brooke Shepard
- Department of Biological Sciences, Oakland University, Rochester, Michigan
| | | | - Harland S Winter
- Pediatric IBD Center, Mass General Hospital for Children, Boston, Massachusetts
| | - Dimitrios Iliopoulos
- Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, California
| | - Charalabos Pothoulakis
- Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, California
| | - Eunok Im
- College of Pharmacy, Pusan National University, Busan, Korea
| | - Sang Hoon Rhee
- Department of Biological Sciences, Oakland University, Rochester, Michigan
| |
Collapse
|
8
|
Pulido R. PTEN Inhibition in Human Disease Therapy. Molecules 2018; 23:molecules23020285. [PMID: 29385737 PMCID: PMC6017825 DOI: 10.3390/molecules23020285] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/26/2018] [Accepted: 01/28/2018] [Indexed: 12/19/2022] Open
Abstract
The tumor suppressor PTEN is a major homeostatic regulator, by virtue of its lipid phosphatase activity against phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3], which downregulates the PI3K/AKT/mTOR prosurvival signaling, as well as by its protein phosphatase activity towards specific protein targets. PTEN catalytic activity is crucial to control cell growth under physiologic and pathologic situations, and it impacts not only in preventing tumor cell survival and proliferation, but also in restraining several cellular regeneration processes, such as those associated with nerve injury recovery, cardiac ischemia, or wound healing. In these conditions, inhibition of PTEN catalysis is being explored as a potentially beneficial therapeutic intervention. Here, an overview of human diseases and conditions in which PTEN inhibition could be beneficial is presented, together with an update on the current status of specific small molecule inhibitors of PTEN enzymatic activity, their use in experimental models, and their limitations as research or therapeutic drugs.
Collapse
Affiliation(s)
- Rafael Pulido
- Biomarkers in Cancer Unit, Biocruces Health Research Institute, 48903 Barakaldo, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain.
| |
Collapse
|
9
|
Augello G, Puleio R, Emma MR, Cusimano A, Loria GR, McCubrey JA, Montalto G, Cervello M. A PTEN inhibitor displays preclinical activity against hepatocarcinoma cells. Cell Cycle 2016; 15:573-83. [PMID: 26794644 DOI: 10.1080/15384101.2016.1138183] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Phosphatase and tensin homolog (PTEN) gene is considered a tumor suppressor gene. However, PTEN mutations rarely occur in hepatocellular carcinoma (HCC), whereas heterozygosity of PTEN, resulting in reduced PTEN expression, has been observed in 32-44% of HCC patients. In the present study, we investigated the effects of the small molecule PTEN inhibitor VO-OHpic in HCC cells. VO-OHpic inhibited cell viability, cell proliferation and colony formation, and induced senescence-associated β-galactosidase activity in Hep3B (low PTEN expression) and to a lesser extent in PLC/PRF/5 (high PTEN expression) cells, but not in PTEN-negative SNU475 cells. VO-OHpic synergistically inhibited cell viability when combined with PI3K/mTOR and RAF/MEK/ERK pathway inhibitors, but only in Hep3B cells, and significantly inhibited tumor growth in nude mice bearing xenografts of Hep3B cells. Therefore, we demonstrated for the first time that VO-OHpic inhibited cell growth and induced senescence in HCC cells with low PTEN expression, and that the combination of VO-OHpic with PI3K/mTOR and RAF/MEK/ERK inhibitors resulted in a more effective tumor cell kill. Our findings, hence, provide proof-of-principle evidence that pharmacological inhibition of PTEN may represent a promising approach for HCC therapy in a subclass of patients with a low PTEN expression.
Collapse
Affiliation(s)
- Giuseppa Augello
- a Institute of Biomedicine and Molecular Immunology "Alberto Monroy,", National Research Council (CNR) , Palermo , Italy
| | - Roberto Puleio
- b Istituto Zooprofilattico Sperimentale della Sicilia "A Mirri,", Area Diagnostica Specialistica, Laboratorio di Istopatologia ed Immunoistochimica , Palermo , Italy
| | - Maria Rita Emma
- a Institute of Biomedicine and Molecular Immunology "Alberto Monroy,", National Research Council (CNR) , Palermo , Italy
| | - Antonella Cusimano
- a Institute of Biomedicine and Molecular Immunology "Alberto Monroy,", National Research Council (CNR) , Palermo , Italy
| | - Guido R Loria
- b Istituto Zooprofilattico Sperimentale della Sicilia "A Mirri,", Area Diagnostica Specialistica, Laboratorio di Istopatologia ed Immunoistochimica , Palermo , Italy
| | - James A McCubrey
- c Department of Microbiology and Immunology , Brody School of Medicine at East Carolina University , Greenville , NC , USA
| | - Giuseppe Montalto
- a Institute of Biomedicine and Molecular Immunology "Alberto Monroy,", National Research Council (CNR) , Palermo , Italy.,d Biomedical Department of Internal Medicine and Specialties (DiBiMIS), University of Palermo , Palermo , Italy
| | - Melchiorre Cervello
- a Institute of Biomedicine and Molecular Immunology "Alberto Monroy,", National Research Council (CNR) , Palermo , Italy
| |
Collapse
|
10
|
McCubrey JA, Rakus D, Gizak A, Steelman LS, Abrams SL, Lertpiriyapong K, Fitzgerald TL, Yang LV, Montalto G, Cervello M, Libra M, Nicoletti F, Scalisi A, Torino F, Fenga C, Neri LM, Marmiroli S, Cocco L, Martelli AM. Effects of mutations in Wnt/β-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity-Diverse effects on cell growth, metabolism and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2942-2976. [PMID: 27612668 DOI: 10.1016/j.bbamcr.2016.09.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase that participates in an array of critical cellular processes. GSK-3 was first characterized as an enzyme that phosphorylated and inactivated glycogen synthase. However, subsequent studies have revealed that this moon-lighting protein is involved in numerous signaling pathways that regulate not only metabolism but also have roles in: apoptosis, cell cycle progression, cell renewal, differentiation, embryogenesis, migration, regulation of gene transcription, stem cell biology and survival. In this review, we will discuss the roles that GSK-3 plays in various diseases as well as how this pivotal kinase interacts with multiple signaling pathways such as: PI3K/PTEN/Akt/mTOR, Ras/Raf/MEK/ERK, Wnt/beta-catenin, hedgehog, Notch and TP53. Mutations that occur in these and other pathways can alter the effects that natural GSK-3 activity has on regulating these signaling circuits that can lead to cancer as well as other diseases. The novel roles that microRNAs play in regulation of the effects of GSK-3 will also be evaluated. Targeting GSK-3 and these other pathways may improve therapy and overcome therapeutic resistance.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA.
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Steve L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine at East Carolina University, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, USA
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Massimo Libra
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | | | - Aurora Scalisi
- Unit of Oncologic Diseases, ASP-Catania, Catania 95100, Italy
| | - Francesco Torino
- Department of Systems Medicine, Chair of Medical Oncology, Tor Vergata University of Rome, Rome, Italy
| | - Concettina Fenga
- Department of Biomedical, Odontoiatric, Morphological and Functional Images, Occupational Medicine Section - Policlinico "G. Martino" - University of Messina, Messina 98125, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Sandra Marmiroli
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| |
Collapse
|
11
|
Chen Q, Yue F, Li W, Zou J, Xu T, Huang C, Zhang Y, Song K, Huang G, Xu G, Huang H, Li J, Liu L. Potassium Bisperoxo(1,10-phenanthroline)oxovanadate (bpV(phen)) Induces Apoptosis and Pyroptosis and Disrupts the P62-HDAC6 Protein Interaction to Suppress the Acetylated Microtubule-dependent Degradation of Autophagosomes. J Biol Chem 2015; 290:26051-8. [PMID: 26363065 DOI: 10.1074/jbc.m115.653568] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a cellular process that controls and executes the turnover of dysfunctional organelles and misfolded or abnormally aggregated proteins. Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) activates the initiation of autophagy. Autophagosomes migrate along acetylated microtubules to fuse with lysosomes to execute the degradation of the engulfed substrates that usually bind with sequestosome 1 (SQSTM1, p62). Microtubule-associated protein 1 light chain 3 (LC3) traces the autophagy process by converting from the LC3-I to the LC3-II isoform and serves as a major marker of autophagy flux. Potassium bisperoxo(1,10-phenanthroline)oxovanadate (bpV(phen)) is an insulin mimic and a PTEN inhibitor and has the potential to treat different diseases. Here we show that bpV(phen) enhances the ubiquitination of p62, reduces the stability of p62, disrupts the interaction between p62 and histone deacetylase 6 (HDAC6), activates the deacetylase activity of HDAC6 on α-tubulin, and impairs stable acetylated microtubules. Microtubular destabilization leads to the blockade of autophagosome-lysosome fusion and accumulation of autophagosomes. Autophagy defects lead to oxidative stress and lysosomal rupture, which trigger different types of cell death, including apoptosis and pyroptosis. The consistent results from multiple systems, including mouse and different types of mammalian cells, are different from the predicted function of bpV(phen) as a PTEN inhibitor to activate autophagy flux. In addition, levels of p62 are reduced but not elevated when autophagosomal degradation is blocked, revealing a novel function of p62 in autophagy regulation. Therefore, it is necessary to pay attention to the roles of bpV(phen) in autophagy, apoptosis, and pyroptosis when it is developed as a drug.
Collapse
Affiliation(s)
- Qi Chen
- From the School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province 230032, China, the Department of Anesthesiology, Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui Province, 230601, China, the Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and
| | - Fei Yue
- the Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and
| | - Wenjiao Li
- the Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and
| | - Jing Zou
- the Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and
| | - Tao Xu
- From the School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province 230032, China
| | - Cheng Huang
- From the School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province 230032, China
| | - Ye Zhang
- the Department of Anesthesiology, Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui Province, 230601, China
| | - Kun Song
- the Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and
| | - Guanqun Huang
- the Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and
| | - Guibin Xu
- the Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and
| | - Hai Huang
- the Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and
| | - Jun Li
- From the School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province 230032, China,
| | - Leyuan Liu
- the Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and the Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas 77843
| |
Collapse
|
12
|
Targeting PTEN using small molecule inhibitors. Methods 2015; 77-78:63-8. [DOI: 10.1016/j.ymeth.2015.02.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 02/15/2015] [Accepted: 02/16/2015] [Indexed: 12/22/2022] Open
|
13
|
Li J, Wang H, Zhong Q, Zhu X, Chen SJ, Qian Y, Costakis J, Bunney G, Beiser DG, Leff AR, Lewandowski ED, ÓDonnell JM, Vanden Hoek TL. A novel pharmacological strategy by PTEN inhibition for improving metabolic resuscitation and survival after mouse cardiac arrest. Am J Physiol Heart Circ Physiol 2015; 308:H1414-22. [PMID: 25795713 DOI: 10.1152/ajpheart.00748.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 03/17/2015] [Indexed: 01/04/2023]
Abstract
Sudden cardiac arrest (SCA) is a leading cause of death in the United States. Despite return of spontaneous circulation, patients die due to post-SCA syndrome that includes myocardial dysfunction, brain injury, impaired metabolism, and inflammation. No medications improve SCA survival. Our prior work suggests that optimal Akt activation is critical for cooling protection and SCA recovery. Here, we investigate a small inhibitor of PTEN, an Akt-related phosphatase present in heart and brain, as a potential therapy in improving cardiac and neurological recovery after SCA. Anesthetized adult female wild-type C57BL/6 mice were randomized to pretreatment of VO-OHpic (VO) 30 min before SCA or vehicle control. Mice underwent 8 min of KCl-induced asystolic arrest followed by CPR. Resuscitated animals were hemodynamically monitored for 2 h and observed for 72 h. Outcomes included heart pressure-volume loops, energetics (phosphocreatine and ATP from (31)P NMR), protein phosphorylation of Akt, GSK3β, pyruvate dehydrogenase (PDH) and phospholamban, circulating inflammatory cytokines, plasma lactate, and glucose as measures of systemic metabolic recovery. VO reduced deterioration of left ventricular maximum pressure, maximum rate of change in the left ventricular pressure, and Petco2 and improved 72 h neurological intact survival (50% vs. 10%; P < 0.05). It reduced plasma lactate, glucose, IL-1β, and Pre-B cell colony enhancing factor, while increasing IL-10. VO increased phosphorylation of Akt and GSK3β in both heart and brain, and cardiac phospholamban phosphorylation while reducing p-PDH. Moreover, VO improved cardiac bioenergetic recovery. We concluded that pharmacologic PTEN inhibition enhances Akt activation, improving metabolic, cardiovascular, and neurologic recovery with increased survival after SCA. PTEN inhibitors may be a novel pharmacologic strategy for treating SCA.
Collapse
Affiliation(s)
- Jing Li
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Huashan Wang
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Qiang Zhong
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois; Department of Emergency Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science & Technology, China
| | - Xiangdong Zhu
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Sy-Jou Chen
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois; Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taiwan
| | - Yuanyu Qian
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois; Emergency Department, Chinese PLA General Hospital, Beijing, China
| | - Jim Costakis
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Gabrielle Bunney
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - David G Beiser
- Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Alan R Leff
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - E Douglas Lewandowski
- Program in Integrative Cardiac Metabolism, Center for Cardiovascular Research, and Department of Physiology and Biophysics, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - J Michael ÓDonnell
- Program in Integrative Cardiac Metabolism, Center for Cardiovascular Research, and Department of Physiology and Biophysics, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Terry L Vanden Hoek
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois;
| |
Collapse
|
14
|
Chemical intervention tools to probe phosphoinositide-dependent signalling. Biochem Soc Trans 2014; 42:1343-8. [DOI: 10.1042/bst20140186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Chemical intervention tools have been beneficial to many investigations elucidating signalling networks and interactions. The present review summarizes the current status of chemical tools to probe phosphoinositide metabolism and signalling. In particular, phosphoinositide-targeting tools are compared with protein-targeting tools with respect to their unique advantages and possible applications.
Collapse
|
15
|
Zhu X, Shao ZH, Li C, Li J, Zhong Q, Learoyd J, Meliton A, Meliton L, Leff AR, Vanden Hoek TL. TAT-protein blockade during ischemia/reperfusion reveals critical role for p85 PI3K-PTEN interaction in cardiomyocyte injury. PLoS One 2014; 9:e95622. [PMID: 24752319 PMCID: PMC3994094 DOI: 10.1371/journal.pone.0095622] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/27/2014] [Indexed: 11/19/2022] Open
Abstract
Recent work shows that cooling protection after mouse cardiac arrest and cardiomyocyte ischemia is mediated by Akt activation. The PI3K p85 subunit can either augment or inhibit Akt activation depending on its binding to p110 or PTEN respectively. To further clarify the role of PI3K p85 in cardioprotection, we studied novel TAT-p85 fusion proteins that selectively inhibit PI3K p85 binding. We hypothesized that TAT fused p85 lacking the PTEN binding site (TAT-ΔPTEN p85) would enhance Akt phosphorylation to afford cardioprotection. Conversely, TAT fused p85 lacking the p110 binding site (TAT-Δp110p85) would decrease Akt phosphorylation and abrogate cardioprotection. Microscopy and Western blot analysis demonstrated that TAT fusion protein was transduced into cardiomyocytes within 5 min and remained more than 2 h. Inhibition of PI3K/Akt by TAT-Δp110 p85 significantly increased cell death from 44.6±2.7% to 92.5±3.4% after simulated ischemia and reperfusion. By contrast, PTEN inhibition using TAT-ΔPTEN p85 decreased cell death to 11.9±5.3%, a similar level of cardioprotection seen with past cooling studies. Additional studies with the small molecule PTEN inhibitor VO-OHpic confirmed that PTEN inhibition was highly protective against cell death induced by ischemia and reperfusion. We conclude that blockade of p85-PTEN interaction and PTEN inhibition may be promising strategies for rescuing the heart from ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Xiangdong Zhu
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Zuo-Hui Shao
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Changqing Li
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Jing Li
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Qiang Zhong
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Jonathan Learoyd
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Angelo Meliton
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Lucille Meliton
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Alan R. Leff
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Terry L. Vanden Hoek
- Program in Advanced Resuscitation Medicine, Center for Cardiovascular Research, and Department of Emergency Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
16
|
Cheng KC, Asakawa A, Li YX, Chung HH, Amitani H, Ueki T, Cheng JT, Inui A. Silymarin induces insulin resistance through an increase of phosphatase and tensin homolog in Wistar rats. PLoS One 2014; 9:e84550. [PMID: 24404172 PMCID: PMC3880317 DOI: 10.1371/journal.pone.0084550] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 11/15/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND AIMS Phosphatase and tensin homolog (PTEN) is a phosphoinositide phosphatase that regulates crucial cellular functions, including insulin signaling, lipid and glucose metabolism, as well as survival and apoptosis. Silymarin is the active ingredient in milk thistle and exerts numerous effects through the activation of PTEN. However, the effect of silymarin on the development of insulin resistance remains unknown. METHODS Wistar rats fed fructose-rich chow or normal chow were administered oral silymarin to identify the development of insulin resistance using the homeostasis model assessment of insulin resistance and hyperinsulinemic- euglycemic clamping. Changes in PTEN expression in skeletal muscle and liver were compared using western blotting analysis. Further investigation was performed in L6 cells to check the expression of PTEN and insulin-related signals. PTEN deletion in L6 cells was achieved by small interfering ribonucleic acid transfection. RESULTS Oral administration of silymarin at a dose of 200 mg/kg once daily induced insulin resistance in normal rats and enhanced insulin resistance in fructose-rich chow-fed rats. An increase of PTEN expression was observed in the skeletal muscle and liver of rats with insulin resistance. A decrease in the phosphorylation of Akt in L6 myotube cells, which was maintained in a high-glucose condition, was also observed. Treatment with silymarin aggravated high-glucose-induced insulin resistance. Deletion of PTEN in L6 cells reversed silymarin-induced impaired insulin signaling and glucose uptake. CONCLUSIONS Silymarin has the ability to disrupt insulin signaling through increased PTEN expression. Therefore, silymarin should be used carefully in type-2 diabetic patients.
Collapse
Affiliation(s)
- Kai-Chun Cheng
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Akihiro Asakawa
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Ying-Xiao Li
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hsien-Hui Chung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Haruka Amitani
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takatoshi Ueki
- Department of Neuroanatomy, Hamamatsu University School of Medicine. Hamamatsu, Japan
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City, Taiwan
| | - Akio Inui
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- * E-mail:
| |
Collapse
|
17
|
Ding J, Guo J, Yuan Q, Yuan F, Chen H, Tian H. Inhibition of phosphatase and tensin homolog deleted on chromosome 10 decreases rat cortical neuron injury and blood-brain barrier permeability, and improves neurological functional recovery in traumatic brain injury model. PLoS One 2013; 8:e80429. [PMID: 24312220 PMCID: PMC3842922 DOI: 10.1371/journal.pone.0080429] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 10/02/2013] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Recent evidence has supported the neuroprotective effect of bpV (pic), an inhibitor of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), in models of ischemic stroke. However, whether PTEN inhibitors improve long-term functional recovery after traumatic brain injury (TBI) and whether PTEN affects blood brain barrier (BBB) permeability need further elucidation. The present study was performed to address these issues. METHODS Adult Sprague-Dawley rats were subjected to fluid percussion injury (FPI) after treatment with a well-established PTEN inhibitor bpV (pic) or saline starting 24 h before FPI. Western blotting, real-time quantitative PCR, or immunostaining was used to measure PTEN, p-Akt, or MMP-9 expression. We determined the presence of neuron apoptosis by TUNEL assay. Evans Blue dye extravasation was measured to evaluate the extent of BBB disruption. Functional recovery was assessed by the neurological severity score (NSS), and Kaplan-Meier analysis was used for survival analysis. RESULTS PTEN expression was up-regulated after TBI. After bpV (pic) treatment, p-Akt was also up-regulated. We found that bpV (pic) significantly decreased BBB permeability and reduced the number of TUNEL-positive cells. We further demonstrated that PTEN inhibition improved neurological function recovery in the early stage after TBI. CONCLUSION These data suggest that treatment with the PTEN inhibitor bpV (pic) has a neuroprotective effect in TBI rats.
Collapse
Affiliation(s)
- Jun Ding
- Department of Neurosurgery, Shanghai 6 th People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
18
|
Lai CTM, Ng EKO, Chow PC, Kwong A, Cheung YF. Circulating microRNA expression profile and systemic right ventricular function in adults after atrial switch operation for complete transposition of the great arteries. BMC Cardiovasc Disord 2013; 13:73. [PMID: 24040857 PMCID: PMC3847493 DOI: 10.1186/1471-2261-13-73] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 09/13/2013] [Indexed: 11/16/2022] Open
Abstract
Background Data on the use of circulating microRNAs (miRNAs) as biomarkers of cardiovascular diseases are emerging. Little, however, is known on the expression profile of circulating of microRNAs in congenital heart malformations with a systemic right ventricle that is prone to functional impairment. We aimed to test the hypothesis that circulating miRNA profile is altered in patients late after atrial switch operation for complete transposition of the great arteries (TGA) and further explored possible relationships between alteration of circulating miRNAs and systemic ventricular contractility. Methods Circulating miRNA expression profiling of serum samples from 5 patients and 5 healthy controls was performed. The results were validated in 26 patients and 20 controls using real-time quantitative reverse-transcription polymerase chain reaction for candidate miRNAs with fold changes >3 by expression profiling. Systemic ventricular myocardial acceleration during isovolumic contraction (IVA) was determined by colour tissue Doppler echocardiography. Results Compared with controls, patients had significantly lower systemic ventricular IVA (p = 0.002). Of the 23 upregulated miRNAs identified by profiling, 11 were validated to be increased in patients compared with controls: miR-16, miR-106a, miR-144*, miR-18a, miR-25, miR-451, miR-486-3p, miR-486-5p, miR-505*, let-7e and miR-93. Among the validated 11 miRNAs, miR-18a (r = −0.45, p = 0.002) and miR-486-5p (r = −0.35, p = 0.018) correlated negatively with systemic ventricular IVA for the whole cohort. Conclusions A distinct serum miRNA expression signature exists in adults with complete TGA after atrial switch operation, with serum miR-18a and miR-486-5p being associated with systemic ventricular contractility.
Collapse
Affiliation(s)
- Clare T M Lai
- Division of Paediatric Cardiology, Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hongkong, China.
| | | | | | | | | |
Collapse
|
19
|
Andreux PA, Williams EG, Koutnikova H, Houtkooper RH, Champy MF, Henry H, Schoonjans K, Williams RW, Auwerx J. Systems genetics of metabolism: the use of the BXD murine reference panel for multiscalar integration of traits. Cell 2012; 150:1287-99. [PMID: 22939713 DOI: 10.1016/j.cell.2012.08.012] [Citation(s) in RCA: 176] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 06/06/2012] [Accepted: 08/03/2012] [Indexed: 01/22/2023]
Abstract
Metabolic homeostasis is achieved by complex molecular and cellular networks that differ significantly among individuals and are difficult to model with genetically engineered lines of mice optimized to study single gene function. Here, we systematically acquired metabolic phenotypes by using the EUMODIC EMPReSS protocols across a large panel of isogenic but diverse strains of mice (BXD type) to study the genetic control of metabolism. We generated and analyzed 140 classical phenotypes and deposited these in an open-access web service for systems genetics (www.genenetwork.org). Heritability, influence of sex, and genetic modifiers of traits were examined singly and jointly by using quantitative-trait locus (QTL) and expression QTL-mapping methods. Traits and networks were linked to loci encompassing both known variants and novel candidate genes, including alkaline phosphatase (ALPL), here linked to hypophosphatasia. The assembled and curated phenotypes provide key resources and exemplars that can be used to dissect complex metabolic traits and disorders.
Collapse
Affiliation(s)
- Pénélope A Andreux
- Laboratory of Integrative and Systems Physiology, School of Life Sciences, École Polytechnique Fédérale de Lausanne 1015, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Shi GD, OuYang YP, Shi JG, Liu Y, Yuan W, Jia LS. PTEN deletion prevents ischemic brain injury by activating the mTOR signaling pathway. Biochem Biophys Res Commun 2010; 404:941-5. [PMID: 21185267 DOI: 10.1016/j.bbrc.2010.12.085] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 12/18/2010] [Indexed: 11/19/2022]
Abstract
It is increasingly clear that the tumor suppressor PTEN (phosphatase and tensin homolog deleted on chromosome 10) is a negative regulator of neuronal cell survival. However, its molecular mechanisms remain poorly understood. Here we found that PTEN/mTOR is critical for controlling neuronal cell death after ischemic brain injury. Male rats were subjected to MCAO (middle cerebral artery occlusion) followed by pretreating with bpv (pic), a potent inhibitor for PTEN, or by intra-cerebroventricular infusion of PTEN siRNA. bpv (pic) significantly decreased infarct volume and reduced the number of TUNEL-positive cells. We further demonstrated that although bpv (pic) did not affect brain injury-induced mTOR protein expression, bpv (pic) prevented decrease in phosphorylation of mTOR, and the subsequent decrease in S6. Similarly, down-regulation of PTEN expression also reduced the number of TUNEL-positive cells, and increased phospho-mTOR. These data suggest that PTEN deletion prevents neuronal cell death resulting from ischemic brain injury and that its neuroprotective effects are mediated by increasing the injury-induced mTOR phosphorylation.
Collapse
Affiliation(s)
- G D Shi
- Department of Orthopaedics Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, PR China
| | | | | | | | | | | |
Collapse
|