1
|
Ruggiero C, Lalli E. Targeting the cytoskeleton against metastatic dissemination. Cancer Metastasis Rev 2021; 40:89-140. [PMID: 33471283 DOI: 10.1007/s10555-020-09936-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/08/2020] [Indexed: 02/08/2023]
Abstract
Cancer is a pathology characterized by a loss or a perturbation of a number of typical features of normal cell behaviour. Indeed, the acquisition of an inappropriate migratory and invasive phenotype has been reported to be one of the hallmarks of cancer. The cytoskeleton is a complex dynamic network of highly ordered interlinking filaments playing a key role in the control of fundamental cellular processes, like cell shape maintenance, motility, division and intracellular transport. Moreover, deregulation of this complex machinery contributes to cancer progression and malignancy, enabling cells to acquire an invasive and metastatic phenotype. Metastasis accounts for 90% of death from patients affected by solid tumours, while an efficient prevention and suppression of metastatic disease still remains elusive. This results in the lack of effective therapeutic options currently available for patients with advanced disease. In this context, the cytoskeleton with its regulatory and structural proteins emerges as a novel and highly effective target to be exploited for a substantial therapeutic effort toward the development of specific anti-metastatic drugs. Here we provide an overview of the role of cytoskeleton components and interacting proteins in cancer metastasis with a special focus on small molecule compounds interfering with the actin cytoskeleton organization and function. The emerging involvement of microtubules and intermediate filaments in cancer metastasis is also reviewed.
Collapse
Affiliation(s)
- Carmen Ruggiero
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, CNRS, 660 route des Lucioles-Sophia Antipolis, 06560, Valbonne, France.
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560, Valbonne, France.
| | - Enzo Lalli
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
- Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des Lucioles - Sophia Antipolis, 06560, Valbonne, France
| |
Collapse
|
2
|
Rahmanzade R. Redefinition of tumor capsule: Rho-dependent clustering of cancer-associated fibroblasts in favor of tensional homeostasis. Med Hypotheses 2019; 135:109425. [PMID: 31760246 DOI: 10.1016/j.mehy.2019.109425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 09/29/2019] [Accepted: 10/09/2019] [Indexed: 11/16/2022]
Abstract
Fibroblasts are the most frequent cells of the connective tissues. Having the ability to sense and respond to mechanical stimuli in addition to the biochemical ones makes them crucial for such a composite-like and tension-preserving tissue. Over the last decade, the investigation of the role of these cells in tumor progression was a hot topic of research in tumor biology. Literatures almost unanimously describe the re-education of stromal fibroblasts by tumor cells in favor of tumor progression, which resulted in the birth of a new nomenclature, the cancer-associated fibroblasts. On the other hand, some studies reported anti-tumor roles for these cells. Herein, author suggests that the previously described pro-migratory and pro-contractile contexts, which respectively results in divergent and convergent distribution of fibroblasts by changing Rho-Rac1 balance, could be applied for cancer-associated fibroblasts as well. Based on this proposed concept, stromal fibroblasts could represent different roles, either pro-tumor or anti-tumor, during the course of tumor progression. In the earlier phases, they tend to assemble along tumor-stroma interface in the form of tumor capsules in order to resist tumor growth and to maintain tensional homeostasis in stroma. But in later phases, after being chronically subjected to tumor-induced chemical and mechanical stimuli, they will gradually lose their substantial abilities to oppose tumor expansion and, in contrary, will promote tumorigenesis. In summary, this paper redefines tumor capsule from chemical and mechanical standpoints as Rho-dependent clustering of cancer-associated fibroblasts in favor of tensional homeostasis. Furthermore, it proposes that stromal fibroblasts will undergo some irreversible epigenetic changes in Rac1- and Rho-related proteins through tumor-stroma crosstalk, which irreversibly diminish their ability of capsule formation. Finally, the author discusses the possible researches helping us to assess the proposed concept and its clinical implications.
Collapse
Affiliation(s)
- Ramin Rahmanzade
- Biomedical Research & Training, University Hospital Basel, Mittlere Strasse 91, 4031 Basel, Switzerland.
| |
Collapse
|
3
|
Gandalovičová A, Rosel D, Fernandes M, Veselý P, Heneberg P, Čermák V, Petruželka L, Kumar S, Sanz-Moreno V, Brábek J. Migrastatics-Anti-metastatic and Anti-invasion Drugs: Promises and Challenges. Trends Cancer 2018; 3:391-406. [PMID: 28670628 PMCID: PMC5482322 DOI: 10.1016/j.trecan.2017.04.008] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In solid cancers, invasion and metastasis account for more than 90% of mortality. However, in the current armory of anticancer therapies, a specific category of anti-invasion and antimetastatic drugs is missing. Here, we coin the term ‘migrastatics’ for drugs interfering with all modes of cancer cell invasion and metastasis, to distinguish this class from conventional cytostatic drugs, which are mainly directed against cell proliferation. We define actin polymerization and contractility as target mechanisms for migrastatics, and review candidate migrastatic drugs. Critical assessment of these antimetastatic agents is warranted, because they may define new options for the treatment of solid cancers. Local invasion and metastasis, rather than clonal proliferation, are the dominant features of solid cancer. However, a specific category of anti-invasion and antimetastatic drugs is missing for treatment of solid cancer We propose the term ‘migrastatics’ for drugs interfering with all modes of cancer cell invasiveness and, consequently, with their ability to metastasize (e.g., inhibiting not only local invasion, but also extravasation and metastatic colonization). In solid cancer, drug resistance is the main cause of treatment failure, and is attributed to mutations of the target. Since targeting the cause, although academically desirable, may be futile, a pragmatic and near-term option is to move downstream, to common denominators of cell migration and/or invasion, such as actin polymerization and actomyosin-mediated contractility.
Collapse
Affiliation(s)
- Aneta Gandalovičová
- Department of Cell Biology, Charles University, Viničná 7, Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242, Vestec u Prahy, Czech Republic
| | - Daniel Rosel
- Department of Cell Biology, Charles University, Viničná 7, Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242, Vestec u Prahy, Czech Republic
| | | | - Pavel Veselý
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Petr Heneberg
- Charles University, Department of Internal Medicine, Third Faculty of Medicine, Prague, Czech Republic
| | - Vladimír Čermák
- Department of Cell Biology, Charles University, Viničná 7, Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242, Vestec u Prahy, Czech Republic
| | - Luboš Petruželka
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Sunil Kumar
- Ayurveda Molecular Modeling, Hyderabad, Telangana, India
| | - Victoria Sanz-Moreno
- Tumor Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London, UK.
| | - Jan Brábek
- Department of Cell Biology, Charles University, Viničná 7, Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242, Vestec u Prahy, Czech Republic.
| |
Collapse
|
4
|
Zuckerman V, Sokolov E, Swet JH, Ahrens WA, Showlater V, Iannitti DA, Mckillop IH. Expression and function of lysophosphatidic acid receptors (LPARs) 1 and 3 in human hepatic cancer progenitor cells. Oncotarget 2016; 7:2951-67. [PMID: 26701886 PMCID: PMC4823083 DOI: 10.18632/oncotarget.6696] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/16/2015] [Indexed: 01/26/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary cancer of the liver and is characterized by rapid tumor expansion and metastasis. Lysophosphatidic acid (LPA) signaling, via LPA receptors 1–6 (LPARs1–6), regulates diverse cell functions including motility, migration, and proliferation, yet the role of LPARs in hepatic tumor pathology is poorly understood. We sought to determine the expression and function of endothelial differentiation gene (EDG) LPARs (LPAR1–3) in human HCC and complimentary in vitro models. Human HCC were characterized by significantly elevated LPAR1/LPAR3 expression in the microenvironment between the tumor and non-tumor liver (NTL), a finding mirrored in human SKHep1 cells. Analysis of human tissue and human hepatic tumor cells in vitro revealed cells that express LPAR3 (HCC-NTL margin in vivo and SKHep1 in vitro) also express cancer stem cell markers in the absence of hepatocyte markers. Treatment of SKHep1 cells with exogenous LPA led to significantly increased cell motility but not proliferation. Using pharmacological agents and cells transfected to knock-down LPAR1 or LPAR3 demonstrated LPA-dependent cell migration occurs via an LPAR3-Gi-ERK-pathway independent of LPAR1. These data suggest cells that stain positive for both LPAR3 and cancer stem cell markers are distinct from the tumor mass per se, and may mediate tumor invasiveness/expansion via LPA-LPAR3 signaling.
Collapse
Affiliation(s)
| | - Eugene Sokolov
- Department of Surgery, Carolinas Medical Center, Charlotte, NC, USA 28203
| | - Jacob H Swet
- Department of Surgery, Carolinas Medical Center, Charlotte, NC, USA 28203
| | - William A Ahrens
- Department of Pathology, Carolinas Medical Center, Charlotte, NC, USA 28203
| | - Victor Showlater
- Department of Surgery, Carolinas Medical Center, Charlotte, NC, USA 28203
| | - David A Iannitti
- Department of Surgery, Carolinas Medical Center, Charlotte, NC, USA 28203
| | - Iain H Mckillop
- Department of Surgery, Carolinas Medical Center, Charlotte, NC, USA 28203
| |
Collapse
|
5
|
Kubo N, Araki K, Kuwano H, Shirabe K. Cancer-associated fibroblasts in hepatocellular carcinoma. World J Gastroenterol 2016; 22:6841-6850. [PMID: 27570421 PMCID: PMC4974583 DOI: 10.3748/wjg.v22.i30.6841] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/09/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023] Open
Abstract
The hepatic stellate cells in the liver are stimulated sustainably by chronic injury of the hepatocytes, activating myofibroblasts, which produce abundant collagen. Myofibroblasts are the major source of extracellular proteins during fibrogenesis, and may directly, or secreted products, contribute to carcinogenesis and tumor progression. Cancer-associated fibroblasts (CAFs) are one of the components of the tumor microenvironment that promote the proliferation and invasion of cancer cells by secreting various growth factors and cytokines. CAFs crosstalk with cancer cells stimulates tumor progression by creating a favorable microenvironment for progression, invasion, and metastasis through the epithelial-mesenchymal transition. Basic studies on CAFs have advanced, and the role of CAFs in tumors has been elucidated. In particular, for hepatocellular carcinoma, carcinogenesis from cirrhosis is a known fact, and participation of CAFs in carcinogenesis is supported. In this review, we discuss the current literature on the role of CAFs and CAF-related signaling in carcinogenesis, crosstalk with cancer cells, immunosuppressive effects, angiogenesis, therapeutic targets, and resistance to chemotherapy. The role of CAFs is important in cancer initiation and progression. CAFtargeted therapy may be effective for suppression not only of fibrosis but also cancer progression.
Collapse
|
6
|
Borin TF, Arbab AS, Gelaleti GB, Ferreira LC, Moschetta MG, Jardim-Perassi BV, Iskander ASM, Varma NRS, Shankar A, Coimbra VB, Fabri VA, de Oliveira JG, de Campos Zuccari DAP. Melatonin decreases breast cancer metastasis by modulating Rho-associated kinase protein-1 expression. J Pineal Res 2016; 60:3-15. [PMID: 26292662 PMCID: PMC4996347 DOI: 10.1111/jpi.12270] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/14/2015] [Indexed: 01/09/2023]
Abstract
The occurrence of metastasis, an important breast cancer prognostic factor, depends on cell migration/invasion mechanisms, which can be controlled by regulatory and effector molecules such as Rho-associated kinase protein (ROCK-1). Increased expression of this protein promotes tumor growth and metastasis, which can be restricted by ROCK-1 inhibitors. Melatonin has shown oncostatic, antimetastatic, and anti-angiogenic effects and can modulate ROCK-1 expression. Metastatic and nonmetastatic breast cancer cell lines were treated with melatonin as well as with specific ROCK-1 inhibitor (Y27632). Cell viability, cell migration/invasion, and ROCK-1 gene expression and protein expression were determined in vitro. In vivo lung metastasis study was performed using female athymic nude mice treated with either melatonin or Y27832 for 2 and 5 wk. The metastases were evaluated by X-ray computed tomography and single photon emission computed tomography (SPECT) and by immunohistochemistry for ROCK-1 and cytokeratin proteins. Melatonin and Y27632 treatments reduced cell viability and invasion/migration of both cell lines and decreased ROCK-1 gene expression in metastatic cells and protein expression in nonmetastatic cell line. The numbers of 'hot' spots (lung metastasis) identified by SPECT images were significantly lower in treated groups. ROCK-1 protein expression also was decreased in metastatic foci of treated groups. Melatonin has shown to be effective in controlling metastatic breast cancer in vitro and in vivo, not only via inhibition of the proliferation of tumor cells but also through direct antagonism of metastatic mechanism of cells rendered by ROCK-1 inhibition. When Y27632 was used, the effects were similar to those found with melatonin treatment.
Collapse
Affiliation(s)
- Thaiz Ferraz Borin
- Laboratory of Molecular Investigation of Cancer – LIMC, Department of Molecular Biology, Faculdade de Medicina de Sao Jose do Rio Preto – FAMERP, Sao Jose do Rio Preto, SP, Brazil
| | - Ali Syed Arbab
- Tumor angiogenesis laboratory, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - Gabriela Bottaro Gelaleti
- Laboratory of Molecular Investigation of Cancer – LIMC, Department of Molecular Biology, Faculdade de Medicina de Sao Jose do Rio Preto – FAMERP, Sao Jose do Rio Preto, SP, Brazil
- Universidade Estadual Paulista Julio de Mesquita Filho – IBILCE/UNESP, Sao Jose do Rio Preto, SP, Brazil
| | - Lívia Carvalho Ferreira
- Laboratory of Molecular Investigation of Cancer – LIMC, Department of Molecular Biology, Faculdade de Medicina de Sao Jose do Rio Preto – FAMERP, Sao Jose do Rio Preto, SP, Brazil
- Universidade Estadual Paulista Julio de Mesquita Filho – IBILCE/UNESP, Sao Jose do Rio Preto, SP, Brazil
| | - Marina Gobbe Moschetta
- Laboratory of Molecular Investigation of Cancer – LIMC, Department of Molecular Biology, Faculdade de Medicina de Sao Jose do Rio Preto – FAMERP, Sao Jose do Rio Preto, SP, Brazil
| | - Bruna Victorasso Jardim-Perassi
- Laboratory of Molecular Investigation of Cancer – LIMC, Department of Molecular Biology, Faculdade de Medicina de Sao Jose do Rio Preto – FAMERP, Sao Jose do Rio Preto, SP, Brazil
| | - ASM Iskander
- Tumor angiogenesis laboratory, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - Nadimpalli Ravi S. Varma
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, MI, USA
| | - Adarsh Shankar
- Tumor angiogenesis laboratory, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - Verena Benedick Coimbra
- Laboratory of Molecular Investigation of Cancer – LIMC, Department of Molecular Biology, Faculdade de Medicina de Sao Jose do Rio Preto – FAMERP, Sao Jose do Rio Preto, SP, Brazil
| | - Vanessa Alves Fabri
- Laboratory of Molecular Investigation of Cancer – LIMC, Department of Molecular Biology, Faculdade de Medicina de Sao Jose do Rio Preto – FAMERP, Sao Jose do Rio Preto, SP, Brazil
| | | | - Debora Aparecida Pires de Campos Zuccari
- Laboratory of Molecular Investigation of Cancer – LIMC, Department of Molecular Biology, Faculdade de Medicina de Sao Jose do Rio Preto – FAMERP, Sao Jose do Rio Preto, SP, Brazil
- Universidade Estadual Paulista Julio de Mesquita Filho – IBILCE/UNESP, Sao Jose do Rio Preto, SP, Brazil
| |
Collapse
|
7
|
Iyengar S, Zhan C, Lu J, Korngold R, Schwartz DH. Treatment with a rho kinase inhibitor improves survival from graft-versus-host disease in mice after MHC-haploidentical hematopoietic cell transplantation. Biol Blood Marrow Transplant 2014; 20:1104-11. [PMID: 24796280 DOI: 10.1016/j.bbmt.2014.04.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/24/2014] [Indexed: 11/26/2022]
Abstract
Acute graft-versus-host disease (GVHD) is a major complication of allogeneic hematopoietic cell transplantation (HCT) and the main cause of nonrelapse mortality during the first 100 days post-transplant. Although GVHD can be prevented by extensive removal of mature donor T cells from the donor hematopoietic stem cell population, doing so eliminates any potential allogeneic graft-versus-tumor (GVT) effect also mediated by donor T cells and results in unacceptable rates of cancer relapse. One potential solution to this problem of separating GVHD development from a GVT response is to prevent T cell-mediated GVHD in the intestinal tract (IT) while preserving systemic antihost alloreactivity of donor T cells that target residual tumor cells expressing host alloantigens. We examined the ability of the anti-inflammatory rho kinase inhibitor, fasudil, given orally and intraperitoneally, to prevent GVHD in a C3H → B6C3F1 mouse model of MHC-haploidentical bone marrow transplantation. Fasudil-treated recipients of anti-thy-1 mAb + C' treated bone marrow (ATBM) cells plus T cells had a 73% 90-day survival compared with 25% among untreated ATBM + T cell recipients (P < .0001). Severe initial weight loss was similar in the 2 groups, but less diarrhea was observed among treated animals, and fasudil-treated survivors recovered more weight than untreated survivors. Skin inflammation occurred and resolved between weeks 2 and 8 with similar severity and kinetics in both treated and untreated surviving animals, indicating persistent alloreactivity. Day 10 post-transplantation splenocytes from fasudil-treated mice, containing mature donor T cells, and day 98 splenocytes, containing mature donor and de novo thymus-derived T cells, exhibited alloreactivity against host parental antigens, as assessed by in vitro IFN-γ production and rounds of allostimulated proliferation, respectively. These data support the idea that targeted treatment of the IT with rho kinase inhibitors can ameliorate lethal GVHD while preserving systemic alloreactivity. The results also suggest that similar mechanisms of IT-specific tolerance or resistance to GVHD operate in fasudil-treated and untreated long-term survivors of allogeneic ATBM + T cells.
Collapse
Affiliation(s)
- Sujatha Iyengar
- Jurist Department of Research, Hackensack University Medical Center, Hackensack, New Jersey.
| | - Caixin Zhan
- Jurist Department of Research, Hackensack University Medical Center, Hackensack, New Jersey
| | - Jordan Lu
- Jurist Department of Research, Hackensack University Medical Center, Hackensack, New Jersey
| | - Robert Korngold
- Jurist Department of Research, Hackensack University Medical Center, Hackensack, New Jersey
| | - David H Schwartz
- Jurist Department of Research, Hackensack University Medical Center, Hackensack, New Jersey
| |
Collapse
|
8
|
Willier S, Butt E, Grunewald TGP. Lysophosphatidic acid (LPA) signalling in cell migration and cancer invasion: a focussed review and analysis of LPA receptor gene expression on the basis of more than 1700 cancer microarrays. Biol Cell 2013; 105:317-33. [PMID: 23611148 DOI: 10.1111/boc.201300011] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 04/16/2013] [Indexed: 12/27/2022]
Abstract
Lysophosphatidic acid (LPA) is a ubiquitously present signalling molecule involved in diverse cellular processes such as cell migration, proliferation and differentiation. LPA acts as an autocrine and/or paracrine signalling molecule via different G-protein-coupled LPA receptors (LPARs) that trigger a broad range of intracellular signalling cascades, especially the RHOA pathway. Mounting evidence suggests a crucial role of the LPA/LPAR-axis in cancer cell metastasis and promising studies are underway to investigate the therapeutic potential of LPAR-antagonists. This review summarises current knowledge on how LPA promotes cytoskeletal remodelling to enhance the migratory and invasive properties of cells, which may ultimately contribute to cancer metastasis. Furthermore, we provide comprehensive transcriptome analyses of published microarrays of more than 350 normal tissues and more than 1700 malignant tissues to define the expression signatures of LPARs and the LPA-generating enzymes autotaxin (ATX) and lipase member 1 (LIPI). These analyses demonstrate that ATX is highly expressed in a variety of carcinomas and sarcomas, whereas LIPI is almost exclusively overexpressed in highly aggressive Ewing's sarcomas, which underscores the potential contribution of LPA in metastatic disease. In addition, these analyses show that different cancer entities display distinct expression signatures of LPARs that distinguish them from one another. Finally, we discuss current approaches to specifically target the LPA/LPAR circuits in experimental cancer therapy.
Collapse
Affiliation(s)
- Semjon Willier
- Institute for Clinical Biochemistry and Pathobiochemistry, University of Würzburg, Würzburg, Germany
| | | | | |
Collapse
|
9
|
Garrido-Gómez T, Dominguez F, Quiñonero A, Estella C, Vilella F, Pellicer A, Simon C. Annexin A2 is critical for embryo adhesiveness to the human endometrium by RhoA activation through F-actin regulation. FASEB J 2012; 26:3715-27. [PMID: 22645245 DOI: 10.1096/fj.12-204008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Annexin A2 (ANXA2) is present in vivo in the mid- and late-secretory endometria and is mainly localized in the luminal epithelium. Our aim was to evaluate its function in regulating the human implantation process. With an in vitro adhesion model, constructed to evaluate how the mouse embryo and JEG-3 spheroids attach to human endometrial epithelial cells, we demonstrated that ANXA2 inhibition significantly diminishes embryo adhesiveness. ANXA2 is also implicated in endometrial epithelial cell migration and trophoblast outgrowth. ANXA2 was seen to be linked to the RhoA/ROCK pathway and to regulate cell adhesion. We noted that ANXA2 inhibition significantly reduces active RhoA, although RhoA inactivation does not alter the ANXA2 levels. RhoA inactivation and ROCK inhibition also moderate embryo adhesiveness to endometrial epithelial cells. We corroborated that the induction of constitutively active RhoA partially reverses the effects of ANXA2 inhibition on endometrial adhesiveness. These molecules colocalize on the plasma membrane of endometrial epithelial cells, and a large proportion of ANXA2 and RhoA are colocalized in the F-actin networks. The functional effects of ANXA2 inhibition and RhoA/ROCK inactivation are associated with significant alterations in F-actin organization and its depolymerization. ANXA2 may act upstream of the RhoA/ROCK pathway by regulating F-actin remodeling and is a key factor in human endometrial adhesiveness.
Collapse
Affiliation(s)
- Tamara Garrido-Gómez
- Fundación IVI, Instituto Universitario IVI, Universidad de Valencia, Fundación Investigación Clínico de Valencia Instituto de Investigacion Sanitaria, Valencia, Spain
| | | | | | | | | | | | | |
Collapse
|
10
|
Ren X, Ma Y, Xu M, Chen T, Zhang Z, Zhang Y. Construction, modification and evaluation of apolipoprotein A-I promoter-driven shRNA expression vectors against hTERT. Plasmid 2011; 65:42-50. [DOI: 10.1016/j.plasmid.2010.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 11/04/2010] [Accepted: 11/08/2010] [Indexed: 11/29/2022]
|
11
|
Park SY, Jeong KJ, Panupinthu N, Yu S, Lee J, Han JW, Kim JM, Lee JS, Kang J, Park CG, Mills GB, Lee HY. Lysophosphatidic acid augments human hepatocellular carcinoma cell invasion through LPA1 receptor and MMP-9 expression. Oncogene 2010; 30:1351-9. [PMID: 21102517 DOI: 10.1038/onc.2010.517] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lysophosphatidic acid (LPA), produced extracellularly by autotaxin (ATX), has diverse biological activities implicated in tumor initiation and progression, including increasing cell survival, angiogenesis, invasion and metastasis. ATX, LPA and the matrix metalloproteinase (MMP)-9 have all been implicated in hepatocellular carcinoma (HCC) invasion and metastasis. We, thus sought to determine whether ATX with subsequent LPA production and action, including induction of MMP-9 could provide a unifying mechanism. ATX transcripts and LPA receptor type 1 (LPA1) protein are elevated in HCC compared with normal tissues. Silencing or pharmacological inhibition of LPA1 significantly attenuated LPA-induced MMP-9 expression and HCC cell invasion. Further, reducing MMP-9 activity or expression significantly inhibits LPA-induced HCC cell invasion, demonstrating that MMP-9 is downstream of LPA1. Inhibition of phosphoinositide-3 kinase (PI3K) signaling or dominant-negative mutants of protein kinase Cδ and p38 mitogen-activated protein kinase (MAPK) abrogated LPA-induced MMP-9 expression and subsequent invasion. We thus demonstrate a mechanistic cascade of ATX-producing LPA with LPA activating LPA1 and inducing MMP-9 through coordinate activation of the PI3K and the p38 MPAK signaling cascades, providing novel biomarkers and potential therapeutic targets for HCC.
Collapse
Affiliation(s)
- S Y Park
- Department of Pharmacology, College of Medicine, Konyang University, Daejeon, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Tanikawa T, Kurohane K, Imai Y. Regulatory effect of lysophosphatidic acid on lymphocyte migration. Biol Pharm Bull 2010; 33:204-8. [PMID: 20118541 DOI: 10.1248/bpb.33.204] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Lysophosphatidic acid (LPA) is a lipid mediator that is known to exhibit chemotactic activity toward a variety of cancer cells. However, its effect on the immune system has not been studied extensively. Another lipid mediator, sphingosine-1-phosphate (S1P), has been shown to influence lymphocyte recirculation by regulating lymphocyte egress from lymphoid organs. In this study, we found that LPA inhibits spontaneous migration of mouse splenic lymphocytes through a chemorepulsive effect. We also demonstrated that LPA inhibits chemokine CCL21-induced lymphocyte migration. This inhibitory effect on CCL21-induced migration was observed for both T and B cells. The involvement of a receptor, LPA(1), LPA(2) or LPA(3), in the inhibition of the CCL21-induced migration was confirmed with a synthetic agonist, oleyl thiophosphate. Considering that the signaling by CCL21 through cognate receptor CCR7 contributes to lymphocyte homing and dendritic cell trafficking to lymph nodes, LPA may play a role as a key regulator of these processes. The inhibitory effect of LPA is in remarkable contrast to the effect of S1P receptor signaling, which is known to potentiate lymphocyte chemotaxis involving CCR7.
Collapse
Affiliation(s)
- Takashi Tanikawa
- Laboratory of Microbiology and Immunology and the Global COE Program, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8526, Japan
| | | | | |
Collapse
|
13
|
Lu H, Knutson KL, Gad E, Disis ML. The tumor antigen repertoire identified in tumor-bearing neu transgenic mice predicts human tumor antigens. Cancer Res 2007; 66:9754-61. [PMID: 17018635 DOI: 10.1158/0008-5472.can-06-1083] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
FVB/N mice transgenic for nontransforming rat neu develop spontaneous breast cancers that are neu positive and estrogen receptor negative, mimicking premenopausal human breast cancer. These animals have been widely used as a model for immunobased therapies targeting HER-2/neu. In this study, we used serological analysis of recombinant cDNA expression libraries to characterize the antigenic repertoire of neu transgenic (neu-tg) mice and questioned the ability of this murine model to predict potential human tumor antigens. After screening 3 x 10(6) clones from 3 different cDNA libraries, 15 tumor antigens were identified, including cytokeratin 2-8, glutamyl-prolyl-tRNA synthetase, complement C3, galectin 8, and serine/threonine-rich protein kinase 1. Multiple proteins involved in the Rho/Rho-associated, coiled coil-containing protein kinase (Rock) signal transduction pathway were found to be immunogenic, including Rock1, Rho/Rac guanine nucleotide exchange factor 2, and schistosoma mansoni adult worm antigen preparation 70. All of the identified antigens are self-proteins that are expressed in normal tissues in addition to breast tumors and the majority of the antigens are intracellular proteins. More than half of the mouse tumor antigens have human homologues that have been reported previously as tumor antigens. Finally, the tumor-specific antibody immunity and marked immune cell infiltration that was observed in mice with spontaneous tumors were not observed in mice with transplanted tumors. Our results indicate that neu-tg mice bearing spontaneous tumors develop humoral immunity to their tumors similar to cancer patients and that tumor antigens identified in transgenic mouse may predict immunogenic human homologues.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/biosynthesis
- Antibodies, Neoplasm/blood
- Antibodies, Neoplasm/immunology
- Antigens, Neoplasm/analysis
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- DNA, Complementary/genetics
- Disease Progression
- Female
- Gene Expression Regulation, Neoplastic
- Gene Library
- Genes, erbB-2
- Glycoproteins/genetics
- Glycoproteins/physiology
- Humans
- Male
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mice
- Mice, Transgenic
- Neoplasm Transplantation
- RNA, Messenger/biosynthesis
- RNA, Neoplasm/biosynthesis
- Rats
- Receptor, ErbB-2
- Specific Pathogen-Free Organisms
- Testis/metabolism
Collapse
Affiliation(s)
- Hailing Lu
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98195, USA.
| | | | | | | |
Collapse
|
14
|
Wang D, Dou K, Xiang H, Song Z, Zhao Q, Chen Y, Li Y. Involvement of RhoA in progression of human hepatocellular carcinoma. J Gastroenterol Hepatol 2007; 22:1916-20. [PMID: 17914970 DOI: 10.1111/j.1440-1746.2006.04534.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM The activation of Rho proteins has been shown to lead to loss of polarity in cancer cells, as well as reorganization of the cytoskeleton and facilitation of cell motility, possibly resulting in their malignant potential. The clinicopathological significance of RhoA, however, is not yet well known in the case of hepatocellular carcinoma (HCC). This study evaluated the clinicopathological correlation of RhoA levels with HCC. METHODS The intratumor expression level of Rho was determined and compared with that in adjacent non-cancerous hepatic tissue using quantitative real time RT-PCR and Western blotting in 64 patients with HCC. Relationships between the level of RhoA and clinicopathological factors were examined. RhoA protein expression was confirmed by immunohistochemistry. RESULTS RhoA immunostaining was strong in malignant tissue whereas it was minimal in benign tissue. Tumor tissue of HCC patients demonstrated a copy number of RhoA mRNA that was well correlated with its protein level in each case and was significantly higher than that found in the corresponding non-cancerous liver tissue (P < 0.01). With regard to venous invasion, satellite lesions and advanced pTNM stage, the RhoA level tended to be higher in HCC than that seen in negative tissue (P < 0.05). CONCLUSIONS This is the first demonstration that the expression level of RhoA is correlated with tumor progression and metastasis in HCC. RhoA in tumor tissue might be expected to be not only a good candidate marker for invasive and proliferative tumor cells, but also a molecular target of these cells.
Collapse
Affiliation(s)
- Desheng Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Xi'an, Shannxi Province, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Salhia B, Tran NL, Symons M, Winkles JA, Rutka JT, Berens ME. Molecular pathways triggering glioma cell invasion. Expert Rev Mol Diagn 2006; 6:613-26. [PMID: 16824034 DOI: 10.1586/14737159.6.4.613] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The efficacy of treating malignant gliomas with adjuvant therapies remains largely unsuccessful due to the inability to effectively target invading cells. Although our understanding of glioma oncogenesis has steadily improved, the molecular mechanisms that mediate glioma invasion are still poorly understood. It is clear that genetic alterations in malignant gliomas affect cell proliferation and cell cycle control, which are the targets of most chemotherapeutic agents. However, effective therapy against cell invasion has been less successful. Future treatment protocols must incorporate pharmacotherapeutic strategies that target resistant infiltrative glioma cells as well as proliferating ones. Thus, delineating the point of convergence of signaling pathways, which mediate glioma invasion, proliferation and apoptosis, may identify novel targets that can serve as possible points of therapeutic intervention. The optimization of novel strategies will require reliable preclinical testing using an in vivo animal model of brain invasion. Current applications of existing animal models are not currently optimized or characterized for use in glioma invasion research. As such, the development of a bona fide brain invasion model in vivo must be established. Progress in understanding molecular mechanisms driving glioma invasion will be critical to the success of managing and improving the outcome of patients with this grave disease.
Collapse
Affiliation(s)
- Bodour Salhia
- The Arthur & Sonia Labatt Brain Tumour Research Center, The Hospital for Sick Children, The University of Toronto, Toronto, Ontario, Canada.
| | | | | | | | | | | |
Collapse
|
16
|
Yam JWP, Ko FCF, Chan CY, Yau TO, Tung EKK, Leung THY, Jin DY, Ng IOL. Tensin2 variant 3 is associated with aggressive tumor behavior in human hepatocellular carcinoma. Hepatology 2006; 44:881-90. [PMID: 17006924 DOI: 10.1002/hep.21339] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tensins are a new family of proteins that act as an important link among extracellular matrix, actin cytoskeleton, and signal transduction and have been implicated in human cancers. Tensin2 was initially identified in a search for new tensin family members that share extensive sequence homology with tensin1. Tensin2 was highly expressed in liver tissues. A recent study reported that one of the splicing variants of tensin2, variant 3, promotes cell migration. In the present study, we aimed to elucidate the role of variant 3 in hepatocarcinogenesis by assessing the expression of variant 3 mRNA in hepatocellular carcinoma (HCC) tissue and ectopically expressing variant 3 in HCC cell lines. Analysis of variant 3 expression in human HCC tissue revealed it was overexpressed in 46% (23/50) of tumor tissues as compared with the corresponding nontumorous livers. High expression of variant 3 was significantly associated with venous invasion (P = .037), tumor microsatellite formation (P = .022), and tumor nonencapsulation (P = .049). Our ectopic expression study showed that variant 3 significantly promoted the cell growth and motility of HCC cells. The clonal transfectants of variant 3 were more closely packed and resulted in a higher saturation density than in the control vector transfectants. Variant 3 expression also enhanced the proliferation rate in culture and in vivo tumorigenicity in nude mice. In conclusion, we reveal a novel role for variant 3 in the progression of HCC and suggest the feasibility of elevated variant 3 expression as a tumor progression marker for HCC.
Collapse
Affiliation(s)
- Judy Wai Ping Yam
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Lawler K, Foran E, O'Sullivan G, Long A, Kenny D. Mobility and invasiveness of metastatic esophageal cancer are potentiated by shear stress in a ROCK- and Ras-dependent manner. Am J Physiol Cell Physiol 2006; 291:C668-77. [PMID: 16641163 DOI: 10.1152/ajpcell.00626.2005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To metastasize, tumor cells must adopt different morphological responses to resist shear forces encountered in circulating blood and invade through basement membranes. The Rho and Ras GTPases play a critical role in regulating this dynamic behavior. Recently, we demonstrated shear-induced activation of adherent esophageal metastatic cells, characterized by formation of dynamic membrane blebs. Although membrane blebbing has only recently been characterized as a rounded mode of cellular invasion promoted through Rho kinase (ROCK), the role of shear forces in modulating membrane blebbing activity is unknown. To further characterize membrane blebbing in esophageal metastatic cells (OC-1 cell line), we investigated the role of shear in cytoskeletal remodeling and signaling through ROCK and Ras. Our results show that actin and tubulin colocalize to the cortical ring of the OC-1 cell under static conditions. However, under shear, actin acquires a punctuate distribution and tubulin localizes to the leading edge of the OC-1 cell. We show for the first time that dynamic bleb formation is induced by shear alone independent of integrin-mediated adhesion ( P < 0.001, compared with OC-1 cells). Y-27632, a specific inhibitor of ROCK, causes a significant reduction in shear-induced bleb formation and inhibits integrin αvβ3-Ras colocalization at the leading edge of the cell. Direct measurement of Ras activation shows that the level of GTP-bound Ras is elevated in sheared OC-1 cells and that the shear-induced increase in Ras activity is inhibited by Y-27632. Finally, we show that shear stress significantly increases OC-1 cell invasion ( P < 0.007), an effect negated by the presence of Y-27632. Together our findings suggest a novel physiological role for ROCK and Ras in metastatic cell behavior.
Collapse
Affiliation(s)
- Karen Lawler
- Department of Clinical Pharmacology, Royal College of Surgeons, Dublin, Ireland
| | | | | | | | | |
Collapse
|
18
|
Salhia B, Rutten F, Nakada M, Beaudry C, Berens M, Kwan A, Rutka JT. Inhibition of Rho-kinase affects astrocytoma morphology, motility, and invasion through activation of Rac1. Cancer Res 2005; 65:8792-800. [PMID: 16204049 DOI: 10.1158/0008-5472.can-05-0160] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Malignant astrocytomas are highly invasive neoplasms infiltrating diffusely into regions of normal brain. Whereas the molecular and cellular mechanisms governing astrocytoma invasion remain poorly understood, evidence in other cell systems has implicated a role for the Rho-GTPases in cell motility and invasion. Here, we examine how the inhibition or activation of Rho-kinase (ROCK) affects astrocytoma morphology, motility, and invasion. ROCK was inhibited in astrocytoma cells by using 5 to 100 mumol/L of Y27632 or by expressing the dominant-negative ROCK mutant, RB/PH TT. ROCK activation was achieved by expressing a constitutively active mutant, CAT. ROCK inhibition led to morphologic and cytoskeletal alterations characterized by an increase in the number and length of cell processes, increased membrane ruffling, and collapse of actin stress fibers. Using two-dimensional radial migration and Boyden chamber assays, we show that astrocytoma migration and invasion were increased at least 2-fold by ROCK inhibition. On the contrary, ROCK activation significantly inhibited migration and invasion of astrocytoma cells. Furthermore, using a Rac-GTP pull-down assay, we show that Rac1 is activated as a consequence of ROCK inhibition. Finally, we show that treatment of astrocytoma cells with small interfering RNA duplexes specific for Rac1-reversed stellation, prevented membrane ruffling formation and abrogated the increased motility observed following treatment with Y27632. Our data show that Rac1 plays a major role in astrocytoma morphology, motility, and invasion. These findings warrant further investigation to determine precisely how the modulation of Rac1 and ROCK can be exploited to inhibit glioma invasion.
Collapse
Affiliation(s)
- Bodour Salhia
- The Arthur and Sonia Labatt Brain Tumour Research Center and Division of Neurosurgery, The Hospital for Sick Children, The University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
19
|
Katoh H, Shibata T, Kokubu A, Ojima H, Loukopoulos P, Kanai Y, Kosuge T, Fukayama M, Kondo T, Sakamoto M, Hosoda F, Ohki M, Imoto I, Inazawa J, Hirohashi S. Genetic profile of hepatocellular carcinoma revealed by array-based comparative genomic hybridization: identification of genetic indicators to predict patient outcome. J Hepatol 2005; 43:863-74. [PMID: 16139920 DOI: 10.1016/j.jhep.2005.05.033] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2005] [Revised: 05/17/2005] [Accepted: 05/18/2005] [Indexed: 01/22/2023]
Abstract
BACKGROUND/AIMS We conducted an analysis of chromosomal numerical aberrations and their clinical significance in hepatocellular carcinoma. METHODS We analyzed 87 hepatocellular carcinomas by array-based comparative genomic hybridization with an array containing 800 bacterial artificial chromosome clones. RESULTS Frequent (>30%) chromosomal losses on 1p36.1, 4q21-25, 4q34-35.1, 8p23.3b-11.1, 13q14.1-14.3, 16p13.3, 16q22.1-24.3b, 17p13.3-13.1 and 17p13.3-11, and gains on 1q21-44f, 2q21.2, 2q34, 3q11.2, 5p14.2, 5q13.2-14, 7p22, 7p14.2, 7q21.1, 7q22.3, 7q34, 8q12-24.3 and 17q23, were observed. Recurrent (>5%) amplifications were detected on 1q25, 8q11 and 11q11, and we discovered a novel homozygous deletion at 14q32.11. The extent of chromosomal aberrations correlated significantly with various clinicopathological characteristics of the tumors, and increased in a stepwise manner with the progression of hepatocellular carcinoma. We also identified novel chromosomal alterations that were significantly associated with a range of malignant phenotypes. Multivariate analysis revealed that both chromosomal loss on 17p13.3 and gain on 8q11 are independent prognostic indicators. CONCLUSIONS Our results contribute to a complete description of genomic structural aberrations in relation to hepatocarcinogenesis and provide a valuable basis from which we can begin to understand the characteristics of tumors, predict patient outcomes and discover novel therapeutic targets for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hiroto Katoh
- Pathology Division, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, and Department of Pathology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Faried A, Nakajima M, Sohda M, Miyazaki T, Kato H, Kuwano H. Correlation between RhoA overexpression and tumour progression in esophageal squamous cell carcinoma. Eur J Surg Oncol 2005; 31:410-4. [PMID: 15837049 DOI: 10.1016/j.ejso.2004.12.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2004] [Revised: 12/14/2004] [Accepted: 12/20/2004] [Indexed: 11/28/2022] Open
Abstract
AIMS The aim of this study was to clarify the clinico-pathologic outcome and prognostic significance of RhoA in esophageal squamous cell carcinoma (ESCC). METHODS Immunohistochemical staining for RhoA was performed on surgical specimens obtained from 122 patients with ESCC. RESULTS There were significant correlations among RhoA overexpression and TNM clinical classification (depth of invasion, P=0.028; presence of regional lymph node metastasis, P=0.009; presence of distant metastasis, P=0.003; staging P=0.006), lymphatic invasion (P=0.002), and blood-vessel invasion (P=0.004). The five-year survival rates for ESCC patients with RhoA overexpression were significantly lower than those in patients with RhoA under-expression (P=0.002). CONCLUSIONS Our results demonstrated immunohistochemically that the expression of RhoA protein appeared to be correlated with tumour progression of ESCC. Patients with RhoA overexpression tended to have poor prognosis compared with patients with RhoA under-expression.
Collapse
Affiliation(s)
- A Faried
- Department of General Surgical Science (Surgery I), Graduate School of Medicine, Gunma University, Maebashi, Gunma 371-8511, Japan.
| | | | | | | | | | | |
Collapse
|