151
|
Yan T, Ding F, Zhao Y. Integrated identification of key genes and pathways in Alzheimer's disease via comprehensive bioinformatical analyses. Hereditas 2019; 156:25. [PMID: 31346329 PMCID: PMC6636172 DOI: 10.1186/s41065-019-0101-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/09/2019] [Indexed: 12/23/2022] Open
Abstract
Background Alzheimer's disease (AD) is known to be caused by multiple factors, meanwhile the pathogenic mechanism and development of AD associate closely with genetic factors. Existing understanding of the molecular mechanisms underlying AD remains incomplete. Methods Gene expression data (GSE48350) derived from post-modern brain was extracted from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were derived from hippocampus and entorhinal cortex regions between AD patients and healthy controls and detected via Morpheus. Functional enrichment analyses, including Gene Ontology (GO) and pathway analyses of DEGs, were performed via Cytoscape and followed by the construction of protein-protein interaction (PPI) network. Hub proteins were screened using the criteria: nodes degree≥10 (for hippocampus tissues) and ≥ 8 (for entorhinal cortex tissues). Molecular Complex Detection (MCODE) was used to filtrate the important clusters. University of California Santa Cruz (UCSC) and the database of RNA-binding protein specificities (RBPDB) were employed to identify the RNA-binding proteins of the long non-coding RNA (lncRNA). Results 251 & 74 genes were identified as DEGs, which consisted of 56 & 16 up-regulated genes and 195 & 58 down-regulated genes in hippocampus and entorhinal cortex, respectively. Biological analyses demonstrated that the biological processes and pathways related to memory, transmembrane transport, synaptic transmission, neuron survival, drug metabolism, ion homeostasis and signal transduction were enriched in these genes. 11 genes were identified as hub genes in hippocampus and entorhinal cortex, and 3 hub genes were identified as the novel candidates involved in the pathology of AD. Furthermore, 3 lncRNAs were filtrated, whose binding proteins were closely associated with AD. Conclusions Through GO enrichment analyses, pathway analyses and PPI analyses, we showed a comprehensive interpretation of the pathogenesis of AD at a systematic biology level, and 3 novel candidate genes and 3 lncRNAs were identified as novel and potential candidates participating in the pathology of AD. The results of this study could supply integrated insights for understanding the pathogenic mechanism underlying AD and potential novel therapeutic targets.
Collapse
Affiliation(s)
- Tingting Yan
- Department of Bioengineering, Harbin Institute of Technology, Weihai, 264209 Shandong China
| | - Feng Ding
- Department of Bioengineering, Harbin Institute of Technology, Weihai, 264209 Shandong China
| | - Yan Zhao
- Department of Bioengineering, Harbin Institute of Technology, Weihai, 264209 Shandong China
| |
Collapse
|
152
|
Senoo H, Kamimura Y, Kimura R, Nakajima A, Sawai S, Sesaki H, Iijima M. Phosphorylated Rho-GDP directly activates mTORC2 kinase towards AKT through dimerization with Ras-GTP to regulate cell migration. Nat Cell Biol 2019; 21:867-878. [PMID: 31263268 PMCID: PMC6650273 DOI: 10.1038/s41556-019-0348-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 05/29/2019] [Indexed: 11/19/2022]
Abstract
mTORC2 plays critical roles in metabolism, cell survival and actin cytoskeletal dynamics through the phosphorylation of AKT. Despite its importance to biology and medicine, it is unclear how mTORC2-mediated AKT phosphorylation is controlled. Here, we identify an unforeseen principle by which a GDP-bound form of the conserved small G protein Rho GTPase directly activates mTORC2 in AKT phosphorylation in social amoebae (Dictyostelium discoideum) cells. Using biochemical reconstitution with purified proteins, we demonstrate that Rho-GDP promotes AKT phosphorylation by assembling a supercomplex with Ras-GTP and mTORC2. This supercomplex formation is controlled by the chemoattractant-induced phosphorylation of Rho-GDP at S192 by GSK-3. Furthermore, Rho-GDP rescues defects in both mTORC2-mediated AKT phosphorylation and directed cell migration in Rho-null cells in a manner dependent on phosphorylation of S192. Thus, in contrast to the prevailing view that the GDP-bound forms of G proteins are inactive, our study reveals that mTORC2-AKT signalling is activated by Rho-GDP.
Collapse
Affiliation(s)
- Hiroshi Senoo
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yoichiro Kamimura
- Laboratory for Cell Signaling Dynamics, Quantitative Biology Center, RIKEN, Suita, Japan
| | - Reona Kimura
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akihiko Nakajima
- Department of Basic Science, Graduate School of Arts and Sciences, University of Tokyo, Tokyo, Japan
| | - Satoshi Sawai
- Department of Basic Science, Graduate School of Arts and Sciences, University of Tokyo, Tokyo, Japan
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Miho Iijima
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
153
|
Burridge K, Monaghan-Benson E, Graham DM. Mechanotransduction: from the cell surface to the nucleus via RhoA. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180229. [PMID: 31431179 PMCID: PMC6627015 DOI: 10.1098/rstb.2018.0229] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Cells respond and adapt to their physical environments and to the mechanical forces that they experience. The translation of physical forces into biochemical signalling pathways is known as mechanotransduction. In this review, we focus on two aspects of mechanotransduction. First, we consider how forces exerted on cell adhesion molecules at the cell surface regulate the RhoA signalling pathway by controlling the activities of guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs). In the second part of the review, we discuss how the nucleus contributes to mechanotransduction as a physical structure connected to the cytoskeleton. We focus on recent studies that have either severed the connections between the nucleus and the cytoskeleton, or that have entirely removed the nucleus from cells. These actions reduce the levels of active RhoA, thereby altering the mechanical properties of cells and decreasing their ability to generate tension and respond to external mechanical forces. This article is part of a discussion meeting issue ‘Forces in cancer: interdisciplinary approaches in tumour mechanobiology’.
Collapse
Affiliation(s)
- Keith Burridge
- Department of Cell Biology and Physiology, and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Elizabeth Monaghan-Benson
- Department of Cell Biology and Physiology, and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - David M Graham
- Department of Cell Biology and Physiology, and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
154
|
Wang L, Cheng B, Li H, Wang Y. Proteomics analysis of preadipocytes between fat and lean broilers. Br Poult Sci 2019; 60:522-529. [PMID: 31132862 DOI: 10.1080/00071668.2019.1621989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
1. Reducing excessive chicken body fat deposition is a main goal of the poultry industry. Preadipocytes are important in adipose tissue growth and development. 2. To discover proteins related to chicken fat deposition, two-dimensional fluorescence difference gel electrophoresis (2-D DIGE) was used to identify differentially expressed proteins in preadipocytes derived from Northeast Agricultural University broiler lines divergently selected for abdominal fat content (NEAUHLF). 3. A total of 46 differentially expressed protein spots were found in the preadipocytes between fat and lean broilers. Matrix-assisted laser desorption-ionisation time-of-flight mass spectrometry (MALDI-TOF-MS) analysis showed the protein spots corresponded to 33 different proteins. The proteins were mainly related to biological oxidation, cell proliferation, cytoskeleton, lipid metabolism, molecular chaperone, protein synthesis and signal transduction. 4. From the perspective of protein expression, these results lay a foundation for further study of the genetic mechanism of broiler adipose tissue growth and development.
Collapse
Affiliation(s)
- L Wang
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Chicken Genetics and Breeding , Harbin , P. R. China.,Department of Education of Heilongjiang Province, Key Laboratory of Animal Genetics, Breeding and Reproduction , Harbin , P. R. China.,College of Animal Science and Technology, Northeast Agricultural University , Harbin , P. R. China
| | - B Cheng
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Chicken Genetics and Breeding , Harbin , P. R. China.,Department of Education of Heilongjiang Province, Key Laboratory of Animal Genetics, Breeding and Reproduction , Harbin , P. R. China.,College of Animal Science and Technology, Northeast Agricultural University , Harbin , P. R. China
| | - H Li
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Chicken Genetics and Breeding , Harbin , P. R. China.,Department of Education of Heilongjiang Province, Key Laboratory of Animal Genetics, Breeding and Reproduction , Harbin , P. R. China.,College of Animal Science and Technology, Northeast Agricultural University , Harbin , P. R. China
| | - Y Wang
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Chicken Genetics and Breeding , Harbin , P. R. China.,Department of Education of Heilongjiang Province, Key Laboratory of Animal Genetics, Breeding and Reproduction , Harbin , P. R. China.,College of Animal Science and Technology, Northeast Agricultural University , Harbin , P. R. China
| |
Collapse
|
155
|
Kumar D, Mains RE, Eipper BA, King SM. Ciliary and cytoskeletal functions of an ancient monooxygenase essential for bioactive amidated peptide synthesis. Cell Mol Life Sci 2019; 76:2329-2348. [PMID: 30879092 PMCID: PMC6529398 DOI: 10.1007/s00018-019-03065-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/28/2019] [Accepted: 03/07/2019] [Indexed: 02/07/2023]
Abstract
Many secreted peptides used for cell-cell communication require conversion of a C-terminal glycine to an amide for bioactivity. This reaction is catalyzed only by the integral membrane protein peptidylglycine α-amidating monooxygenase (PAM). PAM has been highly conserved and is found throughout the metazoa; PAM-like sequences are also present in choanoflagellates, filastereans, unicellular and colonial chlorophyte green algae, dinoflagellates and haptophytes. Recent studies have revealed that in addition to playing a key role in peptidergic signaling, PAM also regulates ciliogenesis in vertebrates, planaria and chlorophyte algae, and is required for the stability of actin-based microvilli. Here we briefly introduce the basic principles involved in ciliogenesis, the sequential reactions catalyzed by PAM and the trafficking of PAM through the secretory and endocytic pathways. We then discuss the multi-faceted roles this enzyme plays in the formation and maintenance of cytoskeleton-based cellular protrusions and propose models for how PAM protein and amidating activity might contribute to ciliogenesis. Finally, we consider why some ciliated organisms lack PAM, and discuss the potential ramifications of ciliary localized PAM for the endocrine features commonly observed in patients with ciliopathies.
Collapse
Affiliation(s)
- Dhivya Kumar
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Betty A Eipper
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Stephen M King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| |
Collapse
|
156
|
Kesar S, Paliwal SK, Mishra P, Chauhan M. Quantitative Structure-Activity Relationship Analysis of Selective Rho Kinase Inhibitors as Neuro-regenerator Agents. Turk J Pharm Sci 2019; 16:141-154. [PMID: 32454707 DOI: 10.4274/tjps.galenos.2018.70288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 01/25/2018] [Indexed: 12/01/2022]
Abstract
OBJECTIVES To understand the role of Rho (serine/threonine) kinases in the treatment of neurological segments, attempts have been made to find potent inhibitors of Rho enzyme by a 2D quantitative structure-activity relationship (QSAR) model. MATERIALS AND METHODS QSAR studies were executed on urea-based scaffolds from aniline and benzylamine analogues, which were aligned for generation of a chemometric-based model. Multivariate statistical approaches were applied including linear and nonlinear analysis such as multiple linear regression, partial least square and artificial neural network for the generation of model, and also an application of (in silico) absorption, distribution, metabolism, excretion studies was performed to ascertain the novelty and drug-like properties of the intended molecules. RESULTS Ligand based analysis was implemented and showed excellent statistical relevance such as S value=0.38, F value=48.41, r=0.95, r²=0.91, and r²cv=0.86. Five illuminating variables, i.e., vesicle-associated membrane protein (VAMP) polarization YY component (whole molecule), VAMP dipole Y component (whole molecule), VAMP dipole Z component (whole molecule), Kier ChiV6 path index (whole molecule), and moment of inertia 2 size (whole molecule), were found and they have a profound influence on the potency of the compounds. CONCLUSION The values of standard statistical parameters reveal the predictive power and robustness of this model and also provide valuable insight into the significance of five descriptors. The acquired physicochemical properties (electronic, topological, and steric) show the important structural features required for activity against Rho kinase. After performing Lipinski's rule of five on urea-based derivatives no molecule was violating the rule. Therefore, these features can be effectively employed for the modeling and screening of active neurological agents as novel Rho kinase inhibitors.
Collapse
Affiliation(s)
- Seema Kesar
- Banasthali University, Faculty of Pharmacy, Department of Pharmacy, Rajasthan, India
| | - Sarvesh K Paliwal
- Banasthali University, Faculty of Pharmacy, Department of Pharmacy, Rajasthan, India
| | - Pooja Mishra
- Banasthali University, Faculty of Pharmacy, Department of Pharmacy, Rajasthan, India
| | - Monika Chauhan
- Banasthali University, Faculty of Pharmacy, Department of Pharmacy, Rajasthan, India
| |
Collapse
|
157
|
Dehnavi S, Sadeghi M, Johnston TP, Barreto G, Shohan M, Sahebkar A. The role of protein SUMOylation in rheumatoid arthritis. J Autoimmun 2019; 102:1-7. [PMID: 31078376 DOI: 10.1016/j.jaut.2019.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/02/2019] [Accepted: 05/04/2019] [Indexed: 01/09/2023]
Abstract
Small ubiquitin-like modifier (SUMO) proteins, as a subgroup of post-translational modifiers, act to change the function of proteins. Through their interactions with different targets, immune pathways, and the responses they elicit, can be affected by these SUMO conjugations. Thus, both a change to protein function and involvement in immune pathways has the potential to promote an efficient immune response to either a pathogenic challenge, or the development of an imbalance that could lead to an autoimmune-based disease. Also, a variety of changes such as mutations and polymorphisms can interfere with common functions of these modifications and move an effective immune response in the direction of an autoimmune disease. The present review discusses the general characteristics of SUMO proteins and focuses on their involvement in rheumatoid arthritis as an autoimmune disease.
Collapse
Affiliation(s)
- Sajad Dehnavi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - George Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia; Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Mojtaba Shohan
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
158
|
Phuong TTT, Walker AE, Henson GD, Machin DR, Li DY, Donato AJ, Lesniewski LA. Deletion of Robo4 prevents high-fat diet-induced adipose artery and systemic metabolic dysfunction. Microcirculation 2019; 26:e12540. [PMID: 30825241 DOI: 10.1111/micc.12540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/22/2019] [Accepted: 02/27/2019] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Accumulating evidence suggests the vascular endothelium plays a fundamental role in the pathophysiology of obesity by regulating the functional status of white adipose and systemic metabolism. Robo4 is expressed specifically in endothelial cells and increases vascular stability and inhibits angiogenesis. We sought to determine the role of Robo4 in modulating cardiometabolic function in response to high-fat feeding. METHODS We examined exercise capacity, glucose tolerance, and white adipose tissue artery gene expression, endothelium-dependent dilation (EDD), and angiogenesis in wild type and Robo4 knockout (KO) mice fed normal chow (NC) or a high-fat diet (HFD). RESULTS We found Robo4 deletion enhances exercise capacity in NC-fed mice and HFD markedly increased the expression of the Robo4 ligand, Slit2, in white adipose tissue. Deletion of Robo4 increased angiogenesis in white adipose tissue and protected against HFD-induced impairments in white adipose artery vasodilation and glucose intolerance. CONCLUSIONS We demonstrate a novel functional role for Robo4 in endothelial cell function and metabolic homeostasis in white adipose tissue, with Robo4 deletion protecting against endothelial and metabolic dysfunction associated with a HFD. Our findings suggest that Robo4-dependent signaling pathways may be a novel target in anti-obesity therapy.
Collapse
Affiliation(s)
- Tam T T Phuong
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Ashley E Walker
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Grant D Henson
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Daniel R Machin
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Dean Y Li
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah.,Division of Cardiovascular Medicine Department of Medicine, University of Utah, Salt Lake City, Utah.,Department of Human Genetics, University of Utah, Salt Lake City, Utah
| | - Anthony J Donato
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Salt Lake City Veteran's Affair Medical Center, Geriatrics Research Education and Clinic Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Lisa A Lesniewski
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Salt Lake City Veteran's Affair Medical Center, Geriatrics Research Education and Clinic Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
159
|
Kakiuchi A, Kohno T, Kakuki T, Kaneko Y, Konno T, Hosaka Y, Hata T, Kikuchi S, Ninomiya T, Himi T, Takano K, Kojima T. Rho-kinase and PKCα Inhibition Induces Primary Cilia Elongation and Alters the Behavior of Undifferentiated and Differentiated Temperature-sensitive Mouse Cochlear Cells. J Histochem Cytochem 2019; 67:523-535. [PMID: 30917058 DOI: 10.1369/0022155419841013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Primary cilia, regulated via distinct signal transduction pathways, play crucial roles in various cellular behaviors. However, the full regulatory mechanism involved in primary cilia development during cellular differentiation is not fully understood, particularly for the sensory hair cells of the mammalian cochlea. In this study, we investigated the effects of the Rho-kinase inhibitor Y27632 and PKCα inhibitor GF109203X on primary cilia-related cell behavior in undifferentiated and differentiated temperature-sensitive mouse cochlear precursor hair cells (the conditionally immortalized US/VOT-E36 cell line). Our results indicate that treatment with Y27632 or GF109203X induced primary cilia elongation and tubulin acetylation in both differentiated and undifferentiated cells. Concomitant with cilia elongation, Y27632 treatment also increased Hook2 and cyclinD1 expression, while only Hook2 expression was increased after treatment with GF109203X. In the undifferentiated cells, we observed an increase in the number of S and G2/M stage cells and a decrease of G1 cells after treatment with Y27632, while the opposite was observed after treatment with GF109203X. Finally, while both treatments decreased oxidative stress, only treatment with Y27632, not GF109203X, induced cell cycle-dependent cell proliferation and cell migration.
Collapse
Affiliation(s)
- Akito Kakiuchi
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takuya Kakuki
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yakuto Kaneko
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yukino Hosaka
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomohiro Hata
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shin Kikuchi
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takafumi Ninomiya
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tetsuo Himi
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenichi Takano
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
160
|
Li H, Liu L, Zhuang J, Liu C, Zhou C, Yang J, Gao C, Liu G, Sun C. Deciphering the mechanism of Indirubin and its derivatives in the inhibition of Imatinib resistance using a "drug target prediction-gene microarray analysis-protein network construction" strategy. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:75. [PMID: 30909944 PMCID: PMC6434895 DOI: 10.1186/s12906-019-2471-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/04/2019] [Indexed: 12/25/2022]
Abstract
Background The introduction of imatinib revolutionized the treatment of chronic myeloid leukaemia (CML), substantially extending patient survival. However, imatinib resistance is currently a clinical problem for CML. It is very importantto find a strategy to inhibit imatinib resistance. Methods (1) We Identified indirubin and its derivatives and predicted its putative targets; (2) We downloaded data of the gene chip GSE2810 from the Gene Expression Omnibus (GEO) database and performed GEO2R analysis to obtain differentially expressed genes (DEGs); and (3) we constructed a P-P network of putative targets and DEGs to explore the mechanisms of action and to verify the results of molecular docking. Result We Identified a total of 42 small-molecule compounds, of which 15 affected 11 putative targets, indicating the potential to inhibit imatinib resistance; the results of molecular docking verified these results. Six biomarkers of imatinib resistance were characterised by analysing DEGs. Conclusion The 15 small molecule compounds inhibited imatinib resistance through the cytokine-cytokine receptor signalling pathway, the JAK-stat pathway, and the NF-KB signalling pathway. Indirubin and its derivatives may be new drugsthat can combat imatinib resistance. Electronic supplementary material The online version of this article (10.1186/s12906-019-2471-2) contains supplementary material, which is available to authorized users.
Collapse
|
161
|
An In Vitro Kinase Assay to Assess Rac1 Phosphorylation by ERK. Methods Mol Biol 2019; 1821:131-140. [PMID: 30062409 DOI: 10.1007/978-1-4939-8612-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Recent findings suggest that phosphorylation might further contribute to the tight regulation of Rho GTPases. Interestingly, sequence analysis of Rac1 shows that T108 within the 106PNTP109 motif of Rac1 is likely an ERK phosphorylation site and Rac1 also has an ERK docking site 183KKRKRKCLLL192 (D-site) at the C-terminus. Protein phosphorylation could be assayed by many different methods. Here, we describe an in vitro kinase assay we used to assess Rac1 phosphorylation by ERK. Rac1 phosphorylation is detected based on the transfer of a radiolabeled phosphate from ATP to Rac1 by the phosphotransferase activity of the kinase EKR. This in vitro kinase assay uses commercially available purified active ERK. Substrate Rac1 was generated and purified as a glutathione S-transferase (GST) fusion protein. [γ-32P]ATP is used to radiolabel Rac1. Phosphorylation of Rac1 is viewed by autoradiography.
Collapse
|
162
|
Abstract
Small, monomeric guanine triphosphate hydrolases (GTPases) are ubiquitous cellular integrators of signaling. A signal activates the GTPase, which then binds to an effector molecule to relay a signal inside the cell. The GTPase effector trap flow cytometry assay (G-Trap) utilizes bead-based protein immobilization and dual-color flow cytometry to rapidly and quantitatively measure GTPase activity status in cell or tissue lysates. Beginning with commercial cytoplex bead sets that are color-coded with graded fluorescence intensities of a red (700 nm) wavelength, the bead sets are derivatized to display glutathione on the surface through a detailed protocol described here. A different glutathione-S-transferase-effector protein (GST-effector protein) can then be attached to the surface of each set. For the assay, users can incubate bead sets individually or in a multiplex format with lysates for rapid, selective capture of active, GTP-bound GTPases from a single sample. After that, flow cytometry is used to identify the bead-borne GTPase based on red bead intensity, and the amount of active GTPase per bead is detected using monoclonal antibodies conjugated to a green fluorophore or via labeled secondary antibodies. Three examples are provided to illustrate the efficacy of the effector-functionalized beads for measuring the activation of at least five GTPases in a single lysate from fewer than 50,000 cells.
Collapse
|
163
|
Acebedo AR, Suzuki K, Hino S, Alcantara MC, Sato Y, Haga H, Matsumoto KI, Nakao M, Shimamura K, Takeo T, Nakagata N, Miyagawa S, Nishinakamura R, Adelstein RS, Yamada G. Mesenchymal actomyosin contractility is required for androgen-driven urethral masculinization in mice. Commun Biol 2019; 2:95. [PMID: 30886905 PMCID: PMC6408527 DOI: 10.1038/s42003-019-0336-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/01/2019] [Indexed: 01/23/2023] Open
Abstract
The morphogenesis of mammalian embryonic external genitalia (eExG) shows dynamic differences between males and females. In genotypic males, eExG are masculinized in response to androgen signaling. Disruption of this process can give rise to multiple male reproductive organ defects. Currently, mechanisms of androgen-driven sexually dimorphic organogenesis are still unclear. We show here that mesenchymal-derived actomyosin contractility, by MYH10, is essential for the masculinization of mouse eExG. MYH10 is expressed prominently in the bilateral mesenchyme of male eExG. Androgen induces MYH10 protein expression and actomyosin contractility in the bilateral mesenchyme. Inhibition of actomyosin contractility through blebbistatin treatment and mesenchymal genetic deletion induced defective urethral masculinization with reduced mesenchymal condensation. We also suggest that actomyosin contractility regulates androgen-dependent mesenchymal directional cell migration to form the condensation in the bilateral mesenchyme leading to changes in urethral plate shape to accomplish urethral masculinization. Thus, mesenchymal-derived actomyosin contractility is indispensable for androgen-driven urethral masculinization.
Collapse
Affiliation(s)
- Alvin R. Acebedo
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509 Japan
| | - Kentaro Suzuki
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509 Japan
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811 Japan
| | - Mellissa C. Alcantara
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509 Japan
| | - Yuki Sato
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Hisashi Haga
- Transdisciplinary Life Science Course, Faculty of Advanced Life Science, Hokkaido University, N10-W8, Kita-ku, Sapporo, 060-0810 Japan
| | - Ken-ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research, Shimane University, Izumo, Shimane, 693-8501 Japan
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811 Japan
| | - Kenji Shimamura
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811 Japan
| | - Toru Takeo
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811 Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811 Japan
| | - Shinichi Miyagawa
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509 Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811 Japan
| | - Robert S. Adelstein
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1762 USA
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509 Japan
| |
Collapse
|
164
|
Activated Protein C in Cutaneous Wound Healing: From Bench to Bedside. Int J Mol Sci 2019; 20:ijms20040903. [PMID: 30791425 PMCID: PMC6412604 DOI: 10.3390/ijms20040903] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/14/2019] [Accepted: 02/16/2019] [Indexed: 12/12/2022] Open
Abstract
Independent of its well-known anticoagulation effects, activated protein C (APC) exhibits pleiotropic cytoprotective properties. These include anti-inflammatory actions, anti-apoptosis, and endothelial and epithelial barrier stabilisation. Such beneficial effects have made APC an attractive target of research in a plethora of physiological and pathophysiological processes. Of note, the past decade or so has seen the emergence of its roles in cutaneous wound healing-a complex process involving inflammation, proliferation and remodelling. This review will highlight APC's functions and mechanisms, and detail its pre-clinical and clinical studies on cutaneous wound healing.
Collapse
|
165
|
Lysophosphatidic acid increases mesangial cell proliferation in models of diabetic nephropathy via Rac1/MAPK/KLF5 signaling. Exp Mol Med 2019; 51:1-10. [PMID: 30770784 PMCID: PMC6377648 DOI: 10.1038/s12276-019-0217-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 11/20/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022] Open
Abstract
Mesangial cell proliferation has been identified as a major factor contributing to glomerulosclerosis, which is a typical symptom of diabetic nephropathy (DN). Lysophosphatidic acid (LPA) levels are increased in the glomerulus of the kidney in diabetic mice. LPA is a critical regulator that induces mesangial cell proliferation; however, its effect and molecular mechanisms remain unknown. The proportion of α-SMA+/PCNA+ cells was increased in the kidney cortex of db/db mice compared with control mice. Treatment with LPA concomitantly increased the proliferation of mouse mesangial cells (SV40 MES13) and the expression of cyclin D1 and CDK4. On the other hand, the expression of p27Kip1 was decreased. The expression of Krüppel-like factor 5 (KLF5) was upregulated in the kidney cortex of db/db mice and LPA-treated SV40 MES13 cells. RNAi-mediated silencing of KLF5 reversed these effects and inhibited the proliferation of LPA-treated cells. Mitogen-activated protein kinases (MAPKs) were activated, and the expression of early growth response 1 (Egr1) was subsequently increased in LPA-treated SV40 MES13 cells and the kidney cortex of db/db mice. Moreover, LPA significantly increased the activity of the Ras-related C3 botulinum toxin substrate (Rac1) GTPase in SV40 MES13 cells, and the dominant-negative form of Rac1 partially inhibited the phosphorylation of p38 and upregulation of Egr1 and KLF5 induced by LPA. LPA-induced hyperproliferation was attenuated by the inhibition of Rac1 activity. Based on these results, the Rac1/MAPK/KLF5 signaling pathway was one of the mechanisms by which LPA induced mesangial cell proliferation in DN models.
Collapse
|
166
|
Santoro R, Perrucci GL, Gowran A, Pompilio G. Unchain My Heart: Integrins at the Basis of iPSC Cardiomyocyte Differentiation. Stem Cells Int 2019; 2019:8203950. [PMID: 30906328 PMCID: PMC6393933 DOI: 10.1155/2019/8203950] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
The cellular response to the extracellular matrix (ECM) microenvironment mediated by integrin adhesion is of fundamental importance, in both developmental and pathological processes. In particular, mechanotransduction is of growing importance in groundbreaking cellular models such as induced pluripotent stem cells (iPSC), since this process may strongly influence cell fate and, thus, augment the precision of differentiation into specific cell types, e.g., cardiomyocytes. The decryption of the cellular machinery starting from ECM sensing to iPSC differentiation calls for new in vitro methods. Conveniently, engineered biomaterials activating controlled integrin-mediated responses through chemical, physical, and geometrical designs are key to resolving this issue and could foster clinical translation of optimized iPSC-based technology. This review introduces the main integrin-dependent mechanisms and signalling pathways involved in mechanotransduction. Special consideration is given to the integrin-iPSC linkage signalling chain in the cardiovascular field, focusing on biomaterial-based in vitro models to evaluate the relevance of this process in iPSC differentiation into cardiomyocytes.
Collapse
Affiliation(s)
- Rosaria Santoro
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Aoife Gowran
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Giulio Pompilio
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, via Festa del Perdono 7, Milan, Italy
| |
Collapse
|
167
|
Al-Rekabi Z, Fura AM, Juhlin I, Yassin A, Popowics TE, Sniadecki NJ. Hyaluronan-CD44 interactions mediate contractility and migration in periodontal ligament cells. Cell Adh Migr 2019; 13:138-150. [PMID: 30676222 PMCID: PMC6527381 DOI: 10.1080/19336918.2019.1568140] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The role of hyaluronan (HA) in periodontal healing has been speculated via its interaction with the CD44 receptor. While HA-CD44 interactions have previously been implicated in numerous cell types; effect and mechanism of exogenous HA on periodontal ligament (PDL) cells is less clear. Herein, we examine the effect of exogenous HA on contractility and migration in human and murine PDL cells using arrays of microposts and time-lapse microscopy. Our findings observed HA-treated human PDL cells as more contractile and less migratory than untreated cells. Moreover, the effect of HA on contractility and focal adhesion area was abrogated when PDL cells were treated with Y27632, an inhibitor of rho-dependent kinase, but not when these cells were treated with ML-7, an inhibitor of myosin light chain kinase. Our results provide insight into the mechanobiology of PDL cells, which may contribute towards the development of therapeutic strategies for periodontal healing and tissue regeneration.
Collapse
Affiliation(s)
- Zeinab Al-Rekabi
- a Department of Mechanical Engineering , University of Washington , Seattle , WA , USA
| | - Adriane M Fura
- b Department of Bioengineering , University of Washington , Seattle , WA , USA
| | - Ilsa Juhlin
- a Department of Mechanical Engineering , University of Washington , Seattle , WA , USA
| | - Alaa Yassin
- c Department of Periodontics , University of Washington , Seattle , WA , USA
| | - Tracy E Popowics
- d Department of Oral Health Sciences , University of Washington , Seattle , WA , USA
| | - Nathan J Sniadecki
- a Department of Mechanical Engineering , University of Washington , Seattle , WA , USA.,b Department of Bioengineering , University of Washington , Seattle , WA , USA.,e Institute for Stem Cell and Regenerative Medicine , University of Washington , Seattle , WA , USA
| |
Collapse
|
168
|
Huff LP, Kikuchi DS, Faidley E, Forrester SJ, Tsai MZ, Lassègue B, Griendling KK. Polymerase-δ-interacting protein 2 activates the RhoGEF epithelial cell transforming sequence 2 in vascular smooth muscle cells. Am J Physiol Cell Physiol 2019; 316:C621-C631. [PMID: 30726115 DOI: 10.1152/ajpcell.00208.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Polymerase-δ-interacting protein 2 (Poldip2) controls a wide variety of cellular functions and vascular pathologies. To mediate these effects, Poldip2 interacts with numerous proteins and generates reactive oxygen species via the enzyme NADPH oxidase 4 (Nox4). We have previously shown that Poldip2 can activate the Rho family GTPase RhoA, another signaling node within the cell. In this study, we aimed to better understand how Poldip2 activates Rho family GTPases and the functions of the involved proteins in vascular smooth muscle cells (VSMCs). RhoA is activated by guanine nucleotide exchange factors. Using nucleotide-free RhoA (isolated from bacteria) to pulldown active RhoGEFs, we found that the RhoGEF epithelial cell transforming sequence 2 (Ect2) is activated by Poldip2. Ect2 is a critical RhoGEF for Poldip2-mediated RhoA activation, because siRNA against Ect2 prevented Poldip2-mediated RhoA activity (measured by rhotekin pulldowns). Surprisingly, we were unable to detect a direct interaction between Poldip2 and Ect2, as they did not coimmunoprecipitate. Nox4 is not required for Poldip2-driven Ect2 activation, as Poldip2 overexpression induced Ect2 activation in Nox4 knockout VSMCs similar to wild-type cells. However, antioxidant treatment blocked Poldip2-induced Ect2 activation. This indicates a novel reactive oxygen species-driven mechanism by which Poldip2 regulates Rho family GTPases. Finally, we examined the function of these proteins in VSMCs, using siRNA against Poldip2 or Ect2 and determined that Poldip2 and Ect2 are both essential for vascular smooth muscle cell cytokinesis and proliferation.
Collapse
Affiliation(s)
- Lauren Parker Huff
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Daniel Seicho Kikuchi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Elizabeth Faidley
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Steven J Forrester
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Michelle Z Tsai
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Bernard Lassègue
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Kathy K Griendling
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| |
Collapse
|
169
|
Schwager SC, Taufalele PV, Reinhart-King CA. Cell-Cell Mechanical Communication in Cancer. Cell Mol Bioeng 2019; 12:1-14. [PMID: 31565083 PMCID: PMC6764766 DOI: 10.1007/s12195-018-00564-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022] Open
Abstract
Communication between cancer cells enables cancer progression and metastasis. While cell-cell communication in cancer has primarily been examined through chemical mechanisms, recent evidence suggests that mechanical communication through cell-cell junctions and cell-ECM linkages is also an important mediator of cancer progression. Cancer and stromal cells remodel the ECM through a variety of mechanisms, including matrix degradation, cross-linking, deposition, and physical remodeling. Cancer cells sense these mechanical environmental changes through cell-matrix adhesion complexes and subsequently alter their tension between both neighboring cells and the surrounding matrix, thereby altering the force landscape within the microenvironment. This communication not only allows cancer cells to communicate with each other, but allows stromal cells to communicate with cancer cells through matrix remodeling. Here, we review the mechanisms of intercellular force transmission, the subsequent matrix remodeling, and the implications of this mechanical communication on cancer progression.
Collapse
Affiliation(s)
- Samantha C. Schwager
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, Nashville, TN 37235 USA
| | - Paul V. Taufalele
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, Nashville, TN 37235 USA
| | - Cynthia A. Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, Nashville, TN 37235 USA
| |
Collapse
|
170
|
Kahl BS, Dreyling M, Gordon LI, Martin P, Quintanilla-Martinez L, Sotomayor EM. Recent advances and future directions in mantle cell lymphoma research: report of the 2018 mantle cell lymphoma consortium workshop. Leuk Lymphoma 2019; 60:1853-1865. [PMID: 30696305 DOI: 10.1080/10428194.2019.1571205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell non-Hodgkin lymphoma characterized by the t(11;14) chromosomal translocation. This translocation most often results in overexpression of cyclin D1. MCL is clinically heterogeneous, outcomes are generally poor, and no standard treatment has been established. The recent approvals of ibrutinib and acalabrutinib have provided an additional therapeutic option; however, resistance has emerged as a significant issue and presents the need for more detailed studies of resistance mechanisms. Recent clinical trials have provided new perspectives on the relative efficacy and safety of various approaches for both transplant-eligible and transplant-ineligible patients. Multiple novel strategies are being evaluated in the treatment of MCL, including both targeted agents and cellular immunotherapies. At the Lymphoma Research Foundation's 13th MCL Workshop, researchers gathered to discuss research findings, clinical trial results, and future directions related to MCL, its biology, and its treatment. This report, which includes a summary of each presentation, aims to review recent findings in MCL research and highlight potential areas for future study.
Collapse
Affiliation(s)
- Brad S Kahl
- a Washington University School of Medicine , St. Louis , MO , USA
| | - Martin Dreyling
- b Department of Medicine III , University Hospital, LMU Munich , Munchen , Germany
| | - Leo I Gordon
- c Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - Peter Martin
- d Weill Cornell Medicine Division of Hematology-Oncology , New York , NY , USA
| | | | | |
Collapse
|
171
|
Lampasona AA, Czaplinski K. Hnrnpab regulates neural cell motility through transcription of Eps8. RNA (NEW YORK, N.Y.) 2019; 25:45-59. [PMID: 30314980 PMCID: PMC6298563 DOI: 10.1261/rna.067413.118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/01/2018] [Indexed: 05/05/2023]
Abstract
Cell migration requires a complicated network of structural and regulatory proteins. Changes in cellular motility can impact migration as a result of cell-type or developmental stage regulated expression of critical motility genes. Hnrnpab is a conserved RNA-binding protein found as two isoforms produced by alternative splicing. Its expression is enriched in the subventricular zone (SVZ) and the rostral migratory stream within the brain, suggesting possible support of the migration of neural progenitor cells in this region. Here we show that the migration of cells from the SVZ of developing Hnrnpab-/- mouse brains is impaired. An RNA-seq analysis to identify Hnrnpab-dependent cell motility genes led us to Eps8, and in agreement with the change in cell motility, we show that Eps8 is decreased in Hnrnpab-/- SVZ tissue. We scrutinized the motility of Hnrnpab-/- cells and confirmed that the decreases in both cell motility and Eps8 are restored by ectopically coexpressing both alternatively spliced Hnrnpab isoforms, therefore these variants are surprisingly nonredundant for cell motility. Our results support a model where both Hnrnpab isoforms work in concert to regulate Eps8 transcription in the mouse SVZ to promote the normal migration of neural cells during CNS development.
Collapse
Affiliation(s)
- Alexa A Lampasona
- Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York 11749, USA
- Centers for Molecular Medicine, Stony Brook University, Stony Brook, New York 11749, USA
| | - Kevin Czaplinski
- Centers for Molecular Medicine, Stony Brook University, Stony Brook, New York 11749, USA
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York 11749, USA
| |
Collapse
|
172
|
Weissenrieder JS, Reilly JE, Neighbors JD, Hohl RJ. Inhibiting geranylgeranyl diphosphate synthesis reduces nuclear androgen receptor signaling and neuroendocrine differentiation in prostate cancer cell models. Prostate 2019; 79:21-30. [PMID: 30106164 DOI: 10.1002/pros.23707] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/23/2018] [Indexed: 11/09/2022]
Abstract
BACKGROUND Following androgen deprivation for the treatment of advanced adenocarcinoma of the prostate, tumors can progress to neuroendocrine prostate cancer (NEPC). This transdifferentiation process is poorly understood, but trafficking of transcriptional factors and/or cytoskeletal rearrangements may be involved. We observed the role of geranylgeranylation in this process by treatment with digeranyl bisphosphonate (DGBP), a selective inhibitor of geranylgeranyl pyrophosphate synthase which blocks the prenylation of small GTPases such as Rho and Rab family proteins, including Cdc42 and Rac1. METHODS We examined the therapeutic potential of DGBP in LNCaP, C4-2B4, and 22Rv1 cell culture models. Cell morphology and protein expression were quantified to observe the development of the neuroendocrine phenotype in androgen-deprivation and abiraterone-treated LNCaP models of NEPC development. Luciferase reporter assays were utilized to examine AR activity, and immunofluorescence visualized the localization of AR within the cell. RESULTS Essential genes in the isoprenoid pathway, such as HMGCR, MVK, GGPS1, and GGT1, were highly expressed in a subset of castration resistant prostate cancers reported by Beltran et al. Under treatment with DGBP, nuclear localization of AR decreased in LNCaP, 22Rv1, and C4-2B4 cell lines, luciferase reporter activity was reduced in LNCaP and 22Rv1, and AR target gene transcription also decreased in LNCaP. Conversely, nuclear localization of AR was enhanced by the addition of GGOH. Finally, induction of the NEPC structural and molecular phenotype via androgen deprivation in LNCaP cells was inhibited by DGBP in a GGOH-dependent manner. CONCLUSIONS DGBP is a novel compound with the potential to reduce AR transcriptional activity and inhibit PCa progression to NEPC phenotype. These results suggest that DGBP may be used to block cell growth and metastasis in both hormone therapy sensitive and resistant paradigms.
Collapse
Affiliation(s)
- Jillian S Weissenrieder
- Departments of Medicine and Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania
| | | | - Jeffrey D Neighbors
- Department of Pharmacology and Medicine Penn State College of Medicine, Hershey, Pennsylvania
| | - Raymond J Hohl
- Departments of Medicine and Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
173
|
Xiu Y, Zhang H, Wang S, Gan T, Wei M, Zhou S, Chen S. cDNA cloning, characterization, and expression analysis of the Rac1 and Rac2 genes from Cynoglossus semilaevis. FISH & SHELLFISH IMMUNOLOGY 2019; 84:998-1006. [PMID: 30399403 DOI: 10.1016/j.fsi.2018.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/26/2018] [Accepted: 11/01/2018] [Indexed: 06/08/2023]
Abstract
Rac1 and Rac2, belonging to the small Rho GTPase family, play an important role during the immune responses. In this study, a Rac1 homolog (CsRac1) and a Rac2 homolog (CsRac2) were cloned from the Cynoglossus semilaevis. The full-length of CsRac1 and CsRac2 cDNA was 1219 bp and 1047 bp, respectively. Both CsRac1 and CsRac2 contain a 579 bp open reading frame (ORF) which encoding a 192 amino acids putative protein. The predicted molecular weight of CsRac1 and CsRac2 was 21.41 kDa and 21.35 kDa, and their theoretical pI was 8.50 and 7.91, respectively. Sequence analysis showed that the conserved RHO domain was detected both from amino acid of CsRac1 and CsRac2. Homologous analysis showed that CsRac1 and CsRac2 share high conservation with other counterparts from different species. The CsRac1 and CsRac2 transcript showed wide tissue distribution, in which CsRac1 and CsRac2 exhibit the highest expression level in liver and gill, respectively. The expression level of CsRac1 and CsRac2 fluctuated in the liver and gill tissues at different time points after challenged by Vibrio harveyi. Specifically, CsRac1 and CsRac2 were significantly up-regulated at 48 h and 96 h post injection. Moreover, the knocking down of CsRac1 and CsRac2 in cell line (TSHKC) reduced the expression of CsPAK1, CsIL1-β and CsTNF-α. The present data suggests that CsRac1 and CsRac2 might play important roles in the innate immunity of half-smooth tongue sole.
Collapse
Affiliation(s)
- Yunji Xiu
- Marine Science and Engineering College, Qingdao Agricultural University, Qingdao, 266109, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology Yellow Sea Fisheries Research Institute, CAFS, Qingdao, 266071, China; Key Lab of Sustainable Development of Marine Fisheries, Ministry of Agriculture, Qingdao, 266071, China
| | - Hongxiang Zhang
- Marine Science and Engineering College, Qingdao Agricultural University, Qingdao, 266109, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology Yellow Sea Fisheries Research Institute, CAFS, Qingdao, 266071, China; Key Lab of Sustainable Development of Marine Fisheries, Ministry of Agriculture, Qingdao, 266071, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Shuangyan Wang
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology Yellow Sea Fisheries Research Institute, CAFS, Qingdao, 266071, China; Key Lab of Sustainable Development of Marine Fisheries, Ministry of Agriculture, Qingdao, 266071, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Tian Gan
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology Yellow Sea Fisheries Research Institute, CAFS, Qingdao, 266071, China; Key Lab of Sustainable Development of Marine Fisheries, Ministry of Agriculture, Qingdao, 266071, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Min Wei
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology Yellow Sea Fisheries Research Institute, CAFS, Qingdao, 266071, China; Key Lab of Sustainable Development of Marine Fisheries, Ministry of Agriculture, Qingdao, 266071, China
| | - Shun Zhou
- Marine Science and Engineering College, Qingdao Agricultural University, Qingdao, 266109, China
| | - Songlin Chen
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology Yellow Sea Fisheries Research Institute, CAFS, Qingdao, 266071, China; Key Lab of Sustainable Development of Marine Fisheries, Ministry of Agriculture, Qingdao, 266071, China.
| |
Collapse
|
174
|
Lao M, Zhan Z, Li N, Xu S, Shi M, Zou Y, Huang M, Zeng S, Liang L, Xu H. Role of small ubiquitin-like modifier proteins-1 (SUMO-1) in regulating migration and invasion of fibroblast-like synoviocytes from patients with rheumatoid arthritis. Exp Cell Res 2018; 375:52-61. [PMID: 30562482 DOI: 10.1016/j.yexcr.2018.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/11/2018] [Accepted: 12/13/2018] [Indexed: 11/28/2022]
Abstract
Rheumatoid arthritis (RA) is featured by erosive cartilage and bone destruction. The enhancing aggressive property of fibroblast-like synoviocytes (FLSs) plays a critical role in this process. Small ubiquitin-like modifier (SUMO) proteins, including SUMO-1, SUMO-2, SUMO-3 and SUMO-4, participate in regulating many cellular events such as survival, migration and signal transduction in some cell lines. However, their roles in the pathogenesis of RA are not well established. Therefore, we evaluated the role of SUMO proteins in RA FLSs migration and invasion. We found that expression of both SUMO-1 and SUMO-2 was elevated in FLSs and synovial tissues (STs) from patients with RA. SUMO-1 suppression by small interference RNA (siRNA) reduced migration and invasion as well as MMP-1 and MMP-3 expression in RA FLSs. We also demonstrated that SUMO-1 regulated lamellipodium formation during cell migration. To explore further into molecular mechanisms, we evaluated the effect of SUMO-1 knockdown on the activation of Rac1/PAK1, a critical signaling pathway that controls cell motility. Our results indicated that SUMO-1-mediated SUMOylation controlled Rac1 activation and modulated downstream PAK1 activity. Inhibition of Rac1 or PAK1 also decreased migration and invasion of RA FLSs. Our findings suggest that SUMO-1 suppression could be protective against joint destruction in RA by inhibiting aggressive behavior of RA FLSs.
Collapse
Affiliation(s)
- Minxi Lao
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhongping Zhan
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Nan Li
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Siqi Xu
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Maohua Shi
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Rheumatology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Yaoyao Zou
- Department of Rheumatology, The Second Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mingcheng Huang
- Department of Hematology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shan Zeng
- Department of Rheumatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Liuqin Liang
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hanshi Xu
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
175
|
Abdrabou A, Wang Z. Post-Translational Modification and Subcellular Distribution of Rac1: An Update. Cells 2018; 7:cells7120263. [PMID: 30544910 PMCID: PMC6316090 DOI: 10.3390/cells7120263] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/06/2018] [Accepted: 12/10/2018] [Indexed: 12/27/2022] Open
Abstract
Rac1 is a small GTPase that belongs to the Rho family. The Rho family of small GTPases is a subfamily of the Ras superfamily. The Rho family of GTPases mediate a plethora of cellular effects, including regulation of cytoarchitecture, cell size, cell adhesion, cell polarity, cell motility, proliferation, apoptosis/survival, and membrane trafficking. The cycling of Rac1 between the GTP (guanosine triphosphate)- and GDP (guanosine diphosphate)-bound states is essential for effective signal flow to elicit downstream biological functions. The cycle between inactive and active forms is controlled by three classes of regulatory proteins: Guanine nucleotide exchange factors (GEFs), GTPase-activating proteins (GAPs), and guanine-nucleotide-dissociation inhibitors (GDIs). Other modifications include RNA splicing and microRNAs; various post-translational modifications have also been shown to regulate the activity and function of Rac1. The reported post-translational modifications include lipidation, ubiquitination, phosphorylation, and adenylylation, which have all been shown to play important roles in the regulation of Rac1 and other Rho GTPases. Moreover, the Rac1 activity and function are regulated by its subcellular distribution and translocation. This review focused on the most recent progress in Rac1 research, especially in the area of post-translational modification and subcellular distribution and translocation.
Collapse
Affiliation(s)
- Abdalla Abdrabou
- Department of Medical Genetics, and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Zhixiang Wang
- Department of Medical Genetics, and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
176
|
Li Y, Huo S, Fang Y, Zou T, Gu X, Tao Q, Xu H. ROCK Inhibitor Y27632 Induced Morphological Shift and Enhanced Neurite Outgrowth-Promoting Property of Olfactory Ensheathing Cells via YAP-Dependent Up-Regulation of L1-CAM. Front Cell Neurosci 2018; 12:489. [PMID: 30618636 PMCID: PMC6297255 DOI: 10.3389/fncel.2018.00489] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/29/2018] [Indexed: 01/14/2023] Open
Abstract
Olfactory ensheathing cells (OECs) are heterogeneous in morphology, antigenic profiles and functions, and these OEC subpopulations have shown different outcomes following OEC transplantation for central nervous system (CNS) injuries. Morphologically, OECs are divided into two subpopulations, process-bearing (Schwann cells-like) and flattened (astrocytes-like) OECs, which could switch between each other and are affected by extracellular and intracellular factors. However, neither the relationship between the morphology and function of OECs nor their molecular mechanisms have been clarified. In the present study, we first investigated morphological and functional differences of OECs under different cytokine exposure conditions. It demonstrated that OECs mainly displayed a process-bearing shape under pro-inflammatory conditions (lipopolysaccharide, LPS), while they displayed a flattened shape under anti-inflammatory conditions [interleukin-4 (IL-4) and transforming growth factor-β1 (TGF-β1)]. The morphological changes were partially reversible and the Rho-associated coiled-coil-containing protein kinase (ROCK)/F-actin pathway was involved. Functionally, process-bearing OECs under pro-inflammatory conditions showed increased cellular metabolic activity and a higher migratory rate when compared with flattened OECs under anti-inflammatory conditions and significantly promoted neurite outgrowth and extension. Remarkably, the morphological shift towards process-bearing OECs induced by ROCK inhibitor Y27632 enhanced the neurite outgrowth-promoting property of OECs. Furthermore, as the downstream of the ROCK pathway, transcriptional co-activator Yes-associated protein (YAP) mediated morphological shift and enhanced the neurite outgrowth-promoting property of OECs through upregulating the expression of the neural adhesion molecule L1-CAM. Our data provided evidence that OECs with specific shapes correspond to specific functional phenotypes and opened new insights into the potential combination of OECs and small-molecule ROCK inhibitors for the regeneration of CNS injuries.
Collapse
Affiliation(s)
- Yijian Li
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Shujia Huo
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Yajie Fang
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Ting Zou
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Xianliang Gu
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Qin Tao
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Haiwei Xu
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| |
Collapse
|
177
|
de Beco S, Vaidžiulytė K, Manzi J, Dalier F, di Federico F, Cornilleau G, Dahan M, Coppey M. Optogenetic dissection of Rac1 and Cdc42 gradient shaping. Nat Commun 2018; 9:4816. [PMID: 30446664 PMCID: PMC6240110 DOI: 10.1038/s41467-018-07286-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 10/19/2018] [Indexed: 12/22/2022] Open
Abstract
During cell migration, Rho GTPases spontaneously form spatial gradients that define the front and back of cells. At the front, active Cdc42 forms a steep gradient whereas active Rac1 forms a more extended pattern peaking a few microns away. What are the mechanisms shaping these gradients, and what is the functional role of the shape of these gradients? Here we report, using a combination of optogenetics and micropatterning, that Cdc42 and Rac1 gradients are set by spatial patterns of activators and deactivators and not directly by transport mechanisms. Cdc42 simply follows the distribution of Guanine nucleotide Exchange Factors, whereas Rac1 shaping requires the activity of a GTPase-Activating Protein, β2-chimaerin, which is sharply localized at the tip of the cell through feedbacks from Cdc42 and Rac1. Functionally, the spatial extent of Rho GTPases gradients governs cell migration, a sharp Cdc42 gradient maximizes directionality while an extended Rac1 gradient controls the speed. A steep gradient of Cdc42 is at the front of migrating cells, whereas the active Rac1 gradient is graded. Here the authors show that Cdc42 gradients follow the distribution of GEFs and govern direction of migration, while Rac1 gradients require the activity of the GAP β2-chimaerin and control cell speed.
Collapse
Affiliation(s)
- S de Beco
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, Sorbonne Université, CNRS, 75005, Paris, France
| | - K Vaidžiulytė
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, Sorbonne Université, CNRS, 75005, Paris, France
| | - J Manzi
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, Sorbonne Université, CNRS, 75005, Paris, France
| | - F Dalier
- PASTEUR, Département de chimie, École normale supérieure, CNRS UMR 8640, PSL Research University, Sorbonne Université, 75005, Paris, France
| | - F di Federico
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, Sorbonne Université, CNRS, 75005, Paris, France
| | - G Cornilleau
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, Sorbonne Université, CNRS, 75005, Paris, France
| | - M Dahan
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, Sorbonne Université, CNRS, 75005, Paris, France
| | - M Coppey
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, Sorbonne Université, CNRS, 75005, Paris, France.
| |
Collapse
|
178
|
Huang K, Wu LD. Dehydroepiandrosterone: Molecular mechanisms and therapeutic implications in osteoarthritis. J Steroid Biochem Mol Biol 2018; 183:27-38. [PMID: 29787833 DOI: 10.1016/j.jsbmb.2018.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/26/2018] [Accepted: 05/17/2018] [Indexed: 12/25/2022]
Abstract
Dehydroepiandrosterone (DHEA), a 19-carbon steroid hormone primarily synthesized in the adrenal gland, exerts a chondroprotective effect against osteoarthritis (OA) and has been considered an effective candidate of disease-modifying OA drugs (DMOADs) that slow disease progression. We and others previously demonstrated that DHEA exerted a beneficial effect on osteoarthritic cartilage by positively modulating the balance between anabolic and catabolic factors (e.g., MMPs/TIMP-1, ADAMTS/TIMP-3 and cysteine proteinases/cystatin C), inhibiting catabolic signaling pathways (e.g., Wnt/β-catenin), and suppressing proinflammatory cytokines-mediated low-grade synovial inflammation (e.g., IL-1β). However, the full picture of the pharmacological molecular mechanism(s) underlying the activity of DHEA against OA is still incomplete, and a comprehensive and up-to-date review article in this field is unavailable. In this review, recent findings (apart from the well documented pathogenesis of OA) regarding disease-related mechanisms involving low grade synovial inflammation, cartilage matrix stiffness, chondrocyte autophagy and the roles of a variety of catabolic cellular signaling pathways are discussed. Moreover, the possible relationship between these disease-related mechanisms and DHEA action is discussed. Emerging evidence from in vivo and in vitro studies were scrutinized and are concisely presented to demonstrate the investigational and putative mechanisms underlying the anti-OA potential of DHEA.
Collapse
Affiliation(s)
- Kai Huang
- Department of Orthopedic Surgery, Tongde Hospital of Zhejiang Province, China.
| | - Li-Dong Wu
- Department of Orthopedic Surgery, The Second Hospital of Medical College, Zhejiang University, China
| |
Collapse
|
179
|
Development of pipette tip gap closure migration assay (s-ARU method) for studying semi-adherent cell lines. Cytotechnology 2018; 70:1685-1695. [PMID: 30069611 DOI: 10.1007/s10616-018-0245-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 07/25/2018] [Indexed: 10/28/2022] Open
Abstract
This work presents a pipette tip gap closure migration assay prototype tool (semi-adherent relative upsurge-s-ARU-method) to study cell migration or wound healing in semi-adherent cell lines, such as lymph node carcinoma of the prostate (LNCaP). Basically, it consists of a 6-well cover plate modification, where pipette tips with the filter are shortened and fixed vertically to the inner surface of the cover plate, with their heights adjusted to touch the bottom of the well center. This provides a barrier for the inoculated cells to grow on, creating a cell-free gap. Such a uniform gap formed can be used to study migration assay for both adherent as well as semi-adherent cells. After performing time studies, effective measurement of gap area can be carried out conveniently through image analysis software. Here, the prototype was tested for LNCaP cells, treated with testosterone and flutamide as well as with bacteriophages T4 and M13. A scratch assay using PC3 adherent cells was also performed for comparison. It was observed that s-ARU method is suitable for studying LNCaP cells migration assay, as observed from our results with testosterone, flutamide, and bacteriophages (T4 and M13). Our method is a low-cost handmade prototype, which can be an alternative to the other migration assay protocol(s) for both adherent and semi-adherent cell cultures in oncological research along with other biological research applications.
Collapse
|
180
|
Tian Z, Lv X, Zhang M, Wang X, Chen Y, Tang P, Xu P, Zhang L, Wu B, Zhang L. Deletion of epithelial cell-specific Cdc42 leads to enamel hypermaturation in a conditional knockout mouse model. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2623-2632. [DOI: 10.1016/j.bbadis.2018.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/02/2018] [Accepted: 04/16/2018] [Indexed: 12/01/2022]
|
181
|
Wei WS, Chen X, Guo LY, Li XD, Deng MH, Yuan GJ, He LY, Li YH, Zhang ZL, Jiang LJ, Chen RX, Ma XD, Wei S, Ma NF, Liu ZW, Luo JH, Zhou FJ, Xie D. TRIM65 supports bladder urothelial carcinoma cell aggressiveness by promoting ANXA2 ubiquitination and degradation. Cancer Lett 2018; 435:10-22. [PMID: 30075204 DOI: 10.1016/j.canlet.2018.07.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/09/2018] [Accepted: 07/26/2018] [Indexed: 12/23/2022]
Abstract
Clinically, most of human urothelial carcinoma of the bladder (UCB)-related deaths result from tumor metastasis, but the underlying molecular mechanisms are largely unknown. Recently, a growing number of tripartite motif (TRIM) family members have been suggested to be important regulators for tumorigenesis. However, the impact of most TRIM members on UCB pathogenesis is unclear. In this study, TRIM65 was first screened as an important oncogenic factor of UCB from the Cancer Genome Atlas (TCGA) database and was validated by a large cohort of clinical UCB tissues. By in vitro and in vivo experiments, we demonstrated that TRIM65 promotes UCB cell invasive and metastatic capacities. Notably, we showed that TRIM65 modulates cytoskeleton rearrangement and induces UCB cells epithelial-mesenchymal transition by the ubiquitination of ANXA2, ultimately leading to an enhanced invasiveness of UCB cells. Importantly, UCBs with high expression of TRIM65 and low expression of ANXA2 showed the poorest outcome. Collectively, our results suggest that the overexpression of TRIM65 has an essential oncogenic role via ubiquitination of ANXA2 in UCB pathogenesis, and that such could be used as a novel prognostic marker and/or therapeutic target for UCB.
Collapse
Affiliation(s)
- Wen-Su Wei
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China
| | - Xin Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China
| | - Li-Yi Guo
- Department of Oncology, The Sixth People's Hospital of Huizhou, Huiyang, Guangdong, China
| | - Xiang-Dong Li
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China
| | - Ming-Hui Deng
- Department of Oncology, The Sixth People's Hospital of Huizhou, Huiyang, Guangdong, China
| | - Gang-Jun Yuan
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China
| | - Le-Ye He
- Department of Urology, Xiangya Third Hospital, No. 106, 2nd Zhongshan Road, Changsha, China
| | - Yong-Hong Li
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China
| | - Zhi-Lin Zhang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China
| | - Li-Juan Jiang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China
| | - Ri-Xin Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China
| | - Xiao-Dan Ma
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China
| | - Shi Wei
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China; Department of Urology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ning-Fang Ma
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhuo-Wei Liu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China
| | - Jun-Hang Luo
- Department of Urology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fang-Jian Zhou
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China; Department of Urology, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China.
| | - Dan Xie
- State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, China; Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
182
|
Curcumin analogue 1,5-bis(4-hydroxy-3-((4-methylpiperazin-1-yl)methyl)phenyl)penta-1,4-dien-3-one mediates growth arrest and apoptosis by targeting the PI3K/AKT/mTOR and PKC-theta signaling pathways in human breast carcinoma cells. Bioorg Chem 2018. [DOI: 10.1016/j.bioorg.2018.03.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
183
|
Ogienko AA, Yarinich LA, Fedorova EV, Lebedev MO, Andreyeva EN, Pindyurin AV, Baricheva EM. New slbo-Gal4 driver lines for the analysis of border cell migration during Drosophila oogenesis. Chromosoma 2018; 127:475-487. [DOI: 10.1007/s00412-018-0676-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 06/09/2018] [Accepted: 06/28/2018] [Indexed: 12/23/2022]
|
184
|
Li W, Yu X, Ma X, Xie L, Xia Z, Liu L, Yu X, Wang J, Zhou H, Zhou X, Yang Y, Liu H. Deguelin attenuates non-small cell lung cancer cell metastasis through inhibiting the CtsZ/FAK signaling pathway. Cell Signal 2018; 50:131-141. [PMID: 30018008 DOI: 10.1016/j.cellsig.2018.07.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/09/2018] [Accepted: 07/05/2018] [Indexed: 01/13/2023]
Abstract
Lung cancer is the leading cause of cancer-related death among both men and women every year, mainly due to metastasis. Although natural compound deguelin has been reported to inhibited cell migration and invasion in various cancer cells, the details of this regulation progress remain to be fully elucidated. In this study, we investigated the underlying mechanism of deguelin-suppressed metastasis of non-small cell lung cancer (NSCLC) cells. Our results demonstrate that deguelin inhibits NSCLC cell migration, invasion, and metastasis both in vitro and in vivo. These inhibitory effects of deguelin were mediated by suppressing of Cathepsin Z (CtsZ) expression and interrupting the interaction of CtsZ with integrin β3. Moreover, deguelin inhibits the activation of CtsZ downstream FAK/Src/Paxillin signaling. Knockdown of CtsZ mimicked the effect of deguelin on NSCLC cells migration and invasion. Our study reveals that deguelin exerts its anti-metastatic effect both in vitro and in vivo is partly dependent on the suppression of CtsZ signaling. Deguelin would be a potential anti-metastasis agent against NSCLC.
Collapse
Affiliation(s)
- Wei Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Xinfang Yu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Xiaolong Ma
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Li Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhenkun Xia
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Lijun Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xinyou Yu
- Shangdong Lvdu Bio-Industry Co., Ltd., Binzhou, Shangdong 256600, China
| | - Jian Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Huiling Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xinmin Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Haidan Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
185
|
Tang K, Boudreau CG, Brown CM, Khadra A. Paxillin phosphorylation at serine 273 and its effects on Rac, Rho and adhesion dynamics. PLoS Comput Biol 2018; 14:e1006303. [PMID: 29975690 PMCID: PMC6053249 DOI: 10.1371/journal.pcbi.1006303] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/19/2018] [Accepted: 06/13/2018] [Indexed: 12/16/2022] Open
Abstract
Focal adhesions are protein complexes that anchor cells to the extracellular matrix. During migration, the growth and disassembly of these structures are spatiotemporally regulated, with new adhesions forming at the leading edge of the cell and mature adhesions disassembling at the rear. Signalling proteins and structural cytoskeletal components tightly regulate adhesion dynamics. Paxillin, an adaptor protein within adhesions, is one of these proteins. Its phosphorylation at serine 273 (S273) is crucial for maintaining fast adhesion assembly and disassembly. Paxillin is known to bind to a GIT1-βPIX-PAK1 complex, which increases the local activation of the small GTPase Rac. To understand quantitatively the behaviour of this system and how it relates to adhesion assembly/disassembly, we developed a mathematical model describing the dynamics of the small GTPases Rac and Rho as determined by paxillin S273 phosphorylation. Our model revealed that the system possesses bistability, where switching between uninduced (active Rho) and induced (active Rac) states can occur through a change in rate of paxillin phosphorylation or PAK1 activation. The bistable switch is characterized by the presence of memory, minimal change in the levels of active Rac and Rho within the induced and uninduced states, respectively, and the limited regime of monostability associated with the uninduced state. These results were validated experimentally by showing the presence of bimodality in adhesion assembly and disassembly rates, and demonstrating that Rac activity increases after treating Chinese Hamster Ovary cells with okadaic acid (a paxillin phosphatase inhibitor), followed by a modest recovery after 20 min washout. Spatial gradients of phosphorylated paxillin in a reaction-diffusion model gave rise to distinct regions of Rac and Rho activities, resembling polarization of a cell into front and rear. Perturbing several parameters of the model also revealed important insights into how signalling components upstream and downstream of paxillin phosphorylation affect dynamics. Cellular migration is crucial in both physiological and pathological functions. Maintenance of proper migration and development of aberrant migration are effectuated by cellular machinery involving protein complexes, called adhesions, that anchor the cell to its environment. Over time, these adhesions assemble at the leading edge, as the cell extends forward, anchoring the front of the cells to its substrate, while those at the cell rear disassemble, allowing detachment and forward movement. Their dynamics are controlled by a number of regulatory factors, occurring on both cell-wide and adhesion-level scales. The coordination of these regulatory factors is complex, but insights about their dynamics can be gained from the use of mathematical modeling techniques which integrate many of these components together. Here, we developed several molecularly explicit models to explore how local regulation of paxillin, an adhesion protein, interacts with the activities of Rac and Rho to produce cell-wide polarization associated with motility and directionality. By altering paxillin phosphorylation/dephosphorylation within such models, we have advanced our understanding of how a shift from a non-motile state to a highly motile state occurs. Deciphering these key processes quantitatively thus helped us gain insight into the subcellular factors underlying polarity and movement.
Collapse
Affiliation(s)
- Kaixi Tang
- Department of Physiology, McGill University, Montreal, Québec, Canada
| | | | - Claire M. Brown
- Department of Physiology, McGill University, Montreal, Québec, Canada
- Advanced BioImaging Facility (ABIF), McGill University, Montreal, Québec, Canada
- Cell Information Systems, McGill University, Montreal, Québec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
| | - Anmar Khadra
- Department of Physiology, McGill University, Montreal, Québec, Canada
- * E-mail:
| |
Collapse
|
186
|
Wang J, Li H, Yao Y, Ren Y, Lin J, Hu J, Zheng M, Song X, Zhao T, Chen YY, Shen Y, Zhu YJ, Wang LL. β-Elemene Enhances GAP-43 Expression and Neurite Outgrowth by Inhibiting RhoA Kinase Activation in Rats with Spinal Cord Injury. Neuroscience 2018; 383:12-21. [DOI: 10.1016/j.neuroscience.2018.04.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 04/03/2018] [Accepted: 04/28/2018] [Indexed: 12/21/2022]
|
187
|
Integrin α5 down-regulation by miR-205 suppresses triple negative breast cancer stemness and metastasis by inhibiting the Src/Vav2/Rac1 pathway. Cancer Lett 2018; 433:199-209. [PMID: 29964204 DOI: 10.1016/j.canlet.2018.06.037] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 06/21/2018] [Accepted: 06/26/2018] [Indexed: 01/06/2023]
Abstract
Triple negative breast cancer (TNBC) usually displays more aggressive metastasis, the underlying mechanism is unclear. Previous studies showed that microRNA-205 (miR-205) has controversial roles in cancer, however, its role in TNBC metastasis and the underlying mechanism have not been well-understood. In this study we found that miR-205 expression level is extremely low in basal mesenchymal-like highly migratory and invasive TNBC cells. Stably re-expressing miR-205 in TNBC cells significantly reduced their migration, invasion capability and cancer stem cell (CSC)-like property. Nude mouse orthotopic mammary xenograft tumor model study revealed that miR-205 re-expression greatly decreases TNBC tumor growth and abolishes spontaneous lung metastasis. Mechanistic studies demonstrated that miR-205 inhibits TNBC cell metastatic traits and tumor metastasis by down-regulating integrin α5 (ITGA5). Moreover, ITGA5 knockout using the CRISPR/Cas9 technique achieved the same strong inhibitory effect on TNBC cell CSC-like property and tumor metastasis as re-expressing miR-205 did. Further mechanistic studies indicated that ITGA5 down-regulation by miR-205 re-expression impairs TNBC cell metastatic traits by inhibiting the Src/Vav2/Rac1 pathway. Together, our findings suggest that miR-205 and ITGA5 may serve as potential targets for developing effective therapies for metastatic TNBC.
Collapse
|
188
|
Zhai W, Ma Z, Wang W, Song L, Yi J. Paeoniflorin inhibits Rho kinase activation in joint synovial tissues of rats with collagen-induced rheumatoid arthritis. Biomed Pharmacother 2018; 106:255-259. [PMID: 29966968 DOI: 10.1016/j.biopha.2018.06.130] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 06/15/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023] Open
Abstract
Paeoniflorin (PF) has many effects, such as anti-inflammation, immune-regulation, abirritation, and so on. However, the protective mechanisms of PF on rheumatoid arthritis (RA) was not completely known. Thus, we explored deeply the protective mechanisms in a collagen-induced RA (CIA) rat model. CIA was induced in rats by intradermal injection of bovine type II collagen in complete Freund's adjuvant. Later, the CIA rats received oral administration of PF (50 and 100 mg/kg) once a day from the day 21, with the treatment lasting for 14 days. A variety of indicators were measured for evaluation of anti-rheumatism effect, including paw swelling, arthritis scores, and histopathological changes. And the contents of pro-inflammatory cytokines, including tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6) in the serum, as well as p-NF-κB p65 and p-MYPT1 in the joint synovial tissues were detected to explore the possible mechanisms. The results demonstrated that PF treatment significantly ameliorated the symptoms in CIA rats, reduced the levels of pro-inflammatory cytokines and paw swelling, down-regulated the expressions of p-NF-κB p65 and p-MYPT1. The present results revealed that PF could effectively improve collagen-induced RA in rats by inhibiting Rho kinase activation in the joint synovial tissues, in turn down-regulating expression of p-NF-κB p65 and reducing contents of pro-inflammatory cytokines. Moreover, PF may be an effective agent for RA.
Collapse
Affiliation(s)
- Wenjing Zhai
- College of Chemical and Biological Engineering, Yichun University, Yichun 336000, China
| | - Zhihong Ma
- Department of Immunology and Pathobiology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Weijie Wang
- Department of Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Lin Song
- Department of Immunology and Pathobiology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Jianfeng Yi
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun 336000, China; The Center for Translational Medicine, Yichun University, Yichun 336000, China.
| |
Collapse
|
189
|
Tantillo E, Colistra A, Vannini E, Cerri C, Pancrazi L, Baroncelli L, Costa M, Caleo M. Bacterial Toxins and Targeted Brain Therapy: New Insights from Cytotoxic Necrotizing Factor 1 (CNF1). Int J Mol Sci 2018; 19:ijms19061632. [PMID: 29857515 PMCID: PMC6032336 DOI: 10.3390/ijms19061632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/07/2018] [Accepted: 05/12/2018] [Indexed: 01/17/2023] Open
Abstract
Pathogenic bacteria produce toxins to promote host invasion and, therefore, their survival. The extreme potency and specificity of these toxins confer to this category of proteins an exceptionally strong potential for therapeutic exploitation. In this review, we deal with cytotoxic necrotizing factor (CNF1), a cytotoxin produced by Escherichia coli affecting fundamental cellular processes, including cytoskeletal dynamics, cell cycle progression, transcriptional regulation, cell survival and migration. First, we provide an overview of the mechanisms of action of CNF1 in target cells. Next, we focus on the potential use of CNF1 as a pharmacological treatment in central nervous system’s diseases. CNF1 appears to impact neuronal morphology, physiology, and plasticity and displays an antineoplastic activity on brain tumors. The ability to preserve neural functionality and, at the same time, to trigger senescence and death of proliferating glioma cells, makes CNF1 an encouraging new strategy for the treatment of brain tumors.
Collapse
Affiliation(s)
- Elena Tantillo
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
- Fondazione Pisana per la Scienza Onlus (FPS), via Ferruccio Giovannini 13, San Giuliano Terme, 56017 Pisa, Italy.
| | - Antonella Colistra
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
- Departement of Biology, University of Pisa, via Luca Ghini 13, 56126 Pisa, Italy.
| | - Eleonora Vannini
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Chiara Cerri
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
- Fondazione Umberto Veronesi, Piazza Velasca 5, 20122 Milano, Italy.
| | - Laura Pancrazi
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Laura Baroncelli
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Mario Costa
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Matteo Caleo
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
190
|
Bharadwaj R, Sharma S, Arya R, Bhattacharya S, Bhattacharya A. EhRho1 regulates phagocytosis by modulating actin dynamics through EhFormin1 and EhProfilin1 inEntamoeba histolytica. Cell Microbiol 2018; 20:e12851. [DOI: 10.1111/cmi.12851] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/12/2018] [Accepted: 03/25/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Ravi Bharadwaj
- School of Life Sciences; Jawaharlal Nehru University; New Delhi India
| | - Shalini Sharma
- School of Life Sciences; Jawaharlal Nehru University; New Delhi India
| | - Ranjana Arya
- School of Biotechnology; Jawaharlal Nehru University; New Delhi India
| | - Sudha Bhattacharya
- School of Environmental Sciences; Jawaharlal Nehru University; New Delhi India
| | - Alok Bhattacharya
- School of Life Sciences; Jawaharlal Nehru University; New Delhi India
| | | |
Collapse
|
191
|
Lim J, Choi A, Kim HW, Yoon H, Park SM, Tsai CHD, Kaneko M, Kim DS. Constrained Adherable Area of Nanotopographic Surfaces Promotes Cell Migration through the Regulation of Focal Adhesion via Focal Adhesion Kinase/Rac1 Activation. ACS APPLIED MATERIALS & INTERFACES 2018; 10:14331-14341. [PMID: 29649358 DOI: 10.1021/acsami.7b18954] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cell migration is crucial in physiological and pathological processes such as embryonic development and wound healing; such migration is strongly guided by the surrounding nanostructured extracellular matrix. Previous studies have extensively studied the cell migration on anisotropic nanotopographic surfaces; however, only a few studies have reported cell migration on isotropic nanotopographic surfaces. We herein, for the first time, propose a novel concept of adherable area on cell migration using isotropic nanopore surfaces with sufficient nanopore depth by adopting a high aspect ratio. As the pore size of the nanopore surface was controlled to 200, 300, and 400 nm in a fixed center-to-center distance of 480 nm, it produced 86, 68, and 36% of adherable area, respectively, on the fabricated surface. A meticulous investigation of the cell migration in response to changes in the constrained adherable area of the nanotopographic surface showed 1.4-, 1.5-, and 1.6-fold increase in migration speeds and a 1.4-, 2-, and 2.5-fold decrease in the number of focal adhesions as the adherable area was decreased to 86, 68, and 36%, respectively. Furthermore, a strong activation of FAK/Rac1 signaling was observed to be involved in the promoted cell migration. These results suggest that the reduced adherable area promotes cell migration through decreasing the FA formation, which in turn upregulates FAK/Rac1 activation. The findings in this study can be utilized to control the cell migration behaviors, which is a powerful tool in the research fields involving cell migration such as promoting wound healing and tissue repair.
Collapse
Affiliation(s)
- Jiwon Lim
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-Ro, Nam-gu , Pohang , Gyeongbuk 37673 , Korea
| | - Andrew Choi
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-Ro, Nam-gu , Pohang , Gyeongbuk 37673 , Korea
| | - Hyung Woo Kim
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-Ro, Nam-gu , Pohang , Gyeongbuk 37673 , Korea
| | - Hyungjun Yoon
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-Ro, Nam-gu , Pohang , Gyeongbuk 37673 , Korea
| | - Sang Min Park
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-Ro, Nam-gu , Pohang , Gyeongbuk 37673 , Korea
| | - Chia-Hung Dylan Tsai
- Department of Mechanical Engineering , Osaka University , 1-1 Yamadaoka , Suita , Osaka 565-0871 , Japan
| | - Makoto Kaneko
- Department of Mechanical Engineering , Osaka University , 1-1 Yamadaoka , Suita , Osaka 565-0871 , Japan
| | - Dong Sung Kim
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-Ro, Nam-gu , Pohang , Gyeongbuk 37673 , Korea
| |
Collapse
|
192
|
Petit AP, Garcia-Petit C, Bueren-Calabuig JA, Vuillard LM, Ferry G, Boutin JA. A structural study of the complex between neuroepithelial cell transforming gene 1 (Net1) and RhoA reveals a potential anticancer drug hot spot. J Biol Chem 2018; 293:9064-9077. [PMID: 29695506 DOI: 10.1074/jbc.ra117.001123] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/18/2018] [Indexed: 12/20/2022] Open
Abstract
The GTPase RhoA is a major player in many different regulatory pathways. RhoA catalyzes GTP hydrolysis, and its catalysis is accelerated when RhoA forms heterodimers with proteins of the guanine nucleotide exchange factor (GEF) family. Neuroepithelial cell transforming gene 1 (Net1) is a RhoA-interacting GEF implicated in cancer, but the structural features supporting the RhoA/Net1 interaction are unknown. Taking advantage of a simple production and purification process, here we solved the structure of a RhoA/Net1 heterodimer with X-ray crystallography at 2-Å resolution. Using a panel of several techniques, including molecular dynamics simulations, we characterized the RhoA/Net1 interface. Moreover, deploying an extremely simple peptide-based scanning approach, we found that short peptides (penta- to nonapeptides) derived from the protein/protein interaction region of RhoA could disrupt the RhoA/Net1 interaction and thereby diminish the rate of nucleotide exchange. The most inhibitory peptide, EVKHF, spanning residues 102-106 in the RhoA sequence, displayed an IC50 of ∼100 μm without further modifications. The peptides identified here could be useful in further investigations of the RhoA/Net1 interaction region. We propose that our structural and functional insights might inform chemical approaches for transforming the pentapeptide into an optimized pseudopeptide that antagonizes Net1-mediated RhoA activation with therapeutic anticancer potential.
Collapse
Affiliation(s)
- Alain-Pierre Petit
- From the Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery and
| | - Christel Garcia-Petit
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom and
| | | | - Laurent M Vuillard
- Pôle d'Expertise Biotechnologie, Chimie, Biologie, Institut de Recherches SERVIER, 78290 Croissy-sur-Seine, France
| | - Gilles Ferry
- Pôle d'Expertise Biotechnologie, Chimie, Biologie, Institut de Recherches SERVIER, 78290 Croissy-sur-Seine, France
| | - Jean A Boutin
- Pôle d'Expertise Biotechnologie, Chimie, Biologie, Institut de Recherches SERVIER, 78290 Croissy-sur-Seine, France
| |
Collapse
|
193
|
Lehners M, Dobrowinski H, Feil S, Feil R. cGMP Signaling and Vascular Smooth Muscle Cell Plasticity. J Cardiovasc Dev Dis 2018; 5:jcdd5020020. [PMID: 29671769 PMCID: PMC6023364 DOI: 10.3390/jcdd5020020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 12/11/2022] Open
Abstract
Cyclic GMP regulates multiple cell types and functions of the cardiovascular system. This review summarizes the effects of cGMP on the growth and survival of vascular smooth muscle cells (VSMCs), which display remarkable phenotypic plasticity during the development of vascular diseases, such as atherosclerosis. Recent studies have shown that VSMCs contribute to the development of atherosclerotic plaques by clonal expansion and transdifferentiation to macrophage-like cells. VSMCs express a variety of cGMP generators and effectors, including NO-sensitive guanylyl cyclase (NO-GC) and cGMP-dependent protein kinase type I (cGKI), respectively. According to the traditional view, cGMP inhibits VSMC proliferation, but this concept has been challenged by recent findings supporting a stimulatory effect of the NO-cGMP-cGKI axis on VSMC growth. Here, we summarize the relevant studies with a focus on VSMC growth regulation by the NO-cGMP-cGKI pathway in cultured VSMCs and mouse models of atherosclerosis, restenosis, and angiogenesis. We discuss potential reasons for inconsistent results, such as the use of genetic versus pharmacological approaches and primary versus subcultured cells. We also explore how modern methods for cGMP imaging and cell tracking could help to improve our understanding of cGMP’s role in vascular plasticity. We present a revised model proposing that cGMP promotes phenotypic switching of contractile VSMCs to VSMC-derived plaque cells in atherosclerotic lesions. Regulation of vascular remodeling by cGMP is not only an interesting new therapeutic strategy, but could also result in side effects of clinically used cGMP-elevating drugs.
Collapse
Affiliation(s)
- Moritz Lehners
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany.
| | - Hyazinth Dobrowinski
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany.
| | - Susanne Feil
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany.
| | - Robert Feil
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
194
|
Tang Y, He Y, Zhang P, Wang J, Fan C, Yang L, Xiong F, Zhang S, Gong Z, Nie S, Liao Q, Li X, Li X, Li Y, Li G, Zeng Z, Xiong W, Guo C. LncRNAs regulate the cytoskeleton and related Rho/ROCK signaling in cancer metastasis. Mol Cancer 2018; 17:77. [PMID: 29618386 PMCID: PMC5885413 DOI: 10.1186/s12943-018-0825-x] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 03/20/2018] [Indexed: 02/08/2023] Open
Abstract
Some of the key steps in cancer metastasis are the migration and invasion of tumor cells; these processes require rearrangement of the cytoskeleton. Actin filaments, microtubules, and intermediate filaments involved in the formation of cytoskeletal structures, such as stress fibers and pseudopodia, promote the invasion and metastasis of tumor cells. Therefore, it is important to explore the mechanisms underlying cytoskeletal regulation. The ras homolog family (Rho) and Rho-associated coiled-coil containing protein serine/threonine kinase (ROCK) signaling pathway is involved in the regulation of the cytoskeleton. Moreover, long noncoding RNAs (lncRNAs) have essential roles in tumor migration and guide gene regulation during cancer progression. LncRNAs can regulate the cytoskeleton directly or may influence the cytoskeleton via Rho/ROCK signaling during tumor migration. In this review, we focus on the regulatory association between lncRNAs and the cytoskeleton and discuss the pathways and mechanisms involved in the regulation of cancer metastasis.
Collapse
Affiliation(s)
- Yanyan Tang
- Department of Colorectal Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yi He
- Department of Colorectal Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Ping Zhang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,School of Electronics and Information Engineering, Hunan University of Science and Engineering, Yongzhou, Hunan, China
| | - Jinpeng Wang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Chunmei Fan
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Liting Yang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Fang Xiong
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shanshan Zhang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaojian Gong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Shaolin Nie
- Department of Colorectal Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- Department of Colorectal Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiayu Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Guiyuan Li
- Department of Colorectal Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- Department of Colorectal Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- Department of Colorectal Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Can Guo
- Department of Colorectal Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
195
|
Pan Y, Jiang S, Hou Q, Qiu D, Shi J, Wang L, Chen Z, Zhang M, Duan A, Qin W, Zen K, Liu Z. Dissection of Glomerular Transcriptional Profile in Patients With Diabetic Nephropathy: SRGAP2a Protects Podocyte Structure and Function. Diabetes 2018; 67:717-730. [PMID: 29242313 DOI: 10.2337/db17-0755] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/26/2017] [Indexed: 01/19/2023]
Abstract
Podocytes play a pivotal role in maintaining glomerular filtration function through their interdigitated foot processes. However, the mechanisms that govern the podocyte cytoskeletal rearrangement remain unclear. Through analyzing the transcriptional profile of renal biopsy specimens from patients with diabetic nephropathy (DN) and control donors, we identify SLIT-ROBO ρGTPase-activating protein 2a (SRGAP2a) as one of the main hub genes strongly associated with proteinuria and glomerular filtration in type 2 DN. Immunofluorescence staining and Western blot analysis revealed that human and mouse SRGAP2a is primarily localized at podocytes and largely colocalized with synaptopodin. Moreover, podocyte SRGAP2a is downregulated in patients with DN and db/db mice at both the mRNA and the protein level. SRGAP2a reduction is observed in cultured podocytes treated with tumor growth factor-β or high concentrations of glucose. Functional and mechanistic studies show that SRGAP2a suppresses podocyte motility through inactivating RhoA/Cdc42 but not Rac1. The protective role of SRGAP2a in podocyte function also is confirmed in zebrafish, in which knockdown of SRGAP2a, a SRGAP2 ortholog in zebrafish, recapitulates podocyte foot process effacement. Finally, increasing podocyte SRGAP2a levels in db/db mice through administration of adenovirus-expressing SRGAP2a significantly mitigates podocyte injury and proteinuria. The results demonstrate that SRGAP2a protects podocytes by suppressing podocyte migration.
Collapse
Affiliation(s)
- Yu Pan
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Song Jiang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Qing Hou
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Dandan Qiu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jingsong Shi
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Ling Wang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Zhaohong Chen
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Aiping Duan
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Weisong Qin
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
196
|
Sotillos S, Aguilar-Aragon M, Hombría JCG. Functional analysis of the Drosophila RhoGAP Cv-c protein and its equivalence to the human DLC3 and DLC1 proteins. Sci Rep 2018; 8:4601. [PMID: 29545526 PMCID: PMC5854602 DOI: 10.1038/s41598-018-22794-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 03/01/2018] [Indexed: 01/21/2023] Open
Abstract
RhoGAP proteins control the precise regulation of the ubiquitous small RhoGTPases. The Drosophila Crossveinless-c (Cv-c) RhoGAP is homologous to the human tumour suppressor proteins Deleted in Liver Cancer 1-3 (DLC1-3) sharing an identical arrangement of SAM, GAP and START protein domains. Here we analyse in Drosophila the requirement of each Cv-c domain to its function and cellular localization. We show that the basolateral membrane association of Cv-c is key for its epithelial function and find that the GAP domain targeted to the membrane can perform its RhoGAP activity independently of the rest of the protein, implying the SAM and START domains perform regulatory roles. We propose the SAM domain has a repressor effect over the GAP domain that is counteracted by the START domain, while the basolateral localization is mediated by a central, non-conserved Cv-c region. We find that DLC3 and Cv-c expression in the Drosophila ectoderm cause identical effects. In contrast, DLC1 is inactive but becomes functional if the central non-conserved DLC1 domain is substituted for that of Cv-c. Thus, these RhoGAP proteins are functionally equivalent, opening up the use of Drosophila as an in vivo model to analyse pharmacologically and genetically the human DLC proteins.
Collapse
Affiliation(s)
- Sol Sotillos
- CABD (CSIC/JA/Univ. Pablo de Olavide), Seville, Spain.
| | - Mario Aguilar-Aragon
- CABD (CSIC/JA/Univ. Pablo de Olavide), Seville, Spain.,The Francis Crick Institute, London, UK
| | | |
Collapse
|
197
|
Torres P, Castro M, Reyes M, Torres VA. Histatins, wound healing, and cell migration. Oral Dis 2018; 24:1150-1160. [PMID: 29230909 DOI: 10.1111/odi.12816] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 12/20/2022]
Abstract
Wounds in the oral mucosa heal faster and more efficiently than those in the skin, although the mechanisms underlying these differences are not completely clear. In the last 10 years, a group of salivary peptides, the histatins, has gained attention on behalf of their ability to improve several phases of the wound-healing process. In addition to their roles as anti-microbial agents and in enamel maintenance, histatins elicit other biological effects, namely by promoting the migration of different cell types contained in the oral mucosa and in non-oral tissues. Histatins, and specifically histatin-1, promote cell adhesion and migration in oral keratinocytes, gingival and dermal fibroblasts, non-oral epithelial cells, and endothelial cells. This is particularly relevant, as histatin-1 promotes the re-epithelialization phase and the angiogenic responses by increasing epithelial and endothelial cell migration. Although the molecular mechanisms associated with histatin-dependent cell migration remain poorly understood, recent studies have pointed to the control of signaling endosomes and the balance of small GTPases. This review aimed to update the literature on the effects of histatins in cell migration, with a focus on wound healing. We will also discuss the consequences that this increasing field will have in disease and therapy design.
Collapse
Affiliation(s)
- P Torres
- Faculty of Dentistry, Institute for Research in Dental Sciences, Universidad de Chile, Santiago, Chile
| | - M Castro
- Faculty of Dentistry, Institute for Research in Dental Sciences, Universidad de Chile, Santiago, Chile
| | - M Reyes
- Faculty of Dentistry, Institute for Research in Dental Sciences, Universidad de Chile, Santiago, Chile
| | - V A Torres
- Faculty of Dentistry, Institute for Research in Dental Sciences, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| |
Collapse
|
198
|
Ozdemir ES, Jang H, Gursoy A, Keskin O, Li Z, Sacks DB, Nussinov R. Unraveling the molecular mechanism of interactions of the Rho GTPases Cdc42 and Rac1 with the scaffolding protein IQGAP2. J Biol Chem 2018; 293:3685-3699. [PMID: 29358323 PMCID: PMC5846150 DOI: 10.1074/jbc.ra117.001596] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/12/2018] [Indexed: 12/22/2022] Open
Abstract
IQ motif-containing GTPase-activating proteins (IQGAPs) are scaffolding proteins playing central roles in cell-cell adhesion, polarity, and motility. The Rho GTPases Cdc42 and Rac1, in their GTP-bound active forms, interact with all three human IQGAPs. The IQGAP-Cdc42 interaction promotes metastasis by enhancing actin polymerization. However, despite their high sequence identity, Cdc42 and Rac1 differ in their interactions with IQGAP. Two Cdc42 molecules can bind to the Ex-domain and the RasGAP site of the GTPase-activating protein (GAP)-related domain (GRD) of IQGAP and promote IQGAP dimerization. Only one Rac1 molecule might bind to the RasGAP site of GRD and may not facilitate the dimerization, and the exact mechanism of Cdc42 and Rac1 binding to IQGAP is unclear. Using all-atom molecular dynamics simulations, site-directed mutagenesis, and Western blotting, we unraveled the detailed mechanisms of Cdc42 and Rac1 interactions with IQGAP2. We observed that Cdc42 binding to the Ex-domain of GRD of IQGAP2 (GRD2) releases the Ex-domain at the C-terminal region of GRD2, facilitating IQGAP2 dimerization. Cdc42 binding to the Ex-domain promoted allosteric changes in the RasGAP site, providing a binding site for the second Cdc42 in the RasGAP site. Of note, the Cdc42 "insert loop" was important for the interaction of the first Cdc42 with the Ex-domain. By contrast, differences in Rac1 insert-loop sequence and structure precluded its interaction with the Ex-domain. Rac1 could bind only to the RasGAP site of apo-GRD2 and could not facilitate IQGAP2 dimerization. Our detailed mechanistic insights help decipher how Cdc42 can stimulate actin polymerization in metastasis.
Collapse
Affiliation(s)
- E Sila Ozdemir
- From the Departments of Chemical and Biological Engineering and
| | - Hyunbum Jang
- the Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NCI-Frederick, Frederick, Maryland 21702
| | - Attila Gursoy
- Computer Engineering, Koc University, Istanbul 34450, Turkey,
| | - Ozlem Keskin
- From the Departments of Chemical and Biological Engineering and
| | - Zhigang Li
- the Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892, and
| | - David B Sacks
- the Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Ruth Nussinov
- the Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NCI-Frederick, Frederick, Maryland 21702,
- the Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
199
|
Yu SMW, Bonventre JV. Acute Kidney Injury and Progression of Diabetic Kidney Disease. Adv Chronic Kidney Dis 2018; 25:166-180. [PMID: 29580581 DOI: 10.1053/j.ackd.2017.12.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/15/2017] [Accepted: 12/22/2017] [Indexed: 12/23/2022]
Abstract
Diabetic kidney disease, commonly termed diabetic nephropathy (DN), is the most common cause of end-stage kidney disease (ESKD) worldwide. The characteristic histopathology of DN includes glomerular basement membrane thickening, mesangial expansion, nodular glomerular sclerosis, and tubulointerstitial fibrosis. Diabetes is associated with a number of metabolic derangements, such as reactive oxygen species overproduction, hypoxic state, mitochondrial dysfunction, and inflammation. In the past few decades, our knowledge of DN has advanced considerably although much needs to be learned. The traditional paradigm of glomerulus-centered pathophysiology has expanded to the tubule-interstitium, the immune response and inflammation. Biomarkers of proximal tubule injury have been shown to correlate with DN progression, independent of traditional glomerular injury biomarkers such as albuminuria. In this review, we summarize mechanisms of increased susceptibility to acute kidney injury in diabetes mellitus and the roles played by many kidney cell types to facilitate maladaptive responses leading to chronic and end-stage kidney disease.
Collapse
|
200
|
Rose JC, De Laporte L. Hierarchical Design of Tissue Regenerative Constructs. Adv Healthc Mater 2018; 7:e1701067. [PMID: 29369541 DOI: 10.1002/adhm.201701067] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/01/2017] [Indexed: 02/05/2023]
Abstract
The worldwide shortage of organs fosters significant advancements in regenerative therapies. Tissue engineering and regeneration aim to supply or repair organs or tissues by combining material scaffolds, biochemical signals, and cells. The greatest challenge entails the creation of a suitable implantable or injectable 3D macroenvironment and microenvironment to allow for ex vivo or in vivo cell-induced tissue formation. This review gives an overview of the essential components of tissue regenerating scaffolds, ranging from the molecular to the macroscopic scale in a hierarchical manner. Further, this review elaborates about recent pivotal technologies, such as photopatterning, electrospinning, 3D bioprinting, or the assembly of micrometer-scale building blocks, which enable the incorporation of local heterogeneities, similar to most native extracellular matrices. These methods are applied to mimic a vast number of different tissues, including cartilage, bone, nerves, muscle, heart, and blood vessels. Despite the tremendous progress that has been made in the last decade, it remains a hurdle to build biomaterial constructs in vitro or in vivo with a native-like structure and architecture, including spatiotemporal control of biofunctional domains and mechanical properties. New chemistries and assembly methods in water will be crucial to develop therapies that are clinically translatable and can evolve into organized and functional tissues.
Collapse
Affiliation(s)
- Jonas C. Rose
- DWI—Leibniz Institute for Interactive Materials Forckenbeckstr. 50 Aachen D‐52074 Germany
| | - Laura De Laporte
- DWI—Leibniz Institute for Interactive Materials Forckenbeckstr. 50 Aachen D‐52074 Germany
| |
Collapse
|