1
|
Roscovitine Protects From Arterial Injury by Regulating the Expressions of c-Jun and p27 and Inhibiting Vascular Smooth Muscle Cell Proliferation. J Cardiovasc Pharmacol 2017; 69:161-169. [PMID: 28009720 DOI: 10.1097/fjc.0000000000000453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
2
|
Tudzarova S, Mulholland P, Dey A, Stoeber K, Okorokov AL, Williams GH. p53 controls CDC7 levels to reinforce G1 cell cycle arrest upon genotoxic stress. Cell Cycle 2016; 15:2958-2972. [PMID: 27611229 PMCID: PMC5105930 DOI: 10.1080/15384101.2016.1231281] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
DNA replication initiation is a key event in the cell cycle, which is dependent on 2 kinases - CDK2 and CDC7. Here we report a novel mechanism in which p53 induces G1 checkpoint and cell cycle arrest by downregulating CDC7 kinase in response to genotoxic stress. We demonstrate that p53 controls CDC7 stability post-transcriptionally via miR-192/215 and post-translationally via Fbxw7β E3 ubiquitin ligase. The p53-dependent pathway of CDC7 downregulation is interlinked with the p53-p21-CDK2 pathway, as p21-mediated inhibition of CDK2-dependent phosphorylation of CDC7 on Thr376 is required for GSK3ß-phosphorylation and Fbxw7ß-dependent degradation of CDC7. Notably, sustained oncogenic high levels of active CDC7 exert a negative feedback onto p53, leading to unrestrained S-phase progression and accumulation of DNA damage. Thus, p53-dependent control of CDC7 levels is essential for blocking G1/S cell-cycle transition upon genotoxic stress, thereby safeguarding the genome from instability and thus representing a novel general stress response.
Collapse
Affiliation(s)
- Slavica Tudzarova
- a Wolfson Institute for Biomedical Research, Division of Medicine, University College London , London , UK.,b Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles , Los Angeles , CA , USA
| | - Paul Mulholland
- c Department of Pathology , UCL Cancer Institute, University College London , London , UK
| | - Ayona Dey
- a Wolfson Institute for Biomedical Research, Division of Medicine, University College London , London , UK
| | - Kai Stoeber
- c Department of Pathology , UCL Cancer Institute, University College London , London , UK
| | - Andrei L Okorokov
- a Wolfson Institute for Biomedical Research, Division of Medicine, University College London , London , UK
| | - Gareth H Williams
- c Department of Pathology , UCL Cancer Institute, University College London , London , UK.,d Oncologica Ltd, The Science Village, Chesterford Research Park , Cambridge , UK
| |
Collapse
|
3
|
Binukumar BK, Shukla V, Amin ND, Bhaskar M, Skuntz S, Steiner J, Winkler D, Pelech SL, Pant HC. Analysis of the Inhibitory Elements in the p5 Peptide Fragment of the CDK5 Activator, p35, CDKR1 Protein. J Alzheimers Dis 2016; 48:1009-17. [PMID: 26444778 PMCID: PMC4927891 DOI: 10.3233/jad-150412] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Besides the hallmark pathology of amyloid plaques and neurofibrillary tangles, it is well documented that cyclin-dependent kinase 5 (CDK5), a critical neuronal protein kinase in nervous system development, function, and survival, when deregulated and hyperactivated induces Alzheimer's disease (AD) and amyotrophic lateral sclerosis and Parkinson's disease-like phenotypes in mice. In a recent study, we demonstrated that p5, a small, truncated fragment of 24 amino acid residues derived from the CDK5 activator protein 35 (NCK5A, p35), selectively inhibited deregulated CDK5 hyperactivity and ameliorated AD phenotypes in model mice. In this study, we identified the most inhibitory elements in the p5 peptide fragment. Each amino acid residue in p5 was systematically replaced with its homologous residues that may still be able to functionally substitute. The effects of these p5 peptide analogs were studied on the phosphotransferase activities of CDK5/p35, CDK5/p25, ERK1, and GSK3β. The mimetic p5 peptide (A/V substitution at the C-terminus of the peptide) in the sequence, KNAFYERALSIINLMTSKMVQINV (p5-MT) was the most effective inhibitor of CDK5 kinase activity of 79 tested mimetic peptides including the original p5 peptide, KEAFWDRCLSVINLMSSKMLQINA (p5-WT). Replacement of the residues in C-terminus end of the peptide affected CDK5 phosphotransferase activity most significantly. These peptides were strong inhibitors of CDK5, but not the related proline-directed kinases, ERK1 and GSK3β.
Collapse
Affiliation(s)
- B K Binukumar
- National Institute of Neurological Disorders and Strokes, National Institutes of Health, Bethesda, MD, USA
| | - Varsha Shukla
- National Institute of Neurological Disorders and Strokes, National Institutes of Health, Bethesda, MD, USA
| | - Niranjana D Amin
- National Institute of Neurological Disorders and Strokes, National Institutes of Health, Bethesda, MD, USA
| | - Manju Bhaskar
- National Institute of Neurological Disorders and Strokes, National Institutes of Health, Bethesda, MD, USA
| | - Suzanne Skuntz
- National Institute of Neurological Disorders and Strokes, National Institutes of Health, Bethesda, MD, USA
| | - Joseph Steiner
- National Institute of Neurological Disorders and Strokes, National Institutes of Health, Bethesda, MD, USA
| | - Dirk Winkler
- Kinexus Bioinformatics Corporation, and Division of Neurology, Department of Medicine, University of British Columbia, BC, Canada
| | - Steven L Pelech
- Kinexus Bioinformatics Corporation, and Division of Neurology, Department of Medicine, University of British Columbia, BC, Canada
| | - Harish C Pant
- National Institute of Neurological Disorders and Strokes, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Priglinger CS, Obermann J, Szober CM, Merl-Pham J, Ohmayer U, Behler J, Gruhn F, Kreutzer TC, Wertheimer C, Geerlof A, Priglinger SG, Hauck SM. Epithelial-to-Mesenchymal Transition of RPE Cells In Vitro Confers Increased β1,6-N-Glycosylation and Increased Susceptibility to Galectin-3 Binding. PLoS One 2016; 11:e0146887. [PMID: 26760037 PMCID: PMC4712018 DOI: 10.1371/journal.pone.0146887] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 12/24/2015] [Indexed: 12/03/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) of retinal pigment epithelial cells is a crucial event in the onset of proliferative vitreoretinopathy (PVR), the most common reason for treatment failure in retinal detachment surgery. We studied alterations in the cell surface glycan expression profile upon EMT of RPE cells and focused on its relevance for the interaction with galectin-3 (Gal-3), a carbohydrate binding protein, which can inhibit attachment and spreading of human RPE cells in a dose- and carbohydrate-dependent manner, and thus bares the potential to counteract PVR-associated cellular events. Lectin blot analysis revealed that EMT of RPE cells in vitro confers a glycomic shift towards an abundance of Thomsen-Friedenreich antigen, poly-N-acetyllactosamine chains, and complex-type branched N-glycans. Using inhibitors of glycosylation we found that both, binding of Gal-3 to the RPE cell surface and Gal-3-mediated inhibition of RPE attachment and spreading, strongly depend on the interaction of Gal-3 with tri- or tetra-antennary complex type N-glycans and sialylation of glycans but not on complex-type O-glycans. Importantly, we found that β1,6 N-acetylglucosaminyltransferase V (Mgat5), the key enzyme catalyzing the synthesis of tetra- or tri-antennary complex type N-glycans, is increased upon EMT of RPE cells. Silencing of Mgat5 by siRNA and CRISPR-Cas9 genome editing resulted in reduced Gal-3 binding. We conclude from these data that binding of recombinant Gal-3 to the RPE cell surface and inhibitory effects on RPE attachment and spreading largely dependent on interaction with Mgat5 modified N-glycans, which are more abundant on dedifferentiated than on the healthy, native RPE cells. Based on these findings we hypothesize that EMT of RPE cells in vitro confers glycomic changes, which account for high affinity binding of recombinant Gal-3, particularly to the cell surface of myofibroblastic RPE. From a future perspective recombinant Gal-3 may disclose a therapeutic option allowing for selectively targeting RPE cells with pathogenic relevance for development of PVR.
Collapse
Affiliation(s)
- Claudia S. Priglinger
- Department of Ophthalmology, Ludwig-Maximilians-University, Munich, Germany
- * E-mail:
| | - Jara Obermann
- Research Unit Protein Science, Helmholtz Zentrum Munich, German Research Center for Environmental Health (GmbH), Munich, Germany
| | | | - Juliane Merl-Pham
- Research Unit Protein Science, Helmholtz Zentrum Munich, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Uli Ohmayer
- Research Unit Protein Science, Helmholtz Zentrum Munich, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Jennifer Behler
- Research Unit Protein Science, Helmholtz Zentrum Munich, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Fabian Gruhn
- Research Unit Protein Science, Helmholtz Zentrum Munich, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Thomas C. Kreutzer
- Department of Ophthalmology, Ludwig-Maximilians-University, Munich, Germany
| | | | - Arie Geerlof
- Protein Expression and Purification Facility, Institute of Structural Biology, Helmholtz Zentrum Munich, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | | | - Stefanie M. Hauck
- Research Unit Protein Science, Helmholtz Zentrum Munich, German Research Center for Environmental Health (GmbH), Munich, Germany
| |
Collapse
|
5
|
Zhang S, Lu Z, Mao W, Ahmed AA, Yang H, Zhou J, Jennings N, Rodriguez-Aguayo C, Lopez-Berestein G, Miranda R, Qiao W, Baladandayuthapani V, Li Z, Sood AK, Liu J, Le XF, Bast RC. CDK5 Regulates Paclitaxel Sensitivity in Ovarian Cancer Cells by Modulating AKT Activation, p21Cip1- and p27Kip1-Mediated G1 Cell Cycle Arrest and Apoptosis. PLoS One 2015; 10:e0131833. [PMID: 26146988 PMCID: PMC4492679 DOI: 10.1371/journal.pone.0131833] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 06/06/2015] [Indexed: 01/12/2023] Open
Abstract
Cyclin-dependent kinase 5 (CDK5) is a cytoplasmic serine/ threonine kinase. Knockdown of CDK5 enhances paclitaxel sensitivity in human ovarian cancer cells. This study explores the mechanisms by which CDK5 regulates paclitaxel sensitivity in human ovarian cancers. Multiple ovarian cancer cell lines and xenografts were treated with CDK5 small interfering RNA (siRNA) with or without paclitaxel to examine the effect on cancer cell viability, cell cycle arrest and tumor growth. CDK5 protein was measured by immunohistochemical staining of an ovarian cancer tissue microarray to correlate CDK5 expression with overall patient survival. Knockdown of CDK5 with siRNAs inhibits activation of AKT which significantly correlates with decreased cell growth and enhanced paclitaxel sensitivity in ovarian cancer cell lines. In addition, CDK5 knockdown alone and in combination with paclitaxel induced G1 cell cycle arrest and caspase 3 dependent apoptotic cell death associated with post-translational upregulation and nuclear translocation of TP53 and p27Kip1 as well as TP53-dependent transcriptional induction of p21Cip1 in wild type TP53 cancer cells. Treatment of HEYA8 and A2780 wild type TP53 xenografts in nu/nu mice with CDK5 siRNA and paclitaxel produced significantly greater growth inhibition than either treatment alone. Increased expression of CDK5 in human ovarian cancers correlates inversely with overall survival. CDK5 modulates paclitaxel sensitivity by regulating AKT activation, the cell cycle and caspase-dependent apoptosis. CDK5 inhibition can potentiate paclitaxel activity in human ovarian cancer cells.
Collapse
Affiliation(s)
- Shu Zhang
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of General Surgery, the Second Affiliated Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Zhen Lu
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Weiqun Mao
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Ahmed A. Ahmed
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Hailing Yang
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jinhua Zhou
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Nicholas Jennings
- Departments of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Cristian Rodriguez-Aguayo
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Gabriel Lopez-Berestein
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Roberto Miranda
- Departments of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, Untied States of America
| | - Wei Qiao
- Bioinformatics Computer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Veera Baladandayuthapani
- Bioinformatics Computer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Zongfang Li
- Department of General Surgery, the Second Affiliated Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Anil K. Sood
- Departments of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jinsong Liu
- Departments of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, Untied States of America
| | - Xiao-Feng Le
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (RCB); (XFL)
| | - Robert C. Bast
- Departments of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (RCB); (XFL)
| |
Collapse
|
6
|
Zhang SS, Wang W, Zhao CQ, Xie MJ, Li WY, Yang XL, Lv JG. Inhibitory effects of roscovitine on proliferation and migration of vascular smooth muscle cells in vitro. ACTA ACUST UNITED AC 2014; 34:791-795. [PMID: 25480571 DOI: 10.1007/s11596-014-1354-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 10/30/2014] [Indexed: 12/31/2022]
Abstract
Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are the major cause of in-stent restenosis (ISR). Intervention proliferation and migration of VSMCs is an important strategy for antirestenotic therapy. Roscovitine, a second-generation cyclin-dependent kinase inhibitor, can inhibit cell cycle of multiple cell types. We studied the effects of roscovitine on cell cycle distribution, proliferation and migration of VSMCs in vitro by flow cytometry, BrdU incorporation and wound healing assay, respectively. Our results showed that roscovitine increased the proportion of G0/G1 phase cells after 12 h (69.57±3.65 vs. 92.50±1.68, P=0.000), 24 h (80.87±2.24 vs. 90.25±0.79, P=0.000) and 48 h (88.08±3.86 vs. 88.87±2.43, P=0.427) as compared with control group. Roscovitine inhibited proliferation and migration of VSMCs in a concentration-dependent way. With the increase of concentration, roscovitine showed increased capacity for growth and migration inhibition. Roscovitine (30 μmol/L) led to an almost complete VSMCs growth and migration arrest. Combined with its low toxicity and selective inhibition to ISR-VSMCs, roscovitine may be a potential drug in the treatment of vascular stenosis diseases and particularly useful in the prevention and treatment of ISR.
Collapse
Affiliation(s)
- Shuang-Shuang Zhang
- Department of Cardiovascular Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Cardiovascular Medicine, The Central Hospital of Wuhan, Wuhan, 430014, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chong-Qiang Zhao
- Department of Cardiovascular Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Min-Jie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen-Yu Li
- Department of Cardiovascular Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiang-Li Yang
- Department of Cardiovascular Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia-Gao Lv
- Department of Cardiovascular Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
7
|
Gomes H, Romeiro NC, Braz GRC, de Oliveira EAG, Rodrigues C, da Fonseca RN, Githaka N, Isezaki M, Konnai S, Ohashi K, da Silva Vaz I, Logullo C, Moraes J. Identification and structural-functional analysis of cyclin-dependent kinases of the cattle tick Rhipicephalus (Boophilus) microplus. PLoS One 2013; 8:e76128. [PMID: 24146826 PMCID: PMC3795742 DOI: 10.1371/journal.pone.0076128] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 08/20/2013] [Indexed: 01/08/2023] Open
Abstract
Cyclin-dependent kinases (CDKs) are a family of serine/threonine kinases essential for cell cycle progression. Herein, we describe the participation of CDKs in the physiology of Rhipicephalus microplus, the southern cattle tick and an important disease vector. Firstly, amino acid sequences homologous with CDKs of other organisms were identified from a R. microplus transcriptome database in silico. The analysis of the deduced amino acid sequences of CDK1 and CDK10 from R. microplus showed that both have caspase-3/7 cleavage motifs despite their differences in motif position and length of encoded proteins. CDK1 has two motifs (DKRGD and SAKDA) located opposite to the ATP binding site while CDK10 has only one motif (SLLDN) for caspase 3–7 near the ATP binding site. Roscovitine (Rosco), a purine derivative that inhibits CDK/cyclin complexes by binding to the catalytic domain of the CDK molecule at the ATP binding site, which prevents the transfer of ATP's γphosphoryl group to the substrate. To determine the effect of Rosco on tick CDKs, BME26 cells derived from R. microplus embryo cells were utilized in vitro inhibition assays. Cell viability decreased in the Rosco-treated groups after 24 hours of incubation in a concentration-dependent manner and this was observed up to 48 hours following incubation. To our knowledge, this is the first report on characterization of a cell cycle protein in arachnids, and the sensitivity of BME26 tick cell line to Rosco treatment suggests that CDKs are potential targets for novel drug design to control tick infestation.
Collapse
Affiliation(s)
- Helga Gomes
- Laboratório Integrado de Bioquímica Hatisaburo Masuda, NUPEM - UFRJ, campus Macaé, Avenida São José do Barreto, São José do Barreto, Macaé, RJ, Brazil
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, CCS, Bloco H, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, Brazil
| | - Nelilma C. Romeiro
- Laboratório Integrado de Computação Científica, NUPEM - UFRJ, Campus Macaé, São José do Barreto, Macaé, RJ, Brazil
| | - Gloria R. C. Braz
- Instituto Nacional de Ciência e Tecnologia - Entomologia Molecular, Rio de Janeiro, RJ, Brazil
- Departamento de Bioquímica - Instituto de Química, IQ-UFRJ, Rio de Janeiro, RJ, Brazil
| | | | - Camilla Rodrigues
- Laboratório Integrado de Bioquímica Hatisaburo Masuda, NUPEM - UFRJ, campus Macaé, Avenida São José do Barreto, São José do Barreto, Macaé, RJ, Brazil
| | - Rodrigo Nunes da Fonseca
- Laboratório Integrado de Bioquímica Hatisaburo Masuda, NUPEM - UFRJ, campus Macaé, Avenida São José do Barreto, São José do Barreto, Macaé, RJ, Brazil
- Instituto Nacional de Ciência e Tecnologia - Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Naftaly Githaka
- Laboratory of Infectious Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Nishi, Kita-Ku Sapporo, Japan
| | - Masayoshi Isezaki
- Laboratory of Infectious Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Nishi, Kita-Ku Sapporo, Japan
| | - Satoru Konnai
- Laboratory of Infectious Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Nishi, Kita-Ku Sapporo, Japan
| | - Kazuhiko Ohashi
- Laboratory of Infectious Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Nishi, Kita-Ku Sapporo, Japan
| | - Itabajara da Silva Vaz
- Instituto Nacional de Ciência e Tecnologia - Entomologia Molecular, Rio de Janeiro, RJ, Brazil
- Centro de Biotecnologia e Faculdade de Veterinária, UFRGS, Porto Alegre, RS, Brazil
| | - Carlos Logullo
- Instituto Nacional de Ciência e Tecnologia - Entomologia Molecular, Rio de Janeiro, RJ, Brazil
- Laboratório de Química e Função de Proteínas e Peptídeos, Unidade de Experimentação Animal – CBB - UENF, Horto, Campos dos Goytacazes, RJ, Brazil
| | - Jorge Moraes
- Laboratório Integrado de Bioquímica Hatisaburo Masuda, NUPEM - UFRJ, campus Macaé, Avenida São José do Barreto, São José do Barreto, Macaé, RJ, Brazil
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, CCS, Bloco H, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, Brazil
- Instituto Nacional de Ciência e Tecnologia - Entomologia Molecular, Rio de Janeiro, RJ, Brazil
- * E-mail:
| |
Collapse
|
8
|
|
9
|
Mechanisms of inflammation in proliferative vitreoretinopathy: from bench to bedside. Mediators Inflamm 2012; 2012:815937. [PMID: 23049173 PMCID: PMC3463807 DOI: 10.1155/2012/815937] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 08/09/2012] [Indexed: 11/18/2022] Open
Abstract
Proliferative vitreoretinopathy (PVR) is a vision-threatening disease and a common complication of surgery to correct rhegmatogenous retinal detachment (RRD). Several models of the pathogenesis of this disease have been described with some of these models focusing on the role of inflammatory cells and other models focusing on the role of growth factors and cytokines in the vitreous which come into contact with intraretinal and retinal pigment epithelial cells. New experiments have shed light on the pathogenesis of PVR and offer promising avenues for clinical intervention before PVR develops. One such target is the indirect pathway of activation of platelet-derived growth factor receptor alpha (PDGRα), which plays an important role in PVR. Clinical trials assessing the efficacy of 5-fluorouracil (5-FU) and low-molecular-weight heparin (LMWH), daunorubicin, and 13-cis-retinoic acid, among other therapies, have yielded mixed results. Here we review inflammatory and other mechanisms involved in the pathogenesis of PVR, we highlight important clinical trials, and we discuss how findings at the bench have the potential to be translated to the bedside.
Collapse
|
10
|
Contreras-Vallejos E, Utreras E, Gonzalez-Billault C. Going out of the brain: non-nervous system physiological and pathological functions of Cdk5. Cell Signal 2011; 24:44-52. [PMID: 21924349 DOI: 10.1016/j.cellsig.2011.08.022] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 08/29/2011] [Indexed: 12/23/2022]
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine/threonine kinase that is mostly active in the nervous system, where it regulates several processes such as neuronal migration, actin and microtubule dynamics, axonal guidance, and synaptic plasticity, among other processes. In addition to these known functions, in the past few years, novel roles for Cdk5 outside of the nervous system have been proposed. These include roles in gene transcription, vesicular transport, apoptosis, cell adhesion, and migration in many cell types and tissues such as pancreatic cells, muscle cells, neutrophils, and others. In this review, we will summarize the recently studied non-neuronal functions of Cdk5, with a thorough analysis of the biological consequences of these novel roles.
Collapse
Affiliation(s)
- Erick Contreras-Vallejos
- Department of Biology and Institute for Cell Dynamics and Biotechnology, Faculty of Sciences, Universidad de Chile, Santiago, Chile.
| | | | | |
Collapse
|
11
|
Idowu MA. Cyclin-Dependent Kinases as Drug Targets for Cell Growth and Proliferation Disorders. A Role for Systems Biology Approach in Drug Development. Part I—Cyclin-Dependent Kinases as Drug Targets in Cancer. BIOTECHNOL BIOTEC EQ 2011. [DOI: 10.5504/bbeq.2011.0107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
12
|
Lei H, Velez G, Cui J, Samad A, Maberley D, Matsubara J, Kazlauskas A. N-acetylcysteine suppresses retinal detachment in an experimental model of proliferative vitreoretinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:132-40. [PMID: 20489144 DOI: 10.2353/ajpath.2010.090604] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Proliferative vitreoretinopathy (PVR) is a complication that develops in 5% to 10% of patients who undergo surgery to correct a detached retina. The only treatment option for PVR is surgical intervention, which has a limited success rate that diminishes in patients with recurring PVR. Our recent studies revealed that antioxidants prevented intracellular signaling events that were essential for experimental PVR. The purpose of this study was to test whether N-acetyl-cysteine (NAC), an antioxidant used in a variety of clinical settings, was capable of protecting rabbits from PVR. Vitreous-driven activation of PDGFRalpha and cellular responses intrinsic to PVR (contraction of collagen gels and cell proliferation) were blocked by concentrations of NAC that were well below the maximum tolerated dose. Furthermore, intravitreal injection of NAC effectively protected rabbits from developing retinal detachment, which is the sight-robbing phase of PVR. Finally, these observations with an animal model appear relevant to clinical PVR because NAC prevented human PVR vitreous-induced contraction of primary RPE cells derived from a human PVR membrane. Our observations demonstrate that antioxidants significantly inhibited experimental PVR, and suggest that antioxidants have the potential to function as a PVR prophylactic in patients undergoing retinal surgery to repair a detached retina.
Collapse
Affiliation(s)
- Hetian Lei
- Department of Ophthalmology, Schepens Eye Research Institute, Harvard Medical School, 20 Staniford St, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Cao X, Liu M, Tuo J, Shen D, Chan CC. The effects of quercetin in cultured human RPE cells under oxidative stress and in Ccl2/Cx3cr1 double deficient mice. Exp Eye Res 2010; 91:15-25. [PMID: 20361964 DOI: 10.1016/j.exer.2010.03.016] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 03/16/2010] [Accepted: 03/18/2010] [Indexed: 12/17/2022]
Abstract
Quercetin, a member of the flavonoid family, is one of the most prominent dietary antioxidants. This study investigates the mechanisms for the effects of quercetin on cultured human RPE cells and in Ccl2/Cx3cr1 double knock-out (DKO) mice, which spontaneously develop progressive retinal lesions mimicking age-related macular degeneration (AMD). In the in vitro experiment, cultured ARPE-19 cells were exposed to 1 mM H(2)O(2) with or without 50 muM quercetin for 2 h. Cellular viability, mitochondrial function, and apoptosis were assessed using crystal violet staining, MTT assay, and comet assay, respectively. Apoptotic molecular transcripts of BCL-2, BAX, FADD, CASPASE-3 and CASPASE-9 were measured by RQ-PCR. COX activity and nitric oxide (NO) level were determined in the supernatant of the culture medium. Quercetin treatment protected ARPE-19 cells from H(2)O(2)-induced oxidative injury, enhanced BCL-2 transcript levels, increased the BCL-2/BAX ratio, suppressed the transcription of pro-apoptotic factors such as BAX, FADD, CASPASE-3 and CASPASE-9, inhibited the transcription of inflammatory factors such as TNF-alpha, COX-2 and INOS, and decreased the levels of COX and NO in the culture medium. In the in vivo experiment, DKO and C57/B6 mice were treated with 25 mg/kg/day quercetin by intraperitoneal injection daily for two months. Funduscopy was performed monthly. After two months, serum was collected to measure NADP(+)/NADPH, COX, PGE-2, and NO levels. The eyes were harvested for histology and A2E measurement. Ocular transcripts of Bcl-2, Bax, Cox-2, Inos, Tnf-alpha, Fas, FasL and Caspase-3 were detected by RQ-PCR. Quercetin treatment did not reverse the progression of retinal lesions in DKO mice funduscopically or histologically. Although quercetin treatment could recover systemic anti-oxidative capacity, suppress the systemic expression of NO, COX and PGE-2, and decrease ocular A2E levels, it could not effectively suppress the transcripts of the ocular inflammatory factors Tnf-alpha, Cox-2 and Inos, or the pro-apoptotic factors Fas, FasL and Caspase-3 in DKO mice. Our data demonstrate that quercetin can protect human RPE cells from oxidative stress in vitro via inhibition of pro-inflammatory molecules and direct inhibition of the intrinsic apoptosis pathway. However, quercetin (25 mg/kg/day) does not improve the retinal AMD-like lesions in the Ccl2(-/-)/Cx3cr1(-/-) mice, likely due to its insufficient suppression of the inflammatory and apoptosis pathways in the eye.
Collapse
Affiliation(s)
- Xiaoguang Cao
- Immunopathology Section, Laboratory of Immunology; National Eye Institute, National Institutes of Health, Bethesda, MD 20892-1857, USA
| | | | | | | | | |
Collapse
|