1
|
Cabrera-Rodríguez R, Pérez-Yanes S, Lorenzo-Sánchez I, Trujillo-González R, Estévez-Herrera J, García-Luis J, Valenzuela-Fernández A. HIV Infection: Shaping the Complex, Dynamic, and Interconnected Network of the Cytoskeleton. Int J Mol Sci 2023; 24:13104. [PMID: 37685911 PMCID: PMC10487602 DOI: 10.3390/ijms241713104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
HIV-1 has evolved a plethora of strategies to overcome the cytoskeletal barrier (i.e., actin and intermediate filaments (AFs and IFs) and microtubules (MTs)) to achieve the viral cycle. HIV-1 modifies cytoskeletal organization and dynamics by acting on associated adaptors and molecular motors to productively fuse, enter, and infect cells and then traffic to the cell surface, where virions assemble and are released to spread infection. The HIV-1 envelope (Env) initiates the cycle by binding to and signaling through its main cell surface receptors (CD4/CCR5/CXCR4) to shape the cytoskeleton for fusion pore formation, which permits viral core entry. Then, the HIV-1 capsid is transported to the nucleus associated with cytoskeleton tracks under the control of specific adaptors/molecular motors, as well as HIV-1 accessory proteins. Furthermore, HIV-1 drives the late stages of the viral cycle by regulating cytoskeleton dynamics to assure viral Pr55Gag expression and transport to the cell surface, where it assembles and buds to mature infectious virions. In this review, we therefore analyze how HIV-1 generates a cell-permissive state to infection by regulating the cytoskeleton and associated factors. Likewise, we discuss the relevance of this knowledge to understand HIV-1 infection and pathogenesis in patients and to develop therapeutic strategies to battle HIV-1.
Collapse
Affiliation(s)
- Romina Cabrera-Rodríguez
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| | - Silvia Pérez-Yanes
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| | - Iria Lorenzo-Sánchez
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| | - Rodrigo Trujillo-González
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
- Analysis Department, Faculty of Mathematics, Universidad de La Laguna (ULL), 38200 La Laguna, Spain
| | - Judith Estévez-Herrera
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| | - Jonay García-Luis
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| | - Agustín Valenzuela-Fernández
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| |
Collapse
|
2
|
Bozic I, Savic D, Lavrnja I. Astrocyte phenotypes: Emphasis on potential markers in neuroinflammation. Histol Histopathol 2020; 36:267-290. [PMID: 33226087 DOI: 10.14670/hh-18-284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Astrocytes, the most abundant glial cells in the central nervous system (CNS), have numerous integral roles in all CNS functions. They are essential for synaptic transmission and support neurons by providing metabolic substrates, secreting growth factors and regulating extracellular concentrations of ions and neurotransmitters. Astrocytes respond to CNS insults through reactive astrogliosis, in which they go through many functional and molecular changes. In neuroinflammatory conditions reactive astrocytes exert both beneficial and detrimental functions, depending on the context and heterogeneity of astrocytic populations. In this review we profile astrocytic diversity in the context of neuroinflammation; with a specific focus on multiple sclerosis (MS) and its best-described animal model experimental autoimmune encephalomyelitis (EAE). We characterize two main subtypes, protoplasmic and fibrous astrocytes and describe the role of intermediate filaments in the physiology and pathology of these cells. Additionally, we outline a variety of markers that are emerging as important in investigating astrocytic biology in both physiological conditions and neuroinflammation.
Collapse
Affiliation(s)
- Iva Bozic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Danijela Savic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
3
|
Kim RR, Chen Z, J. Mann T, Bastard K, F. Scott K, Church WB. Structural and Functional Aspects of Targeting the Secreted Human Group IIA Phospholipase A 2. Molecules 2020; 25:molecules25194459. [PMID: 32998383 PMCID: PMC7583969 DOI: 10.3390/molecules25194459] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/20/2020] [Accepted: 09/25/2020] [Indexed: 12/11/2022] Open
Abstract
Human group IIA secretory phospholipase A2 (hGIIA) promotes the proliferation of cancer cells, making it a compelling therapeutic target, but it is also significant in other inflammatory conditions. Consequently, suitable inhibitors of hGIIA have always been sought. The activation of phospholipases A2 and the catalysis of glycerophospholipid substrates generally leads to the release of fatty acids such as arachidonic acid (AA) and lysophospholipid, which are then converted to mediator compounds, including prostaglandins, leukotrienes, and the platelet-activating factor. However, this ability of hGIIA to provide AA is not a complete explanation of its biological role in inflammation, as it has now been shown that it also exerts proinflammatory effects by a catalysis-independent mechanism. This mechanism is likely to be highly dependent on key specific molecular interactions, and the full mechanistic descriptions of this remain elusive. The current candidates for the protein partners that may mediate this catalysis-independent mechanism are also introduced in this review. A key discovery has been that selective inhibition of the catalysis-independent activity of hGIIA is achieved with cyclised derivatives of a pentapeptide, FLSYK, derived from the primary sequence of hGIIA. The effects of hGIIA on cell function appear to vary depending on the pathology studied, and so its mechanism of action is complex and context-dependent. This review is comprehensive and covers the most recent developments in the understanding of the many facets of hGIIA function and inhibition and the insight they provide into their clinical application for disease treatment. A cyclic analogue of FLSYK, c2, the most potent analogue known, has now been taken into clinical trials targeting advanced prostate cancer.
Collapse
Affiliation(s)
- Ryung Rae Kim
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (R.R.K.); (Z.C.); (K.B.)
| | - Zheng Chen
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (R.R.K.); (Z.C.); (K.B.)
| | - Timothy J. Mann
- School of Medicine, Western Sydney University, Centre for Oncology, Education and Research Translation and The Ingham Institute, Liverpool, NSW 2170, Australia;
| | - Karine Bastard
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (R.R.K.); (Z.C.); (K.B.)
| | - Kieran F. Scott
- School of Medicine, Western Sydney University, Centre for Oncology, Education and Research Translation and The Ingham Institute, Liverpool, NSW 2170, Australia;
- Correspondence: (K.F.S.); (W.B.C.); Tel.: +61-2-8738-9026 (K.F.S.); +61-2-9036-6569 (W.B.C.)
| | - W. Bret Church
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (R.R.K.); (Z.C.); (K.B.)
- Correspondence: (K.F.S.); (W.B.C.); Tel.: +61-2-8738-9026 (K.F.S.); +61-2-9036-6569 (W.B.C.)
| |
Collapse
|
4
|
Biskou O, Casanova V, Hooper KM, Kemp S, Wright GP, Satsangi J, Barlow PG, Stevens C. The type III intermediate filament vimentin regulates organelle distribution and modulates autophagy. PLoS One 2019; 14:e0209665. [PMID: 30699149 PMCID: PMC6353089 DOI: 10.1371/journal.pone.0209665] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 12/10/2018] [Indexed: 12/21/2022] Open
Abstract
The cytoskeletal protein vimentin plays a key role in positioning of organelles within the cytosol and has been linked to the regulation of numerous cellular processes including autophagy, however, how vimentin regulates autophagy remains relatively unexplored. Here we report that inhibition of vimentin using the steroidal lactone Withaferin A (WFA) causes vimentin to aggregate, and this is associated with the relocalisation of organelles including autophagosomes and lysosomes from the cytosol to a juxtanuclear location. Vimentin inhibition causes autophagosomes to accumulate, and we demonstrate this results from modulation of mechanistic target of rapamycin (mTORC1) activity, and disruption of autophagosome-lysosome fusion. We suggest that vimentin plays a physiological role in autophagosome and lysosome positioning, thus identifying vimentin as a key factor in the regulation of mTORC1 and autophagy.
Collapse
Affiliation(s)
- Olga Biskou
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Victor Casanova
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Kirsty M. Hooper
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Sadie Kemp
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Graham P. Wright
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Jack Satsangi
- Centre for Genomic & Experimental Medicine, University of Edinburgh, Western General Hospital Campus, Crewe Road, Edinburgh, United Kingdom
- Translational Gastroenterology Unit, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Peter G. Barlow
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
| | - Craig Stevens
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
5
|
Bastounis EE, Yeh YT, Theriot JA. Matrix stiffness modulates infection of endothelial cells by Listeria monocytogenes via expression of cell surface vimentin. Mol Biol Cell 2018; 29:1571-1589. [PMID: 29718765 PMCID: PMC6080647 DOI: 10.1091/mbc.e18-04-0228] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Extracellular matrix stiffness (ECM) is one of the many mechanical forces acting on mammalian adherent cells and an important determinant of cellular function. While the effect of ECM stiffness on many aspects of cellular behavior has been studied previously, how ECM stiffness might mediate susceptibility of host cells to infection by bacterial pathogens is hitherto unexplored. To address this open question, we manufactured hydrogels of varying physiologically relevant stiffness and seeded human microvascular endothelial cells (HMEC-1) on them. We then infected HMEC-1 with the bacterial pathogen Listeria monocytogenes (Lm) and found that adhesion of Lm to host cells increases monotonically with increasing matrix stiffness, an effect that requires the activity of focal adhesion kinase (FAK). We identified cell surface vimentin as a candidate surface receptor mediating stiffness-dependent adhesion of Lm to HMEC-1 and found that bacterial infection of these host cells is decreased when the amount of surface vimentin is reduced. Our results provide the first evidence that ECM stiffness can mediate the susceptibility of mammalian host cells to infection by a bacterial pathogen.
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305
| | - Yi-Ting Yeh
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Julie A Theriot
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
6
|
Crow J, Atay S, Banskota S, Artale B, Schmitt S, Godwin AK. Exosomes as mediators of platinum resistance in ovarian cancer. Oncotarget 2017; 8:11917-11936. [PMID: 28060758 PMCID: PMC5355315 DOI: 10.18632/oncotarget.14440] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/16/2016] [Indexed: 12/21/2022] Open
Abstract
Exosomes have been implicated in the cell-cell transfer of oncogenic proteins and genetic material. We speculated this may be one mechanism by which an intrinsically platinum-resistant population of epithelial ovarian cancer (EOC) cells imparts its influence on surrounding tumor cells. To explore this possibility we utilized a platinum-sensitive cell line, A2780 and exosomes derived from its resistant subclones, and an unselected, platinum-resistant EOC line, OVCAR10. A2780 cells demonstrate a ~2-fold increase in viability upon treatment with carboplatin when pre-exposed to exosomes from platinum-resistant cells as compared to controls. This coincided with increased epithelial to mesenchymal transition (EMT). DNA sequencing of EOC cell lines revealed previously unreported somatic mutations in the Mothers Against Decapentaplegic Homolog 4 (SMAD4) within platinum-resistant cells. A2780 cells engineered to exogenously express these SMAD4 mutations demonstrate up-regulation of EMT markers following carboplatin treatment, are more resistant to carboplatin, and release exosomes which impart a ~1.7-fold increase in resistance in naive A2780 recipient cells as compared to controls. These studies provide the first evidence that acquired SMAD4 mutations enhance the chemo-resistance profile of EOC and present a novel mechanism in which exchange of tumor-derived exosomes perpetuates an EMT phenotype, leading to the development of subpopulations of platinum-refractory cells.
Collapse
Affiliation(s)
- Jennifer Crow
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Safinur Atay
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Samagya Banskota
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Biomedical Engineering, Duke University, Durham, North Carolina, NC, USA
| | - Brittany Artale
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA
| | - Sarah Schmitt
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,University of Kansas Cancer Center, Kansas City, KS, USA
| |
Collapse
|
7
|
Yousaf M, Tayyeb A, Ali G. Expression profiling of adhesion proteins during prenatal and postnatal liver development in rats. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2017; 10:21-28. [PMID: 29033593 PMCID: PMC5614736 DOI: 10.2147/sccaa.s139497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Culturing of primary hepatocytes and stem cell-derived hepatocytes faces a major issue of dedifferentiation due to absence of cell–cell adhesion and 3D structures. One of the possible ways to eliminate the problem of dedifferentiation is mimicking the expression pattern of adhesion proteins during the normal developmental process of liver cells. The purpose of this study was to evaluate the expression pattern of some key adhesion proteins, namely, E-cadherin, N-cadherin, epithelial CAM (EpCAM), intracellular CAM (ICAM), collagen 1α1, α-actinin, β-catenin and vimentin, in the liver tissue during prenatal and postnatal stages. Furthermore, differences in their expression between prenatal, early postnatal and adult stages were highlighted. Wistar rats were used to isolate livers at prenatal Day 14 and 17 as well as on postnatal Day 1, 3, 7 and 14. The liver from adult rats was used as control. Both conventional and real-time quantitative polymerase chain reactions (PCRs) were performed. For most of the adhesion proteins such as E-cadherin, N-cadherin, EpCAM, ICAM, collagen 1α1 and α-actinin, low expression was observed around prenatal Day 14 and an increasing expression was observed in the postnatal period. Moreover, β-catenin and vimentin showed higher expression in the early prenatal period, which decreased gradually in the postnatal period, but still this low expression was considerably higher than that in the adult control rats. This basic knowledge of the regulation of expression of adhesion proteins during different developmental stages indicates their vital role in liver development. This pattern can be further studied and imitated under in vitro conditions to achieve better cell–cell interactions.
Collapse
Affiliation(s)
- Mehwish Yousaf
- National Centre of Excellence in Molecular Biology, University of the Punjab
| | - Asima Tayyeb
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Gibran Ali
- National Centre of Excellence in Molecular Biology, University of the Punjab
| |
Collapse
|
8
|
Fang F, Ni K, Cai Y, Ye Z, Shang J, Shen S, Xiong C. Biological characters of human dermal fibroblasts derived from foreskin of male infertile patients. Tissue Cell 2017; 49:56-63. [DOI: 10.1016/j.tice.2016.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 11/22/2016] [Accepted: 12/07/2016] [Indexed: 12/15/2022]
|
9
|
Castro-Muñozledo F, Meza-Aguilar DG, Domínguez-Castillo R, Hernández-Zequinely V, Sánchez-Guzmán E. Vimentin as a Marker of Early Differentiating, Highly Motile Corneal Epithelial Cells. J Cell Physiol 2016; 232:818-830. [DOI: 10.1002/jcp.25487] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 07/11/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Federico Castro-Muñozledo
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del IPN; México City Mexico
| | - Diana G. Meza-Aguilar
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del IPN; México City Mexico
| | - Rocío Domínguez-Castillo
- Department of Molecular Biomedicine; Centro de Investigación y de Estudios Avanzados del IPN; México City Mexico
| | | | - Erika Sánchez-Guzmán
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del IPN; México City Mexico
| |
Collapse
|
10
|
Yu YTC, Chien SC, Chen IY, Lai CT, Tsay YG, Chang SC, Chang MF. Surface vimentin is critical for the cell entry of SARS-CoV. J Biomed Sci 2016; 23:14. [PMID: 26801988 PMCID: PMC4724099 DOI: 10.1186/s12929-016-0234-7] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/12/2016] [Indexed: 01/19/2023] Open
Abstract
Background Severe acute respiratory syndrome coronavirus (SARS-CoV) caused a global panic due to its high morbidity and mortality during 2002 and 2003. Soon after the deadly disease outbreak, the angiotensin-converting enzyme 2 (ACE2) was identified as a functional cellular receptor in vitro and in vivo for SARS-CoV spike protein. However, ACE2 solely is not sufficient to allow host cells to become susceptible to SARS-CoV infection, and other host factors may be involved in SARS-CoV spike protein-ACE2 complex. Results A host intracellular filamentous cytoskeletal protein vimentin was identified by immunoprecipitation and LC-MS/MS analysis following chemical cross-linking on Vero E6 cells that were pre-incubated with the SARS-CoV spike protein. Moreover, flow cytometry data demonstrated an increase of the cell surface vimentin level by 16.5 % after SARS-CoV permissive Vero E6 cells were treated with SARS-CoV virus-like particles (VLPs). A direct interaction between SARS-CoV spike protein and host surface vimentin was further confirmed by far-Western blotting. In addition, antibody neutralization assay and shRNA knockdown experiments indicated a vital role of vimentin in cell binding and uptake of SARS-CoV VLPs and the viral spike protein. Conclusions A direct interaction between vimentin and SARS-CoV spike protein during viral entry was observed. Vimentin is a putative anti-viral drug target for preventing/reducing the susceptibility to SARS-CoV infection.
Collapse
Affiliation(s)
- Yvonne Ting-Chun Yu
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, First Section, Taipei, 100, Taiwan.
| | - Ssu-Chia Chien
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, First Section, Taipei, 100, Taiwan.
| | - I-Yin Chen
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, First Section, Taipei, 100, Taiwan. .,Institute of Microbiology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, First Section, Taipei, 100, Taiwan.
| | - Chia-Tsen Lai
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, First Section, Taipei, 100, Taiwan.
| | - Yeou-Guang Tsay
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, 112, Taiwan.
| | - Shin C Chang
- Institute of Microbiology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, First Section, Taipei, 100, Taiwan.
| | - Ming-Fu Chang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, First Section, Taipei, 100, Taiwan.
| |
Collapse
|
11
|
Romani R, Fallarino F, Pirisinu I, Calvitti M, Caselli A, Fiaschi T, Gamberi T, Matino D, Talesa VN, Donti E, Puccetti P, Modesti A, Magherini F. Comparative proteomic analysis of two distinct stem-cell populations from human amniotic fluid. MOLECULAR BIOSYSTEMS 2015; 11:1622-32. [PMID: 25811139 DOI: 10.1039/c5mb00018a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human amniotic fluid (AF) contains a variety of stem cells of embryonic and extra-embryonic origins. We characterized two distinct types of stem cells isolated from residual AF material derived from prenatal diagnostic amniocentesis. The two types of cells differed in their morphology and growth kinetics, showing fast (fast human amniotic stem cells; fHASCs) or slow (slow human amniotic stem cells; sHASCs) population-doubling times. Both fHASCs and sHASCs expressed pluripotent stem-cell markers, yet unlike sHASCs, clonogenic fHASCs would generate embryoid bodies and maintain their original phenotype during prolonged in vitro passaging. fHASCs - but not sHASCs - expressed the KLF4, SSEA-4 and CD117 markers. Differential proteomic analysis allowed us to identify the protein patterns specific for either cell type as potentially contributing to their distinct phenotypes. We found thirty-six proteins that were differentially expressed by the two cell types, and those proteins were classified according to their biological and molecular functions. Bioinformatic cluster analysis revealed differential occurrence of cytoskeletal proteins, such as vimentin, F-actin-binding protein, and chloride intracellular channel protein 1. Selected proteins differentially expressed by fHASCs and sHASCs were further characterized by Western blot analysis and confocal microscopy.
Collapse
Affiliation(s)
- Rita Romani
- Department of Experimental Medicine, University of Perugia, Polo Didattico Sant'Andrea delle Fratte, Piazzale Gambuli, 06132 Perugia, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hułas-Stasiak M, Dobrowolski P, Tomaszewska E, Kostro K. Maternal acrylamide treatment reduces ovarian follicle number in newborn guinea pig offspring. Reprod Toxicol 2013; 42:125-31. [PMID: 23994668 DOI: 10.1016/j.reprotox.2013.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 08/05/2013] [Accepted: 08/20/2013] [Indexed: 01/22/2023]
Abstract
Acrylamide is an industrial chemical which has toxic effects on reproduction. In this study, we investigated whether acrylamide administered prenatally can induce follicular atresia in the newborn guinea pig ovary. Another aim was to describe the localization of vimentin filaments and determine their participation in atresia. After prenatal acrylamide treatment, the pool of primordial and primary follicles was significantly reduced. The number of caspase 3 and TUNEL positive oocytes increased compared to the control group. There were no differences in Lamp1 (autophagy marker) staining. A vimentin immunosignal was present in the granulosa cells of primordial, primary and secondary follicles. Interestingly, in contrast to the control group, the oocytes from all follicles in the ACR-treated females were negative for vimentin. These data suggest that prenatal exposure to acrylamide reduced the number of ovarian follicles by inducing follicular atresia mediated by oocyte apoptosis. Acrylamide-induced apoptosis may be associated with destruction of vimentin filaments.
Collapse
Affiliation(s)
- Monika Hułas-Stasiak
- Department of Anatomy and Anthropology, Maria Curie-Sklodowska University, Akademicka St. 19, 20-033 Lublin, Poland.
| | | | | | | |
Collapse
|
13
|
Bargagna-Mohan P, Deokule SP, Thompson K, Wizeman J, Srinivasan C, Vooturi S, Kompella UB, Mohan R. Withaferin A effectively targets soluble vimentin in the glaucoma filtration surgical model of fibrosis. PLoS One 2013; 8:e63881. [PMID: 23667686 PMCID: PMC3648549 DOI: 10.1371/journal.pone.0063881] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 04/09/2013] [Indexed: 01/23/2023] Open
Abstract
Withaferin A (WFA) is a natural product that binds to soluble forms of the type III intermediate filament (IF) vimentin. Currently, it is unknown under what pathophysiological contexts vimentin is druggable, as cytoskeltal vimentin-IFs are abundantly expressed. To investigate druggability of vimentin, we exploited rabbit Tenon's capsule fibroblast (RbTCF) cell cultures and the rabbit glaucoma filtration surgical (GFS) model of fibrosis. WFA potently caused G₀/G₁ cell cycle inhibition (IC₅₀ 25 nM) in RbTCFs, downregulating ubiquitin E3 ligase skp2 and inducing p27(Kip1) expression. Transforming growth factor (TGF)-ß-induced myofibroblast transformation caused development of cell spheroids with numerous elongated invadopodia, which WFA blocked potently by downregulating soluble vimentin and α-smooth muscle actin (SMA) expression. In the pilot proof-of-concept study using the GFS model, subconjunctival injections of a low WFA dose reduced skp2 expression in Tenon's capsule and increased p27(Kip1) expression without significant alteration to vimentin-IFs. This treatment maintains significant nanomolar WFA concentrations in anterior segment tissues that correspond to WFA's cell cycle targeting activity. A ten-fold higher WFA dose caused potent downregulation of soluble vimentin and skp2 expression, but as found in cell cultures, no further increase in p27(Kip1) expression was observed. Instead, this high WFA dose potently induced vimentin-IF disruption and downregulated α-SMA expression that mimicked WFA activity in TGF-ß-treated RbTCFs that blocked cell contractile activity at submicromolar concentrations. These findings illuminate that localized WFA injection to ocular tissues exerts pharmacological control over the skp2-p27(Kip1) pathway by targeting of soluble vimentin in a model of surgical fibrosis.
Collapse
Affiliation(s)
- Paola Bargagna-Mohan
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Sunil P. Deokule
- Ophthalmology and Visual Science, University of Kentucky, Lexington, Kentucky, United States of America
| | - Kyle Thompson
- Ophthalmology and Visual Science, University of Kentucky, Lexington, Kentucky, United States of America
| | - John Wizeman
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Cidambi Srinivasan
- Statistics, University of Kentucky, Lexington, Kentucky, United States of America
| | - Sunil Vooturi
- Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Uday B. Kompella
- Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Royce Mohan
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| |
Collapse
|
14
|
Cogli L, Progida C, Bramato R, Bucci C. Vimentin phosphorylation and assembly are regulated by the small GTPase Rab7a. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1283-93. [PMID: 23458836 PMCID: PMC3787733 DOI: 10.1016/j.bbamcr.2013.02.024] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 02/19/2013] [Accepted: 02/20/2013] [Indexed: 11/30/2022]
Abstract
Intermediate filaments are cytoskeletal elements important for cell architecture. Recently it has been discovered that intermediate filaments are highly dynamic and that they are fundamental for organelle positioning, transport and function thus being an important regulatory component of membrane traffic. We have identified, using the yeast two-hybrid system, vimentin, a class III intermediate filament protein, as a Rab7a interacting protein. Rab7a is a member of the Rab family of small GTPases and it controls vesicular membrane traffic to late endosomes and lysosomes. In addition, Rab7a is important for maturation of phagosomes and autophagic vacuoles. We confirmed the interaction in HeLa cells by co-immunoprecipitation and pull-down experiments, and established that the interaction is direct using bacterially expressed recombinant proteins. Immunofluorescence analysis on HeLa cells indicate that Rab7a-positive vesicles sometimes overlap with vimentin filaments. Overexpression of Rab7a causes an increase in vimentin phosphorylation at different sites and causes redistribution of vimentin in the soluble fraction. Consistently, Rab7a silencing causes an increase of vimentin present in the insoluble fraction (assembled). Also, expression of Charcot–Marie–Tooth 2B-causing Rab7a mutant proteins induces vimentin phosphorylation and increases the amount of vimentin in the soluble fraction. Thus, modulation of expression levels of Rab7a wt or expression of Rab7a mutant proteins changes the assembly of vimentin and its phosphorylation state indicating that Rab7a is important for the regulation of vimentin function. ► We searched for new Rab7a interacting proteins and we found vimentin. ► We demonstrated that Rab7a interacts directly with vimentin. ► Rab7a influences vimentin's phosphorylation and soluble/insoluble ratio. ► Rab7a regulates vimentin organization and function.
Collapse
Affiliation(s)
- Laura Cogli
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | | | | | | |
Collapse
|
15
|
Differentiating rectal carcinoma by an immunohistological analysis of carcinomas of pelvic organs based on the NCBI Literature Survey and the Human Protein Atlas database. Surg Today 2012; 42:515-25. [PMID: 22441574 DOI: 10.1007/s00595-012-0167-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 08/01/2011] [Indexed: 12/27/2022]
Abstract
The treatments and prognoses of pelvic organ carcinomas differ, depending on whether the primary tumor originated in the rectum, urinary bladder, prostate, ovary, or uterus; therefore, it is essential to diagnose pathologically the primary origin and stages of these tumors. To establish the panels of immunohistochemical markers for differential diagnosis, we reviewed 91 of the NCBI articles on these topics and found that the results correlated closely with those of the public protein database, the Human Protein Atlas. The results revealed the panels of immunohistochemical markers for the differential diagnosis of rectal adenocarcinoma, in which [+] designates positivity in rectal adenocarcinoma and [-] designates negativity in rectal adenocarcinoma: from bladder adenocarcinoma, CDX2[+], VIL1[+], KRT7[-], THBD[-] and UPK3A[-]; from prostate adenocarcinoma, CDX2[+], VIL1[+], CEACAM5[+], KLK3(PSA)[-], ACPP(PAP)[-] and SLC45A3(prostein)[-]; and from ovarian mucinous adenocarcinoma, CEACAM5[+], VIL1[+], CDX2[+], KRT7[-] and MUC5AC[-]. The panels of markers distinguishing ovarian serous adenocarcinoma, cervical carcinoma, and endometrial adenocarcinoma were also represented. Such a comprehensive review on the differential diagnosis of carcinomas of pelvic organs has not been reported before. Thus, much information has been accumulated in public databases to provide an invaluable resource for clinicians and researchers.
Collapse
|
16
|
Challa AA, Stefanovic B. A novel role of vimentin filaments: binding and stabilization of collagen mRNAs. Mol Cell Biol 2011; 31:3773-89. [PMID: 21746880 PMCID: PMC3165730 DOI: 10.1128/mcb.05263-11] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 07/04/2011] [Indexed: 12/17/2022] Open
Abstract
The stem-loop in the 5' untranslated region (UTR) of collagen α1(I) and α2(I) mRNAs (5'SL) is the key element regulating their stability and translation. Stabilization of collagen mRNAs is the predominant mechanism for high collagen expression in fibrosis. LARP6 binds the 5'SL of α1(I) and α2(I) mRNAs with high affinity. Here, we report that vimentin filaments associate with collagen mRNAs in a 5'SL- and LARP6-dependent manner and stabilize collagen mRNAs. LARP6 interacts with vimentin filaments through its La domain and colocalizes with the filaments in vivo. Knockdown of LARP6 by small interfering RNA (siRNA) or mutation of the 5'SL abrogates the interaction of collagen mRNAs with vimentin filaments. Vimentin knockout fibroblasts produce reduced amounts of type I collagen due to decreased stability of collagen α1(I) and α2(I) mRNAs. Disruption of vimentin filaments using a drug or by expression of dominant-negative desmin reduces type I collagen expression, primarily due to decreased stability of collagen mRNAs. RNA fluorescence in situ hybridization (FISH) experiments show that collagen α1(I) and α2(I) mRNAs are associated with vimentin filaments in vivo. Thus, vimentin filaments may play a role in the development of tissue fibrosis by stabilizing collagen mRNAs. This finding will serve as a rationale for targeting vimentin in the development of novel antifibrotic therapies.
Collapse
Affiliation(s)
- Azariyas A. Challa
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, Florida 32306
| | - Branko Stefanovic
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, Florida 32306
| |
Collapse
|
17
|
Park JE, Kim HT, Lee S, Lee YS, Choi UK, Kang JH, Choi SY, Kang TC, Choi MS, Kwon OS. Differential expression of intermediate filaments in the process of developing hepatic steatosis. Proteomics 2011; 11:2777-89. [DOI: 10.1002/pmic.201000544] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Revised: 03/19/2011] [Accepted: 04/13/2011] [Indexed: 12/18/2022]
|
18
|
Ozaki N, Matheis KA, Gamber M, Feidl T, Nolte T, Kalkuhl A, Deschl U. Identification of genes involved in gentamicin-induced nephrotoxicity in rats--a toxicogenomic investigation. ACTA ACUST UNITED AC 2009; 62:555-66. [PMID: 19664912 DOI: 10.1016/j.etp.2009.07.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Revised: 07/08/2009] [Accepted: 07/15/2009] [Indexed: 01/19/2023]
Abstract
For the application of microarray technology as an additional endpoint in toxicological studies, there is a need to understand associations between pathological processes and gene expression alterations. In the current study, we investigated gentamicin as a nephrotoxic model compound. Gene expression changes of the kidney in response to a dose of 80 mg/kg gentamicin were analyzed by using DNA microarray technology and alterations in gene expression were associated with results from conventional histopathological investigations and with the described pathomechanisms of gentamicin. Under the conditions of our experiment, the mRNA level of 211 genes were found to be deregulated by gentamicin. The gentamicin-induced affection of proximal convoluted tubules was associated with a strong up-regulation of mRNAs encoding for proteins which are used as nephrotoxicity markers in urine and plasma such as Kim-1, Osteopontin and TIMP1. Candidate marker genes for nephrotoxicity such as GATM were deregulated. Gentamicin-induced lysosomal phospholipidosis was indicated by deregulation of lysosomal located gene products such as ATP6V1D, a subunit of the lysosomal H+ transporting ATPase. Effects on glucose transport and metabolism were indicated by the down-regulation on SGLT-2 and glucose-6-phosphatase. Renal cell apoptosis was indicated by up-regulated genes as TP53 and BAX. The role of oxidative stress in gentamicin toxicity was reflected by deregulation of transferrin receptor and heme oxygenase. The results of the study show the potential of microarray technology to study a complex mechanism of toxicity in a single study.
Collapse
Affiliation(s)
- N Ozaki
- Department of Molecular & Cellular Biology, Kobe Pharma Research Institute, Nippon Boehringer Ingelheim Co., Ltd. 6-7-5, Minatojima-Minamimachi Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | | | | | | | | | | | | |
Collapse
|
19
|
Proteomic analysis identifies proteins that continue to grow hepatic stem-like cells without differentiation. Cytotechnology 2008; 57:137-43. [PMID: 19003158 PMCID: PMC2553666 DOI: 10.1007/s10616-008-9122-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Accepted: 01/07/2008] [Indexed: 10/28/2022] Open
Abstract
To understand the molecular mechanism underlying vigorous proliferative activity of hepatic stem-like (HSL) cells, we performed two-dimensional electrophoresis to identify the proteins statistically more abundant in rapidly growing undifferentiated HSL cells than in sodium butyrate-treated differentiated HSL cells. Matrix-assisted laser desorption/ionization-time of flight mass spectrometry and Mascot search identified 6 proteins including prohibitin, vimentin, ezrin, annexin A3, acidic ribosomal phosphoprotein P0 and Grp75. Prohibitin and vimentin control the mitogen-activated protein (MAP) kinase pathway. Ezrin is phosphorylated by various protein-tyrosine kinases and modulates interactions between cytoskeletal and membrane proteins. Annexin A3 has a role in DNA synthesis. Acidic ribosomal phosphoprotein P0 and Grp75 play in protein synthesis. These results suggest that the proteins related to the MAP kinase cascade had some role in continuous proliferation of HSL cells without differentiation.
Collapse
|
20
|
Brauksiepe B, Mujica AO, Herrmann H, Schmidt ER. The Serine/threonine kinase Stk33 exhibits autophosphorylation and phosphorylates the intermediate filament protein Vimentin. BMC BIOCHEMISTRY 2008; 9:25. [PMID: 18811945 PMCID: PMC2567967 DOI: 10.1186/1471-2091-9-25] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Accepted: 09/23/2008] [Indexed: 02/02/2023]
Abstract
Background Colocalization of Stk33 with vimentin by double immunofluorescence in certain cells indicated that vimentin might be a target for phosphorylation by the novel kinase Stk33. We therefore tested in vitro the ability of Stk33 to phosphorylate recombinant full length vimentin and amino-terminal truncated versions thereof. In order to prove that Stk33 and vimentin are also in vivo associated proteins co-immunoprecipitation experiments were carried out. For testing the enzymatic activity of immunoprecipitated Stk33 we incubated precipitated Stk33 with recombinant vimentin proteins. To investigate whether Stk33 binds directly to vimentin, an in vitro co-sedimentation assay was performed. Results The results of the kinase assays demonstrate that Stk33 is able to specifically phosphorylate the non-α-helical amino-terminal domain of vimentin in vitro. Furthermore, co-immunoprecipitation experiments employing cultured cell extracts indicate that Stk33 and vimentin are associated in vivo. Immunoprecipitated Stk33 has enzymatic activity as shown by successful phosphorylation of recombinant vimentin proteins. The results of the co-sedimentation assay suggest that vimentin binds directly to Stk33 and that no additional protein mediates the association. Conclusion We hypothesize that Stk33 is involved in the in vivo dynamics of the intermediate filament cytoskeleton by phosphorylating vimentin.
Collapse
|
21
|
Abstract
Vimentin is one of the intermediate filaments that functions in structural support, signal transduction and organelle positioning of a cell. In the present study, we report the contribution of vimentin in mitochondrial morphology and organization. Using subcellular fractionation, immunoprecipitation and fluorescence microscopy analyses, we found that vimentin was associated with mitochondria. Knockdown of vimentin resulted in mitochondrial fragmentation, swelling and disorganization. We further demonstrated that the vimentin cytoskeleton co-localized and interacted with mitochondria to a greater extent than other cytoskeletal components known to support mitochondria. Our results also suggest that vimentin could participate in the mitochondrial association of microtubules. As mitochondrial morphologies determine mitochondrial function, our findings revealed a potentially important relationship between the vimentin-based intermediate filaments and the regulation of mitochondria.
Collapse
|
22
|
Changes in cell morphology and cytoskeletal organization are induced by human mitotic checkpoint gene, Bub1. Biochem Biophys Res Commun 2008; 365:691-7. [DOI: 10.1016/j.bbrc.2007.11.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Accepted: 11/06/2007] [Indexed: 02/02/2023]
|
23
|
Bargagna-Mohan P, Hamza A, Kim YE, Ho YK(A, Mor-Vaknin N, Wendschlag N, Liu J, Evans RM, Markovitz DM, Zhan CG, Kim KB, Mohan R. The tumor inhibitor and antiangiogenic agent withaferin A targets the intermediate filament protein vimentin. ACTA ACUST UNITED AC 2007; 14:623-34. [PMID: 17584610 PMCID: PMC3228641 DOI: 10.1016/j.chembiol.2007.04.010] [Citation(s) in RCA: 244] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 03/30/2007] [Accepted: 04/19/2007] [Indexed: 11/22/2022]
Abstract
The natural product withaferin A (WFA) exhibits antitumor and antiangiogenesis activity in vivo, which results from this drug's potent growth inhibitory activities. Here, we show that WFA binds to the intermediate filament (IF) protein, vimentin, by covalently modifying its cysteine residue, which is present in the highly conserved alpha-helical coiled coil 2B domain. WFA induces vimentin filaments to aggregate in vitro, an activity manifested in vivo as punctate cytoplasmic aggregates that colocalize vimentin and F-actin. WFA's potent dominant-negative effect on F-actin requires vimentin expression and induces apoptosis. Finally, we show that WFA-induced inhibition of capillary growth in a mouse model of corneal neovascularization is compromised in vimentin-deficient mice. These findings identify WFA as a chemical genetic probe of IF functions, and illuminate a potential molecular target for withanolide-based therapeutics for treating angioproliferative and malignant diseases.
Collapse
Affiliation(s)
| | - Adel Hamza
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | | | - Yik Khuan (Abby) Ho
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Nirit Mor-Vaknin
- Department of Internal Medicine, Division of Infectious Diseases
| | | | - Junjun Liu
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Robert M. Evans
- Department of Pathology, University of Colorado Health Sciences Center, Denver, CO 80045, USA
| | - David M. Markovitz
- Department of Internal Medicine, Division of Infectious Diseases
- Cellular and Molecular Biology Program, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Royce Mohan
- Department of Ophthalmology and Visual Sciences
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
- Correspondence:
| |
Collapse
|
24
|
Ngan CY, Yamamoto H, Seshimo I, Tsujino T, Man-i M, Ikeda JI, Konishi K, Takemasa I, Ikeda M, Sekimoto M, Matsuura N, Monden M. Quantitative evaluation of vimentin expression in tumour stroma of colorectal cancer. Br J Cancer 2007; 96:986-92. [PMID: 17325702 PMCID: PMC2360104 DOI: 10.1038/sj.bjc.6603651] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recent studies have identified vimentin, a type III intermediate filament, among genes differentially expressed in tumours with more invasive features, suggesting an association between vimentin and tumour progression. The aim of this study, was to investigate whether vimentin expression in colon cancer tissue is of clinical relevance. We performed immunostaining in 142 colorectal cancer (CRC) samples and quantified the amount of vimentin expression using computer-assisted image analysis. Vimentin expression in the tumour stroma of CRC was associated with shorter survival. Overall survival in the high vimentin expression group was 71.2% compared with 90.4% in the low-expression group (P=0.002), whereas disease-free survival for the high-expression group was 62.7% compared with 86.7% for the low-expression group (P=0.001). Furthermore, the prognostic power of vimentin for disease recurrence was maintained in both stage II and III CRC. Multivariate analysis suggested that vimentin was a better prognostic indicator for disease recurrence (risk ratio=3.5) than the widely used lymph node status (risk ratio=2.2). Vimentin expression in the tumour stroma may reflect a higher malignant potential of the tumour and may be a useful predictive marker for disease recurrence in CRC patients.
Collapse
Affiliation(s)
- C Y Ngan
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - H Yamamoto
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita city, Osaka 565-0871, Japan; E-mail:
| | - I Seshimo
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - T Tsujino
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - M Man-i
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - J-I Ikeda
- Department of Pathology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - K Konishi
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - I Takemasa
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - M Ikeda
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - M Sekimoto
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - N Matsuura
- Department of Pathology, School of Allied Health Science, Faculty of Medicine, Osaka University, Osaka 565-0871, Japan
| | - M Monden
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
25
|
Garg A, Barnes PF, Porgador A, Roy S, Wu S, Nanda JS, Griffith DE, Girard WM, Rawal N, Shetty S, Vankayalapati R. Vimentin Expressed onMycobacterium tuberculosis-Infected Human Monocytes Is Involved in Binding to the NKp46 Receptor. THE JOURNAL OF IMMUNOLOGY 2006; 177:6192-8. [PMID: 17056548 DOI: 10.4049/jimmunol.177.9.6192] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We previously showed that human NK cells used the NKp46 receptor to lyse Mycobacterium tuberculosis H37Ra-infected monocytes. To identify ligands on H37Ra-infected human mononuclear phagocytes, we used anti-NKp46 to immunoprecipitate NKp46 from NK cells bound to its ligand(s) on H37Ra-infected monocytes. Mass spectrometry analysis identified a 57-kDa molecule, vimentin, as a putative ligand for NKp46. Vimentin expression was significantly up-regulated on the surface of infected monocytes, compared with uninfected cells, and this was confirmed by fluorescence microscopy. Anti-vimentin antiserum inhibited NK cell lysis of infected monocytes, whereas antiserum to actin, another filamentous protein, did not. CHO-K1 cells transfected with a vimentin construct were lysed much more efficiently by NK cells than cells transfected with a control plasmid. This lysis was inhibited by mAb-mediated masking of NKp46 (on NK cells) or vimentin (on infected monocytes). ELISA and Far Western blotting showed that recombinant vimentin bound to a NKp46 fusion protein. These results indicate that vimentin is involved in binding of NKp46 to M. tuberculosis H37Ra-infected mononuclear phagocytes.
Collapse
Affiliation(s)
- Ankita Garg
- Center for Pulmonary and Infectious Disease Control, University of Texas Health Center, 11937 U.S. Highway 271, Tyler, TX 75708, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Boucrot E, Henry T, Borg JP, Gorvel JP, Méresse S. The Intracellular Fate of Salmonella Depends on the Recruitment of Kinesin. Science 2005; 308:1174-8. [PMID: 15905402 DOI: 10.1126/science.1110225] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Salmonella enterica causes a variety of diseases, including gastroenteritis and typhoid fever. The success of this pathogen depends on its capacity to proliferate within host cells in a membrane-bound compartment. We found that the Salmonella-containing vacuole recruited the plus-end-directed motor kinesin. Bacterial effector proteins translocated into the host cell by a type III secretion system antagonistically regulated this event. Among these effectors, SifA targeted SKIP, a host protein that down-regulated the recruitment of kinesin on the bacterial vacuole and, in turn, controlled vacuolar membrane dynamics.
Collapse
Affiliation(s)
- Emmanuel Boucrot
- Centre d'Immunologie de Marseille-Luminy, CNRS-INSERM-Université de la Méditerranée, Parc Scientifique de Luminy, Case 906-13288 Marseille Cedex 9, France
| | | | | | | | | |
Collapse
|
27
|
Perlson E, Hanz S, Ben-Yaakov K, Segal-Ruder Y, Seger R, Fainzilber M. Vimentin-dependent spatial translocation of an activated MAP kinase in injured nerve. Neuron 2005; 45:715-26. [PMID: 15748847 DOI: 10.1016/j.neuron.2005.01.023] [Citation(s) in RCA: 389] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2004] [Revised: 11/16/2004] [Accepted: 01/14/2005] [Indexed: 01/26/2023]
Abstract
How are phosphorylated kinases transported over long intracellular distances, such as in the case of axon to cell body signaling after nerve injury? Here, we show that the MAP kinases Erk1 and Erk2 are phosphorylated in sciatic nerve axoplasm upon nerve injury, concomitantly with the production of soluble forms of the intermediate filament vimentin by local translation and calpain cleavage in axoplasm. Vimentin binds phosphorylated Erks (pErk), thus linking pErk to the dynein retrograde motor via direct binding of vimentin to importin beta. Injury-induced Elk1 activation and neuronal regeneration are inhibited or delayed in dorsal root ganglion neurons from vimentin null mice, and in rats treated with a MEK inhibitor or with a peptide that prevents pErk-vimentin binding. Thus, soluble vimentin enables spatial translocation of pErk by importins and dynein in lesioned nerve.
Collapse
Affiliation(s)
- Eran Perlson
- Department of Biological Chemistry, Weizmann Institute of Science, 76100 Rehovot, Israel
| | | | | | | | | | | |
Collapse
|
28
|
Imamura T, Kanai F, Kawakami T, Amarsanaa J, Ijichi H, Hoshida Y, Tanaka Y, Ikenoue T, Tateishi K, Kawabe T, Arakawa Y, Miyagishi M, Taira K, Yokosuka O, Omata M. Proteomic analysis of the TGF-beta signaling pathway in pancreatic carcinoma cells using stable RNA interference to silence Smad4 expression. Biochem Biophys Res Commun 2004; 318:289-96. [PMID: 15110786 DOI: 10.1016/j.bbrc.2004.04.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2004] [Indexed: 11/24/2022]
Abstract
Smad4 is a tumor-suppressor gene that is lost or mutated in 50% of pancreatic carcinomas. Smad4 is also an intracellular transmitter of transforming growth factor-beta (TGF-beta) signals. Although its tumor-suppressor function is presumed to reside in its capacity to mediate TGF-beta-induced growth inhibition, there seems to be a Smad4-independent TGF-beta signaling pathway. Here, we succeeded in establishing Smad4 knockdown (S4KD) pancreatic cancer cell lines using stable RNA interference. Smad4 protein expression and TGF-beta-Smad4 signaling were impaired in S4KD cells, and we compared the proteomic changes with TGF-beta stimulation using two-dimensional gel electrophoresis (2-DE) and mass spectrometry. We identified five proteins that were up-regulated and seven proteins that were down-regulated; 10 of them were novel targets for TGF-beta. These proteins function in processes such as cytoskeletal regulation, cell cycle, and oxidative stress. Introducing siRNA-mediated gene silencing into proteomics revealed a novel TGF-beta signal pathway that did not involve Smad4.
Collapse
Affiliation(s)
- Takaaki Imamura
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Bacterial cells exhibit a wide variety of shapes. Recent results indicate that the characteristic crescent shape of Caulobacter crescentus depends upon an inter-mediate filament-like protein that localizes to the concave side of the cell.
Collapse
Affiliation(s)
- William Margolin
- Department of Microbiology and Molecular Genetics, University of Texas Medical School, 6431 Fannin, Houston, Texas 77030, USA.
| |
Collapse
|
30
|
Clarke EJ, Allan VJ. Cytokeratin intermediate filament organisation and dynamics in the vegetal cortex of living Xenopus laevis oocytes and eggs. CELL MOTILITY AND THE CYTOSKELETON 2003; 56:13-26. [PMID: 12905528 DOI: 10.1002/cm.10131] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Cytokeratin intermediate filaments are prominent constituents of developing Xenopus oocytes and eggs, forming radial and cortical networks. In order to investigate the dynamics of the cortical cytokeratin network, we expressed EGFP-tagged Xenopus cytokeratin 1(8) in oocytes and eggs. The EGFP-cytokeratin co-assembled with endogenous partner cytokeratin proteins to form fluorescent filaments. Using time-lapse confocal microscopy, cytokeratin filament assembly was monitored in live Xenopus oocytes at different stages of oogenesis, and in the artificially-activated mature egg during the first cell cycle. In stage III to V oocytes, cytokeratin proteins formed a loose cortical geodesic network, which became more tightly bundled in stage VI oocytes. Maturation of oocytes into metaphase II-arrested eggs induced disassembly of the EGFP-cytokeratin network. Imaging live eggs after artificial activation allowed us to observe the reassembly of cytokeratin filaments in the vegetal cortex. The earliest observable structures were loose foci, which then extended into curly filament bundles. The position and orientation of these bundles altered with time, suggesting that forces were acting upon them. During cortical rotation, the cytokeratin network realigned into a parallel array that translocated in a directed manner at 5 microm/minute, relative to stationary cortex. The cytokeratin filaments are, therefore, moving in association with the bulk cytoplasm of the egg, suggesting that they may provide a structural role at the moving interface between cortex and cytoplasm.
Collapse
Affiliation(s)
- Emma J Clarke
- School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | | |
Collapse
|
31
|
Mello ADS, Santos AQ, Funchal C. Correlação entre Hiperglicemia e Células do SNC, com Enfoque na Atividade Glial. ACTA ACUST UNITED AC 2001. [DOI: 10.34024/rnc.2012.v20.8284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Introdução. Entre os mecanismos biológicos que originam o quadro hiperglicêmico a predominância é do diabetes melittus (DM). O DM representa um grupo de desordens metabólicas caracterizadas por hiperglicemia crônica que ocasiona severas alterações celulares e teciduais. Objetivo. O presente trabalho analisou através de revisão da literatura o comportamento de células gliais expostas a elevadas concentrações de glicose, similares às observadas no DM. Método. Foi realizada uma revisão literária através de artigos científicos das bases de dados Pubmed, Science Direct, Scopus e Scielo. Resultados. Foram selecionados artigos e livros entre 1988 e 2009 que discutiam hiperglicemia, sistema nervoso central e que relacionavam hiperglicemia e células gliais. Conclusão. A hiperglicemia crônica proporcionada pelo DM pode influenciar de maneira danosa o metabolismo cerebral exercendo ações sobre a atividade glial. Podendo afetar a sobrevivência neuronal através da excitotoxicidade glutamatérgica e da produção de espécies reativas de oxigênio (ERO) e de espécies reativas de nitrogênio (ERN) que geram como consequência o processo de neuroinflamação. Tal processo inflamatório pode resultar em dano e morte neural caracterizando um processo neurodegerativo.
Collapse
|