51
|
Leonard JE, Fisher TL, Winter LA, Cornelius CA, Reilly C, Smith ES, Zauderer M. Nonclinical Safety Evaluation of VX15/2503, a Humanized IgG4 Anti-SEMA4D Antibody. Mol Cancer Ther 2015; 14:964-72. [PMID: 25657333 DOI: 10.1158/1535-7163.mct-14-0924] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/29/2015] [Indexed: 11/16/2022]
Abstract
The humanized IgG4 monoclonal antibody VX15/2503 bound with 1 to 5 nmol/L affinity to purified recombinant semaphorin 4D (SEMA4D; CD100) produced using murine, rat, cynomolgus macaque, and human sequences. The affinity for native SEMA4D expressed on macaque T lymphocytes was approximately 0.6 nmol/L. Tissues from rats and cynomolgus macaques demonstrated specific staining only with resident lymphocytes. Single-dose and one-month toxicology/PK studies used VX15/2503 dose levels of 0 to 100 mg/kg. No toxicity was observed with either species in these studies, thus the no observed adverse effect level (NOAEL) was 100 mg/kg. Cmax, exposure, and half-life values were similar for both rats and macaques. The NOAEL in a primate maximum feasible dose study was 200 mg/kg. Saturation of T-cell-associated SEMA4D occurred following administration of single doses of 0.1 mg/kg and above; five weekly injections of VX15/2503 at a dose level of 100 mg/kg produced saturation lasting for more than 120 and 130 days, respectively, for rats and primates. Macaques administered five weekly doses of VX15/2503 showed dose-dependent reductions of 2- to 3-fold in T-cell SEMA4D (cSEMA4D) expression levels compared with controls. Reduced cSEMA4D expression levels continued until serum antibody concentrations were 2 to 5 μg/mL, and thereafter normal cSEMA4D levels were restored. On the basis of these data, a phase I clinical study of the safety and tolerability of VX15/2503 was conducted, enrolling adult patients with advanced solid tumor diseases; a single-dose, dose escalation, phase I safety study was also initiated with subjects with multiple sclerosis.
Collapse
|
52
|
Smith ES, Jonason A, Reilly C, Veeraraghavan J, Fisher T, Doherty M, Klimatcheva E, Mallow C, Cornelius C, Leonard JE, Marchi N, Janigro D, Argaw AT, Pham T, Seils J, Bussler H, Torno S, Kirk R, Howell A, Evans EE, Paris M, Bowers WJ, John G, Zauderer M. SEMA4D compromises blood–brain barrier, activates microglia, and inhibits remyelination in neurodegenerative disease. Neurobiol Dis 2015; 73:254-68. [DOI: 10.1016/j.nbd.2014.10.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 10/02/2014] [Accepted: 10/12/2014] [Indexed: 10/24/2022] Open
|
53
|
Michaelis UR. Mechanisms of endothelial cell migration. Cell Mol Life Sci 2014; 71:4131-48. [PMID: 25038776 PMCID: PMC11113960 DOI: 10.1007/s00018-014-1678-0] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/23/2014] [Accepted: 07/07/2014] [Indexed: 01/13/2023]
Abstract
Cell migration plays a central role in a variety of physiological and pathological processes during our whole life. Cellular movement is a complex, tightly regulated multistep process. Although the principle mechanisms of migration follow a defined general motility cycle, the cell type and the context of moving influences the detailed mode of migration. Endothelial cells migrate during vasculogenesis and angiogenesis but also in a damaged vessel to restore vessel integrity. Depending on the situation they migrate individually, in chains or sheets and complex signaling, intercellular signals as well as environmental cues modulate the process. Here, the different modes of cell migration, the peculiarities of endothelial cell migration and specific guidance molecules controlling this process will be reviewed.
Collapse
Affiliation(s)
- U Ruth Michaelis
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany,
| |
Collapse
|
54
|
Ruan SS, Li RC, Han Q, Liu J, Li GL, Song YQ, Wu G. Expression and clinical significance of Semaphorin4D in non-small cell lung cancer and its impact on malignant behaviors of A549 lung cancer cells. ACTA ACUST UNITED AC 2014; 34:491-496. [DOI: 10.1007/s11596-014-1304-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 07/04/2014] [Indexed: 01/22/2023]
|
55
|
Treps L, Le Guelte A, Gavard J. Emerging roles of Semaphorins in the regulation of epithelial and endothelial junctions. Tissue Barriers 2014; 1:e23272. [PMID: 24665374 PMCID: PMC3879177 DOI: 10.4161/tisb.23272] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 12/11/2012] [Accepted: 12/13/2012] [Indexed: 12/13/2022] Open
Abstract
Tissue barriers maintain homeostasis, protect underlying tissues, are remodeled during organogenesis and injury and limit aberrant proliferation and dissemination. In this context, endothelial and epithelial intercellular junctions are the primary targets of various cues. This cellular adaptation requires plasticity and dynamics of adhesion molecules and the associated cytoskeleton, as well as the adhesive-linked signaling platforms. It is therefore not surprising that the guidance molecules from the Semaphorin family arise as novel modifiers of epithelia and endothelia in development and diseases. This review will focus on the actions of Semaphorins, and their cognate receptors, Plexins and Neuropilins, on epithelial and endothelial barrier properties.
Collapse
Affiliation(s)
- Lucas Treps
- CNRS; UMR8104; Paris, France ; Inserm; U1016; Paris, France ; Université Paris Descartes; Sorbonne Paris Cite; Paris, France
| | - Armelle Le Guelte
- CNRS; UMR8104; Paris, France ; Inserm; U1016; Paris, France ; Université Paris Descartes; Sorbonne Paris Cite; Paris, France
| | - Julie Gavard
- CNRS; UMR8104; Paris, France ; Inserm; U1016; Paris, France ; Université Paris Descartes; Sorbonne Paris Cite; Paris, France
| |
Collapse
|
56
|
Ito T, Bai T, Tanaka T, Yoshida K, Ueyama T, Miyajima M, Negishi T, Kawasaki T, Takamatsu H, Kikutani H, Kumanogoh A, Yukawa K. Estrogen-dependent proteolytic cleavage of semaphorin 4D and plexin-B1 enhances semaphorin 4D-induced apoptosis during postnatal vaginal remodeling in pubescent mice. PLoS One 2014; 9:e97909. [PMID: 24841081 PMCID: PMC4026538 DOI: 10.1371/journal.pone.0097909] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/25/2014] [Indexed: 01/15/2023] Open
Abstract
Around the fifth week after birth, the vaginal cavity in female mouse pups opens to the overlaying skin. This postnatal tissue remodeling of the genital tract occurs during puberty, and it largely depends upon hormonally induced apoptosis that mainly occurs in the epithelium at the lower part of the mouse vaginal cavity. Previously, we showed that most BALB/c mice lacking the class IV Semaphorin (Sema4D) develop imperforate vagina and hydrometrocolpos; therefore, we reasoned that the absence of Sema4D-induced apoptosis in vaginal epithelial cells may cause the imperforate vagina. Sema4D signals via the Plexin-B1 receptor; nevertheless detailed mechanisms mediating this hormonally triggered apoptosis are not fully documented. To investigate the estrogen-dependent control of Sema4D signaling during the apoptosis responsible for mouse vaginal opening, we examined structural and functional modulation of Sema4D, Plexin-B1, and signaling molecules by analyzing both wild-type and Sema4D−/− mice with or without ovariectomy. Both the release of soluble Sema4D and the conversion of Plexin-B1 by proteolytic processing in vaginal tissue peaked 5 weeks after birth of wild-type BALB/c mice at the time of vaginal opening. Estrogen supplementation of ovariectomized wild-type mice revealed that both the release of soluble Sema4D and the conversion of Plexin-B1 into an active form were estrogen-dependent and concordant with apoptosis. Estrogen supplementation of ovariectomized Sema4D−/− mice did not induce massive vaginal apoptosis in 5-week-old mice; therefore, Sema4D may be an essential apoptosis-inducing ligand that acts downstream of estrogen action in vaginal epithelium during this postnatal tissue remodeling. Analysis of ovariectomized mice also indicated that Sema4D contributed to estrogen-dependent dephosphorylation of Akt and ERK at the time of vaginal opening. Based on our results, we propose that apoptosis in vaginal epithelium during postnatal vaginal opening is induced by enhanced Sema4D signaling that is caused by estrogen-dependent structural changes of Sema4D and Plexin-B1.
Collapse
Affiliation(s)
- Takuji Ito
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Tao Bai
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Tetsuji Tanaka
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Kenji Yoshida
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Takashi Ueyama
- Department of Anatomy and Cell Biology, Wakayama Medical University, Wakayama, Japan
| | - Masayasu Miyajima
- Laboratory Animal Center, Wakayama Medical University, Wakayama, Japan
| | - Takayuki Negishi
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Takahiko Kawasaki
- Division of Brain Function, National Institute of Genetics, Graduate University for Advanced Studies (Sokendai), Mishima, Japan
| | - Hyota Takamatsu
- Department of Immunopathology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Hitoshi Kikutani
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Kazunori Yukawa
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
- * E-mail:
| |
Collapse
|
57
|
Yeh YM, Chen CY, Huang PR, Hsu CW, Wu CC, Wang TCV. Proteomic analyses of genes regulated by heterogeneous nuclear ribonucleoproteins A/B in Jurkat cells. Proteomics 2014; 14:1357-66. [PMID: 24634410 DOI: 10.1002/pmic.201300549] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/20/2014] [Accepted: 03/04/2014] [Indexed: 11/06/2022]
Abstract
Several lines of evidence suggest that hnRNPs A/B (hnRNPs A1, A2/B1, and A3) play an important role in proliferation, although the functional overlap among members of hnRNPs A/B remains largely unknown. In this study, we have employed RNAi knockdown and proteomic approaches to investigate the biological functions of hnRNPs A/B. Depletion of hnRNP A2, but not A1 or A3, produced a significant inhibition of cellular proliferation in Jurkat cells. Analysis of the proteomes in the cells depleted for hnRNP A1, A2, or A3 has identified a total of 167 differentially expressed proteins in the depleted cells. Network analysis of the proteins altered in the cells depleted for hnRNP A2 revealed that the biological processes likely affected by these proteins are related to cell cycle, cytoskeleton rearrangement, and transcription regulation. Indeed, we have confirmed that the level of RhoA and CrkL was selectively reduced in the cells depleted of hnRNP A2, but not in the cells depleted for hnRNP A1 or A3. Therefore, we suggest that the reduced proliferation observed in the cells depleted of hnRNP A2 may result from its effects on cell adhesion processes in the Jurkat cells.
Collapse
Affiliation(s)
- Yuan-Ming Yeh
- Department of Molecular and Cellular Biology, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | | | | | | | | | | |
Collapse
|
58
|
Liu H, Yang Y, Xiao J, Yang S, Liu Y, Kang W, Li X, Zhang F. Semaphorin 4D expression is associated with a poor clinical outcome in cervical cancer patients. Microvasc Res 2014; 93:1-8. [PMID: 24603190 DOI: 10.1016/j.mvr.2014.02.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 02/20/2014] [Accepted: 02/20/2014] [Indexed: 01/13/2023]
Abstract
Lymphangiogenesis is thought to be essential for cancer progression, making it an important target in cancer therapy. Lymphangiogenic factors (VEGF-C and VEGF-D) are upregulated in various tumors/cancers, and play an important role in lymphangiogenesis and lymph node metastasis. Similarly, semaphorin 4D (Sema4D) is a potent inducer of angiogenesis, and its overexpression is associated with tumor progression and poor prognosis in a variety of malignancies. However, little is known regarding the functional relationship between Sema4D and VEGF-C/VEGF-D and in the mediation of lymphangiogenesis and lymph node metastasis and clinical outcome. The current study aimed to evaluate the effect of Sema4D expression on outcome in patients with cervical cancer, and to explore the molecular mechanism of Sema4D in tumor progression. We evaluated Sema4D expression, density of lymphatic vessels, and invasion of lymphatic vessels with immunohistochemical methods in 232 human cervical cancers with long-term follow-up. Sema4D expression was correlated with patho-clinical parameters and patients' outcome. A cervical cancer cell line was used to investigate the contribution of sema4D to tumor progression by studying the role of Sema4D in VEGF-C/-D and cell migration using reverse transcription-polymerase chain reaction and Western blotting. We observed that Sema4D expression was higher in metastatic cervical cancer than in nonmetastatic cervical cancer (P<0.001). CD34-positive or D2-40-positive lymphatic vessel density was significantly increased in cases with lymph node metastasis compared with those without lymph node metastasis. The increased Sema4D expression was associated with VEGF-C/-D, the presence of lymphatic invasion, the occurrence of lymph node metastasis, and FIGO stage. We also observed a novel association between Sema4D upregulation and poor prognosis in cervical cancer. In vitro, the Sema4D inhibitory antibody and Sema4D-shRNA suppressed VEGF-C and VEGF-D in the human cervical carcinoma cell lines HeLa, Siha, and Caski cells. Invasiveness assay demonstrated that Sema4D could augment the invasive potential of the tumor cells in the cervical cancer lines and induction of cellular invasiveness by Sema4D stimulation could be inhibited by knockdown of plexinB1 by siRNA. Further mechanistic investigations of tumor cell invasiveness showed that Sema4D could induce activation of GTPase Ras homolog gene family, member A (RhoA), MAPK and AKT. In addition, plexinB1 knockdown by siRNA could suppress the Sema4D signal transmitted to MAPK and Akt. Taken together, these results suggest that Sema4D autocrine within tumor cells contributes to enhanced invasion and tumor progression through increased motility of cervical cancer and VEGF-C/-D-mediated lymphangiogenesis. Sema4D might be useful as a molecular marker of poor prognosis in cervical cancer.
Collapse
Affiliation(s)
- Huidong Liu
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Yanmei Yang
- Cancer Institute, Harbin Medical University, 150 Harping Road, 150081 Harbin, China
| | - Jianbing Xiao
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Shucai Yang
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Yan Liu
- Department of General Surgery, Mudanjiang First Hospital, 4 Jianwei Road, 157011 Mudanjiang, China
| | - Wenhui Kang
- Department of Radiotherapy, The Third Affiliated Hospital of Harbin Medical University, 150 Harping Road, 150081 Harbin, China
| | - Xinlei Li
- Department of Anatomy, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China
| | - Fengmin Zhang
- Department of Medical Microbiology, Heilongjiang Province Key Laboratory for Immunity and Infection, Harbin Medical University, 194 Xuefu Road, 150081 Harbin, China.
| |
Collapse
|
59
|
Azzarelli R, Pacary E, Garg R, Garcez P, van den Berg D, Riou P, Ridley AJ, Friedel RH, Parsons M, Guillemot F. An antagonistic interaction between PlexinB2 and Rnd3 controls RhoA activity and cortical neuron migration. Nat Commun 2014; 5:3405. [PMID: 24572910 PMCID: PMC3939360 DOI: 10.1038/ncomms4405] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 02/06/2014] [Indexed: 02/06/2023] Open
Abstract
A transcriptional programme initiated by the proneural factors Neurog2 and Ascl1 controls successive steps of neurogenesis in the embryonic cerebral cortex. Previous work has shown that proneural factors also confer a migratory behaviour to cortical neurons by inducing the expression of the small GTP-binding proteins such as Rnd2 and Rnd3. However, the directionality of radial migration suggests that migrating neurons also respond to extracellular signal-regulated pathways. Here we show that the Plexin B2 receptor interacts physically and functionally with Rnd3 and stimulates RhoA activity in migrating cortical neurons. Plexin B2 competes with p190RhoGAP for binding to Rnd3, thus blocking the Rnd3-mediated inhibition of RhoA and also recruits RhoGEFs to directly stimulate RhoA activity. Thus, an interaction between the cell-extrinsic Plexin signalling pathway and the cell-intrinsic Ascl1-Rnd3 pathway determines the level of RhoA activity appropriate for cortical neuron migration.
Collapse
Affiliation(s)
- Roberta Azzarelli
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
- Present address: Hutchison/MRC Research Centre, University of Cambridge, Box 197, Biomedical Campus, Cambridge CB2 0XZ, UK
| | - Emilie Pacary
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
- Present address: INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, Bordeaux F-33000, France or University Bordeaux, Neurocentre Magendie, Physiopathologie de la plasticité neuronale, U862, Bordeaux F-33000, France
| | - Ritu Garg
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Patricia Garcez
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Debbie van den Berg
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Philippe Riou
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
- Present address: Protein Phosphorylation Laboratory, Cancer Research UK, London Research Institute, Lincoln's Inn Fields Laboratories, London WC2A 3LY, UK
| | - Anne J. Ridley
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Roland H. Friedel
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, New York 10029, USA
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - François Guillemot
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| |
Collapse
|
60
|
The Semaphorin 4D-Plexin-B1-RhoA signaling axis recruits pericytes and regulates vascular permeability through endothelial production of PDGF-B and ANGPTL4. Angiogenesis 2013; 17:261-74. [PMID: 24114199 DOI: 10.1007/s10456-013-9395-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/01/2013] [Indexed: 01/13/2023]
Abstract
Semaphorin 4D (SEMA4D) is a member of a family of transmembrane and secreted proteins that have been shown to act through its receptor Plexin-B1 to regulate axon growth cone guidance, lymphocyte activation, and bone density. SEMA4D is also overexpressed by some malignancies and plays a role in tumor-induced angiogenesis similar to vascular endothelial growth factor (VEGF), a protein that has been targeted as part of some cancer therapies. In an attempt to examine the different effects on tumor growth and vascularity for these two pro-angiogenic factors, we previously noted that while inhibition of both VEGF and SEMA4D restricted tumor vascularity and size, vessels forming under conditions of VEGF blockade retained their association with pericytes while those arising in a background of SEMA4D/Plexin-B1 deficiency did not, an intriguing finding considering that alteration in pericyte association with endothelial cells is an emerging aspect of anti-angiogenic intervention in the treatment of cancer. Here we show through array analysis, immunoblots, migration and co-culture assays and VE-cadherin immunohistochemistry that SEMA4D production by head and neck carcinoma tumor cells induces expression of platelet-derived growth factor-B and angiopoietin-like protein 4 from endothelial cells in a Plexin-B1/Rho-dependent manner, thereby influencing proliferation and differentiation of pericytes and vascular permeability, whereas VEGF lacks these effects. These results partly explain the differences observed between SEMA4D and VEGF in pathological angiogenesis and suggest that targeting SEMA4D function along with VEGF could represent a novel anti-angiogenic therapeutic strategy for the treatment of solid tumors.
Collapse
|
61
|
Li F, Jiang Q, Shi KJ, Luo H, Yang Y, Xu CM. RhoA modulates functional and physical interaction between ROCK1 and Erk1/2 in selenite-induced apoptosis of leukaemia cells. Cell Death Dis 2013; 4:e708. [PMID: 23828571 PMCID: PMC3730416 DOI: 10.1038/cddis.2013.243] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 05/08/2013] [Accepted: 05/30/2013] [Indexed: 12/19/2022]
Abstract
RhoA GTPase dysregulation is frequently reported in various tumours and haematologic malignancies. RhoA, regulating Rho-associated coiled-coil-forming kinase 1 (ROCK1), modulates multiple cell functions, including malignant transformation, metastasis and cell death. Therefore, RhoA/ROCK1 could be an ideal candidate target in cancer treatment. However, the roles of RhoA/ROCK1 axis in apoptosis of leukaemia cells remain elusive. In this study, we explored the effects of RhoA/ROCK1 cascade on selenite-induced apoptosis of leukaemia cells and the underlying mechanism. We found selenite deactivated RhoA/ROCK1 and decreased the association between RhoA and ROCK1 in leukaemia NB4 and Jurkat cells. The inhibited RhoA/ROCK1 signalling enhanced the phosphorylation of Erk1/2 in a Mek1/2-independent manner. Erk1/2 promoted apoptosis of leukaemia cells after it was activated. Intriguingly, it was shown that both RhoA and ROCK1 were present in the multimolecular complex containing Erk1/2. GST pull-down analysis showed ROCK1 had a direct interaction with GST-Erk2. In addition, selenite-induced apoptosis in an NB4 xenograft model was also found to be associated with the RhoA/ROCK1/Erk1/2 pathway. Our data demonstrate that the RhoA/ROCK1 signalling pathway has important roles in the determination of cell fates and the modulation of Erk1/2 activity at the Mek–Erk interplay level.
Collapse
Affiliation(s)
- F Li
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medicine Sciences & School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | |
Collapse
|
62
|
Bird E, Smith PL, Seiffert D, Psaltis GC, Huang J, Schumacher WA. Does plexin-B1, a semaphorin 4D receptor, play a role in thrombosis? Thromb Res 2013; 131:564-5. [PMID: 23642655 DOI: 10.1016/j.thromres.2013.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 11/19/2022]
|
63
|
Kirsch T, Beese M, Wyss K, Klinge U, Haller H, Haubitz M, Fiebeler A. Aldosterone Modulates Endothelial Permeability and Endothelial Nitric Oxide Synthase Activity by Rearrangement of the Actin Cytoskeleton. Hypertension 2013; 61:501-8. [DOI: 10.1161/hypertensionaha.111.196832] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Torsten Kirsch
- From the Division of Nephrology, Hannover Medical School, Hannover, Germany (T.K., M.B., K.W., H.H., M.H., A.F.); Experimental and Clinical Research Center, Charité Medical Faculty and Max-Delbruck Center for Molecular Medicine, Berlin, Germany (A.F.); Department of Surgery, University Hospital RWTH, Aachen, Germany (U.K.)
| | - Michaela Beese
- From the Division of Nephrology, Hannover Medical School, Hannover, Germany (T.K., M.B., K.W., H.H., M.H., A.F.); Experimental and Clinical Research Center, Charité Medical Faculty and Max-Delbruck Center for Molecular Medicine, Berlin, Germany (A.F.); Department of Surgery, University Hospital RWTH, Aachen, Germany (U.K.)
| | - Kristin Wyss
- From the Division of Nephrology, Hannover Medical School, Hannover, Germany (T.K., M.B., K.W., H.H., M.H., A.F.); Experimental and Clinical Research Center, Charité Medical Faculty and Max-Delbruck Center for Molecular Medicine, Berlin, Germany (A.F.); Department of Surgery, University Hospital RWTH, Aachen, Germany (U.K.)
| | - Uwe Klinge
- From the Division of Nephrology, Hannover Medical School, Hannover, Germany (T.K., M.B., K.W., H.H., M.H., A.F.); Experimental and Clinical Research Center, Charité Medical Faculty and Max-Delbruck Center for Molecular Medicine, Berlin, Germany (A.F.); Department of Surgery, University Hospital RWTH, Aachen, Germany (U.K.)
| | - Hermann Haller
- From the Division of Nephrology, Hannover Medical School, Hannover, Germany (T.K., M.B., K.W., H.H., M.H., A.F.); Experimental and Clinical Research Center, Charité Medical Faculty and Max-Delbruck Center for Molecular Medicine, Berlin, Germany (A.F.); Department of Surgery, University Hospital RWTH, Aachen, Germany (U.K.)
| | - Marion Haubitz
- From the Division of Nephrology, Hannover Medical School, Hannover, Germany (T.K., M.B., K.W., H.H., M.H., A.F.); Experimental and Clinical Research Center, Charité Medical Faculty and Max-Delbruck Center for Molecular Medicine, Berlin, Germany (A.F.); Department of Surgery, University Hospital RWTH, Aachen, Germany (U.K.)
| | - Anette Fiebeler
- From the Division of Nephrology, Hannover Medical School, Hannover, Germany (T.K., M.B., K.W., H.H., M.H., A.F.); Experimental and Clinical Research Center, Charité Medical Faculty and Max-Delbruck Center for Molecular Medicine, Berlin, Germany (A.F.); Department of Surgery, University Hospital RWTH, Aachen, Germany (U.K.)
| |
Collapse
|
64
|
Mangialardi G, Katare R, Oikawa A, Meloni M, Reni C, Emanueli C, Madeddu P. Diabetes causes bone marrow endothelial barrier dysfunction by activation of the RhoA-Rho-associated kinase signaling pathway. Arterioscler Thromb Vasc Biol 2013; 33:555-64. [PMID: 23307872 DOI: 10.1161/atvbaha.112.300424] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Diabetes mellitus causes bone marrow (BM) microangiopathy. This study aimed to investigate the mechanisms responsible for BM endothelial dysfunction in diabetes mellitus. METHODS AND RESULTS The analysis of differentially expressed transcripts in BM endothelial cells (BMECs) from type-1 diabetic and nondiabetic mice showed an effect of diabetes mellitus on signaling pathways controlling cell death, migration, and cytoskeletal rearrangement. Type-1 diabetic-BMECs displayed high reactive oxygen species levels, increased expression and activity of RhoA and its associated protein kinases Rho-associated kinase 1/Rho-associated kinase 2, and reduced Akt phosphorylation/activity. Likewise, diabetes mellitus impaired Akt-related BMEC functions, such as migration, network formation, and angiocrine factor-releasing activity, and increased vascular permeability. Moreover, high glucose disrupted BMEC contacts through Src tyrosine kinase phosphorylation of vascular endothelial cadherin. These alterations were prevented by constitutively active Akt (myristoylated Akt), Rho-associated kinase inhibitor Y-27632, and Src inhibitors. Insulin replacement restored BMEC abundance, as assessed by flow cytometry analysis of the endothelial marker MECA32, and endothelial barrier function in BM of type-1 diabetic mice. CONCLUSIONS Redox-dependent activation of RhoA/Rho-associated kinase and Src/vascular endothelial cadherin signaling pathways, together with Akt inactivation, contribute to endothelial dysfunction in diabetic BM. Metabolic control is crucial for maintenance of endothelial cell homeostasis and endothelial barrier function in BM of diabetic mice.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- Experimental Cardiovascular Medicine, Regenerative Medicine Section Bristol Heart Institute, School of Clinical Sciences University of Bristol, Bristol, UK
| | | | | | | | | | | | | |
Collapse
|
65
|
Bijli KM, Fazal F, Rahman A. Regulation of Rela/p65 and endothelial cell inflammation by proline-rich tyrosine kinase 2. Am J Respir Cell Mol Biol 2012; 47:660-8. [PMID: 22842493 PMCID: PMC3547104 DOI: 10.1165/rcmb.2012-0047oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 07/16/2012] [Indexed: 01/01/2023] Open
Abstract
We investigated the role of proline-rich tyrosine kinase 2 (Pyk2) in the mechanism of NF-κB activation and endothelial cell (EC) inflammation induced by thrombin, a procoagulant serine protease released in high amounts during sepsis and other inflammatory conditions. Stimulation of ECs with thrombin resulted in a time-dependent activation of Pyk2. RNA interference knockdown of Pyk2 attenuated thrombin-induced activity of NF-κB and expression of its target genes, vascular cell adhesion molecule-1 and monocyte chemoattractant protein-1. Pyk2 knockdown impaired thrombin-induced activation of IκB kinase (IKK) and phosphorylation (Ser32 and Ser36) of IkappaBα, but, surprisingly, failed to prevent IκBα degradation. However, depletion of IKKα or IKKβ was effective in inhibiting IκBα phosphorylation/degradation, as expected. Intriguingly, Pyk2 knockdown impaired nuclear translocation and DNA binding of RelA/p65, despite the inability to prevent IκBα degradation. In addition, Pyk2 knockdown was associated with inhibition of RelA/p65 phosphorylation at Ser536, which is important for transcriptional activity of RelA/p65. Depletion of IKKα or IKKβ each impaired RelA/p65 phosphorylation. Taken together, these data identify Pyk2 as a critical regulator of EC inflammation by virtue of engaging IKK to promote the release and the transcriptional capacity of RelA/p65, and, additionally, by its ability to facilitate the nuclear translocation of the released RelA/p65. Thus, specific targeting of Pyk2 may be an effective anti-inflammatory strategy in vascular diseases associated with EC inflammation and intravascular coagulation.
Collapse
Affiliation(s)
- Kaiser M Bijli
- Department of Pediatrics (Neonatology), Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Fabeha Fazal
- Department of Pediatrics (Neonatology), Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Arshad Rahman
- Department of Pediatrics (Neonatology), Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York
| |
Collapse
|
66
|
Hota PK, Buck M. Plexin structures are coming: opportunities for multilevel investigations of semaphorin guidance receptors, their cell signaling mechanisms, and functions. Cell Mol Life Sci 2012; 69:3765-805. [PMID: 22744749 PMCID: PMC11115013 DOI: 10.1007/s00018-012-1019-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 04/09/2012] [Accepted: 04/11/2012] [Indexed: 01/13/2023]
Abstract
Plexin transmembrane receptors and their semaphorin ligands, as well as their co-receptors (Neuropilin, Integrin, VEGFR2, ErbB2, and Met kinase) are emerging as key regulatory proteins in a wide variety of developmental, regenerative, but also pathological processes. The diverse arenas of plexin function are surveyed, including roles in the nervous, cardiovascular, bone and skeletal, and immune systems. Such different settings require considerable specificity among the plexin and semaphorin family members which in turn are accompanied by a variety of cell signaling networks. Underlying the latter are the mechanistic details of the interactions and catalytic events at the molecular level. Very recently, dramatic progress has been made in solving the structures of plexins and of their complexes with associated proteins. This molecular level information is now suggesting detailed mechanisms for the function of both the extracellular as well as the intracellular plexin regions. Specifically, several groups have solved structures for extracellular domains for plexin-A2, -B1, and -C1, many in complex with semaphorin ligands. On the intracellular side, the role of small Rho GTPases has been of particular interest. These directly associate with plexin and stimulate a GTPase activating (GAP) function in the plexin catalytic domain to downregulate Ras GTPases. Structures for the Rho GTPase binding domains have been presented for several plexins, some with Rnd1 bound. The entire intracellular domain structure of plexin-A1, -A3, and -B1 have also been solved alone and in complex with Rac1. However, key aspects of the interplay between GTPases and plexins remain far from clear. The structural information is helping the plexin field to focus on key questions at the protein structural, cellular, as well as organism level that collaboratoria of investigations are likely to answer.
Collapse
Affiliation(s)
- Prasanta K. Hota
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
| | - Matthias Buck
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Department of Neuroscience, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Department of Pharmacology, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Comprehensive Cancer Center, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
| |
Collapse
|
67
|
Abstract
Solid tumors not only comprise malignant cells but also other nonmalignant cell types, forming a unique microenvironment that can strongly influence the behavior of tumor cells. Recent advances in the understanding of cancer biology have highlighted the functional role of semaphorins. In fact, semaphorins form a family of molecular signals known to guide and control cell migration during embryo development and in adults. Tumor cells express semaphorins as well as their receptors, plexins and neuropilins. It has been shown that semaphorin signaling can regulate tumor cell behavior. Moreover, semaphorins are important regulators of tumor angiogenesis. Conversely, very little is known about the functional relevance of semaphorin signals for tumor-infiltrating stromal cells, such as leukocytes. In this chapter, we review the current knowledge on the functional role of semaphorins in cancer progression, and we focus on the emerging role of semaphorins in mediating the cross talk between tumor cells and different tumor stromal cells.
Collapse
Affiliation(s)
- Claudia Muratori
- University of Torino Medical School, Institute for Cancer Research (IRCC), Candiolo, Turin, Italy
| | | |
Collapse
|
68
|
Huang YS, Cheng CY, Chueh SH, Hueng DY, Huang YF, Chu CM, Wu ST, Tai MC, Liang CM, Liao MH, Chen CC, Shen LH, Ma KH. Involvement of SHP2 in focal adhesion, migration and differentiation of neural stem cells. Brain Dev 2012; 34:674-84. [PMID: 22118986 DOI: 10.1016/j.braindev.2011.10.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 10/10/2011] [Accepted: 10/26/2011] [Indexed: 11/25/2022]
Abstract
OBJECTIVES SHP2 (Src-homology-2 domain-containing protein tyrosine phosphatase) plays an important role in cell adhesion, migration and cell signaling. However, its role in focal adhesion, differentiation and migration of neural stem cells is still unclear. METHODS In this study, rat neurospheres were cultured in suspension and differentiated neural stem cells were cultured on collagen-coated surfaces. RESULTS The results showed that p-SHP2 co-localized with focal adhesion kinase (FAK) and paxillin in neurospheres and in differentiated neural precursor cells, astrocytes, neurons, and oligodendrocytes. Suppression of SHP2 activity by PTP4 or siRNA-mediated SHP2 silencing caused reduction in the cell migration and neurite outgrowth, and thinning of glial cell processes. Differentiation-induced activation of FAK, Src, paxillin, ERK1/2, and RhoA was decreased by SHP2 inactivation. CONCLUSIONS These results indicate that SHP2 is recruited in focal adhesions of neural stem cells and regulates focal adhesion formation. SHP2-mediated regulation of neural differentiation and migration may be related to formation of focal adhesions and RhoA and ERK1/2 activation.
Collapse
Affiliation(s)
- Yuahn-Sieh Huang
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Witherden DA, Watanabe M, Garijo O, Rieder SE, Sarkisyan G, Cronin SJF, Verdino P, Wilson IA, Kumanogoh A, Kikutani H, Teyton L, Fischer WH, Havran WL. The CD100 receptor interacts with its plexin B2 ligand to regulate epidermal γδ T cell function. Immunity 2012; 37:314-25. [PMID: 22902232 DOI: 10.1016/j.immuni.2012.05.026] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Revised: 05/02/2012] [Accepted: 05/15/2012] [Indexed: 01/15/2023]
Abstract
γδ T cells respond rapidly to keratinocyte damage, providing essential contributions to the skin wound healing process. The molecular interactions regulating their response are unknown. Here, we identify a role for interaction of plexin B2 with the CD100 receptor in epithelial repair. In vitro blocking of plexin B2 or CD100 inhibited γδ T cell activation. Furthermore, CD100 deficiency in vivo resulted in delayed repair of cutaneous wounds due to a disrupted γδ T cell response to keratinocyte damage. Ligation of CD100 in γδ T cells induced cellular rounding via signals through ERK kinase and cofilin. Defects in this rounding process were evident in the absence of CD100-mediated signals, thereby providing a mechanistic explanation for the defective wound healing in CD100-deficient animals. The discovery of immune functions for plexin B2 and CD100 provides insight into the complex cell-cell interactions between epithelial resident γδ T cells and the neighboring cells they support.
Collapse
Affiliation(s)
- Deborah A Witherden
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Binmadi NO, Yang YH, Zhou H, Proia P, Lin YL, Batista De Paula AM, Sena Guimarães AL, Poswar FO, Sundararajan D, Basile JR. Plexin-B1 and semaphorin 4D cooperate to promote perineural invasion in a RhoA/ROK-dependent manner. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1232-1242. [PMID: 22252234 DOI: 10.1016/j.ajpath.2011.12.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 11/07/2011] [Accepted: 12/02/2011] [Indexed: 01/15/2023]
Abstract
Perineural invasion (PNI) is a tropism of tumor cells for nerve bundles located in the surrounding stroma. It is a pathological feature observed in certain tumors, referred to as neurotropic malignancies, that severely limits the ability to establish local control of disease and results in pain, recurrent growth, and distant metastases. Despite the importance of PNI as a prognostic indicator, its biological mechanisms are poorly understood. The semaphorins and their receptors, the plexins, compose a family of proteins originally shown to be important in nerve cell adhesion, axon migration, and proper central nervous system development. Emerging evidence has demonstrated that these factors are expressed in tissues outside of the nervous system and represent a widespread signal transduction system that is involved in the regulation of motility and adhesion in different cell types. We believe that the plexins and semaphorins, which are strongly expressed in both axons and many carcinomas, play a role in PNI. In this study, we show that plexin-B1 is overexpressed in tissues and cell lines from neurotropic malignancies and is attracted to nerves that express its ligand, semaphorin 4D, in a Rho/Rho kinase-dependent manner. We also demonstrate that nerves are attracted to tumors through this same system of proteins, suggesting that both plexin-B1 and semaphorin 4D are important in the promotion of PNI.
Collapse
Affiliation(s)
- Nada O Binmadi
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland; Department of Oral, Basic, and Clinical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ying-Hua Yang
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland
| | - Hua Zhou
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland
| | - Patrizia Proia
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland; Department of Sports Science, University of Palermo, Palermo, Italy
| | - Yi-Ling Lin
- Department of Diagnostic and Surgical Sciences, University of California at Los Angeles, School of Dentistry, Los Angeles, California
| | - Alfredo M Batista De Paula
- Health Science Program, Department of Dentistry, State University of Montes Claros, Minas Gerais, Brazil
| | - André L Sena Guimarães
- Health Science Program, Department of Dentistry, State University of Montes Claros, Minas Gerais, Brazil
| | - Fabiano O Poswar
- Health Science Program, Department of Medicine, State University of Montes Claros, Minas Gerais, Brazil
| | - Devaki Sundararajan
- Department of Oral and Maxillofacial Pathology, Boston University Goldman School of Dental Medicine, Boston, Massachusetts
| | - John R Basile
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland; Marlene and Stuart Greenebaum Cancer Center, Baltimore, Maryland.
| |
Collapse
|
71
|
Neufeld G, Sabag AD, Rabinovicz N, Kessler O. Semaphorins in angiogenesis and tumor progression. Cold Spring Harb Perspect Med 2012; 2:a006718. [PMID: 22315716 PMCID: PMC3253028 DOI: 10.1101/cshperspect.a006718] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The semaphorins were initially described as axon guidance factors, but have recently been implicated in a variety of physiological and developmental functions, including regulation of immune response, angiogenesis, and migration of neural crest cells. The semaphorin family contains more than 30 genes divided into seven subfamilies, all of which are characterized by the presence of a sema domain. The semaphorins transduce their signals by binding to one of the nine receptors belonging to the plexin family, or, in the case of the class 3 semaphorins, by binding to one of the two neuropilin receptors. Additional receptors, which form complexes with these primary semaphorin receptors, are also frequently involved in semaphorin signaling. Recent evidence suggests that some semaphorins can act as antiangiogenic and/or antitumorigenic agents whereas other semaphorins promote tumor progression and/or angiogenesis. Furthermore, loss of endogenous inhibitory semaphorin expression or function on one hand, and overexpression of protumorigenic semaphorins on the other hand, is associated with the progression of some tumor types.
Collapse
Affiliation(s)
- Gera Neufeld
- Cancer and Vascular Biology Research Center, Rappaport Research Institute in the Medical Sciences, Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 31096, Israel.
| | | | | | | |
Collapse
|
72
|
Sema4D, the ligand for Plexin B1, suppresses c-Met activation and migration and promotes melanocyte survival and growth. J Invest Dermatol 2011; 132:1230-8. [PMID: 22189792 PMCID: PMC3305852 DOI: 10.1038/jid.2011.414] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Semaphorins are secreted and membrane bound proteins involved in neural pathfinding, organogenesis, and tumor progression, through Plexin and neuropilins receptors. We recently reported that Plexin B1, the Semaphorin 4D receptor, is a tumor suppressor protein for melanoma, in part, through inhibition of the oncogenic c-Met tyrosine kinase receptor. In this report we show that Sema4D is a protective paracrine factor for normal human melanocyte survival in response to ultraviolet irradiation, that it stimulates proliferation, and regulates the activity of the c-Met receptor. c-Met receptor signaling stimulates melanocyte migration, in part through down-regulation of the cell adhesion molecule E-cadherin. Sema4D suppressed activation of c-Met in response to its ligand hepatocyte growth factor (HGF), and partially blocked the suppressive effects of HGF on E-cadherin expression in melanocytes and HGF-dependent migration. These data demonstrate a role for Plexin B1 in maintenance of melanocyte survival and proliferation in the skin, and suggest that Semaphorin 4D and Plexin B1 act cooperatively with HGF and c-Met to regulate c-Met dependent effects in human melanocytes. Because our data show that Plexin B1 is profoundly down-regulated by UVB in melanocytes, loss of Plexin B1 may accentuate HGF dependent effects on melanocytes, including melanocyte migration.
Collapse
|
73
|
Abstract
Angiogenesis, the formation of new blood vessels from preexisting vasculature, is essential for many physiological processes, and aberrant angiogenesis contributes to some of the most prevalent human diseases, including cancer. Angiogenesis is controlled by delicate balance between pro- and anti-angiogenic signals. While pro-angiogenic signaling has been extensively investigated, how developmentally regulated, naturally occurring anti-angiogenic molecules prevent the excessive growth of vascular and lymphatic vessels is still poorly understood. In this review, we summarize the current knowledge on how semaphorins and their receptors, plexins and neuropilins, control normal and pathological angiogenesis, with an emphasis on semaphorin-regulated anti-angiogenic signaling circuitries in vascular and lymphatic endothelial cells. This emerging body of information may afford the opportunity to develop novel anti-angiogenic therapeutic strategies.
Collapse
|
74
|
Plank C, Zelphati O, Mykhaylyk O. Magnetically enhanced nucleic acid delivery. Ten years of magnetofection-progress and prospects. Adv Drug Deliv Rev 2011; 63:1300-31. [PMID: 21893135 PMCID: PMC7103316 DOI: 10.1016/j.addr.2011.08.002] [Citation(s) in RCA: 204] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 08/18/2011] [Accepted: 08/19/2011] [Indexed: 12/28/2022]
Abstract
Nucleic acids carry the building plans of living systems. As such, they can be exploited to make cells produce a desired protein, or to shut down the expression of endogenous genes or even to repair defective genes. Hence, nucleic acids are unique substances for research and therapy. To exploit their potential, they need to be delivered into cells which can be a challenging task in many respects. During the last decade, nanomagnetic methods for delivering and targeting nucleic acids have been developed, methods which are often referred to as magnetofection. In this review we summarize the progress and achievements in this field of research. We discuss magnetic formulations of vectors for nucleic acid delivery and their characterization, mechanisms of magnetofection, and the application of magnetofection in viral and nonviral nucleic acid delivery in cell culture and in animal models. We summarize results that have been obtained with using magnetofection in basic research and in preclinical animal models. Finally, we describe some of our recent work and end with some conclusions and perspectives.
Collapse
|
75
|
Yang YH, Zhou H, Binmadi NO, Proia P, Basile JR. Plexin-B1 activates NF-κB and IL-8 to promote a pro-angiogenic response in endothelial cells. PLoS One 2011; 6:e25826. [PMID: 22028792 PMCID: PMC3196529 DOI: 10.1371/journal.pone.0025826] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 09/11/2011] [Indexed: 12/13/2022] Open
Abstract
Background The semaphorins and their receptors, the plexins, are proteins related to c-Met and the scatter factors that have been implicated in an expanding signal transduction network involving co-receptors, RhoA and Ras activation and deactivation, and phosphorylation events. Our previous work has demonstrated that Semaphorin 4D (Sema4D) acts through its receptor, Plexin-B1, on endothelial cells to promote angiogenesis in a RhoA and Akt-dependent manner. Since NF-κB has been linked to promotion of angiogenesis and can be activated by Akt in some contexts, we wanted to examine NF-κB in Sema4D treated cells to determine if there was biological significance for the pro-angiogenic phenotype observed in endothelium. Methods/Principal Findings Using RNA interference techniques, gel shifts and NF-κB reporter assays, we demonstrated NF-κB translocation to the nucleus in Sema4D treated endothelial cells occurring downstream of Plexin-B1. This response was necessary for endothelial cell migration and capillary tube formation and protected endothelial cells against apoptosis as well, but had no effect on cell proliferation. We dissected Plexin-B1 signaling with chimeric receptor constructs and discovered that the ability to activate NF-κB was dependent upon Plexin-B1 acting through Rho and Akt, but did not involve its role as a Ras inhibitor. Indeed, inhibition of Rho by C3 toxin and Akt by LY294002 blocked Sema4D-mediated endothelial cell migration and tubulogenesis. We also observed that Sema4D treatment of endothelial cells induced production of the NF-κB downstream target IL-8, a response necessary for angiogenesis. Finally, we could show through co-immunofluorescence for p65 and CD31 that Sema4D produced by tumor xenografts in nude mice activated NF-κB in vessels of the tumor stroma. Conclusion/Significance These findings provide evidence that Sema4D/Plexin-B1-mediated NF-κB activation and IL-8 production is critical in the generation a pro-angiogenic phenotype in endothelial cells and suggests a new therapeutic target for the anti-angiogenic treatment of some cancers.
Collapse
Affiliation(s)
- Ying-Hua Yang
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Hua Zhou
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Nada O. Binmadi
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Patrizia Proia
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America
- Department of Sports Science (DISMOT), University of Palermo, Palermo, Italy
| | - John R. Basile
- Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America
- Marlene and Stuart Greenebaum Cancer Center, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
76
|
Kato S, Kubota K, Shimamura T, Shinohara Y, Kobayashi N, Watanabe S, Yoneda M, Inamori M, Nakamura F, Ishiguro H, Nakaigawa N, Nagashima Y, Taguri M, Kubota Y, Goshima Y, Morita S, Endo I, Maeda S, Nakajima A, Nakagama H. Semaphorin 4D, a lymphocyte semaphorin, enhances tumor cell motility through binding its receptor, plexinB1, in pancreatic cancer. Cancer Sci 2011; 102:2029-37. [PMID: 21812859 DOI: 10.1111/j.1349-7006.2011.02053.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor, for which the development of new biomarkers and therapeutic targets has become critical. The main cause of poor prognosis in PDAC patients is the high invasive and metastatic potential of the cancer. In the present study, we report a new signaling pathway that was found to mediate the enhanced tumor cell motility in pancreatic cancer. Semaphorin 4D (Sema4D) is a ligand known to be expressed on different cell types, and has been reported to be involved in the regulation of immune functions, epithelial morphogenesis, and tumor growth and metastasis. In this study, we revealed for the first time that the cancer tissue cells expressing Sema4D in PDAC are tumor-infiltrating lymphocytes. The overexpression of Sema4D and of its receptor, plexinB1, was found to be significantly correlated with clinical factors, such as lymph node metastasis, distant metastasis, and poor prognosis in patients with PDAC. Through in vitro analysis, we demonstrated that Sema4D can potentiate the invasiveness of pancreatic cancer cells and we identified the downstream molecules. The binding of Sema4D to plexinB1 induced small GTPase Ras homolog gene family, member A activation and resulted in the phosphorylation of MAPK and Akt. In addition, in terms of potential therapeutic application, we clearly demonstrated that the enhanced-cell invasiveness induced by Sema4D could be inhibited by knockdown of plexinB1, suggesting that blockade of plexinB1 might diminish the invasive potential of pancreatic cancer cells. Our findings provide new insight into possible prognostic biomarkers and therapeutic targets in PDAC patients.
Collapse
Affiliation(s)
- Shingo Kato
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Chaturvedi LS, Marsh HM, Basson MD. Role of RhoA and its effectors ROCK and mDia1 in the modulation of deformation-induced FAK, ERK, p38, and MLC motogenic signals in human Caco-2 intestinal epithelial cells. Am J Physiol Cell Physiol 2011; 301:C1224-38. [PMID: 21849669 DOI: 10.1152/ajpcell.00518.2010] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Repetitive deformation enhances intestinal epithelial migration across tissue fibronectin. We evaluated the contribution of RhoA and its effectors Rho-associated kinase (ROK/ROCK) and mammalian diaphanous formins (mDia1) to deformation-induced intestinal epithelial motility across fibronectin and the responsible focal adhesion kinase (FAK), extracellular signal-regulated kinase (ERK), p38, and myosin light chain (MLC) signaling. We reduced RhoA, ROCK1, ROCK2, and mDia1 by smart-pool double-stranded short-interfering RNAs (siRNA) and pharmacologically inhibited RhoA, ROCK, and FAK in human Caco-2 intestinal epithelial monolayers on fibronectin-coated membranes subjected to 10% repetitive deformation at 10 cycles/min. Migration was measured by wound closure. Stimulation of migration by deformation was prevented by exoenzyme C3, Y27632, or selective RhoA, ROCK1, and ROCK2 or mDia1 siRNAs. RhoA, ROCK inhibition, or RhoA, ROCK1, ROCK2, mDia1, and FAK reduction by siRNA blocked deformation-induced nuclear ERK phosphorylation without preventing ERK phosphorylation in the cytoplasmic protein fraction. Furthermore, RhoA, ROCK inhibition or RhoA, ROCK1, ROCK2, and mDia1 reduction by siRNA also blocked strain-induced FAK-Tyr(925), p38, and MLC phosphorylation. These results suggest that RhoA, ROCK, mDia1, FAK, ERK, p38, and MLC all mediate the stimulation of intestinal epithelial migration by repetitive deformation. This pathway may be an important target for interventions to promote mechanotransduced mucosal healing during inflammation.
Collapse
|
78
|
Rho-mediated activation of PI(4)P5K and lipid second messengers is necessary for promotion of angiogenesis by Semaphorin 4D. Angiogenesis 2011; 14:309-19. [PMID: 21538148 DOI: 10.1007/s10456-011-9214-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 04/18/2011] [Indexed: 12/11/2022]
Abstract
Phosphatidylinositol 4-phosphate 5-kinase (PI(4)P5K) is a type I lipid kinase that generates the lipid second messenger phospholipid phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) and functions downstream of RhoA in actin organization. It is known to play an essential role in neurite remodeling, yielding a phenotype identical to that seen in cells treated with Semaphorin 4D (Sema4D), a protein that regulates proliferation, adhesion and migration in many different cell types. Plexin-B1, the receptor for Sema4D, activates RhoA in order to generate a pro-angiogenic signal in endothelial cells. Therefore, we looked in human umbilical vein endothelial cells (HUVEC) to determine if Plexin-B1 exerted control over the cytoskeleton by regulation of PI(4)P5K activity. Here we demonstrate the Rho/Rho Kinase (ROK)-dependent generation of PI(4,5)P(2) upon treatment of HUVEC with Sema4D, as well as co-localization of PI(4)P5Kα with Plexin-B1. Formation of PI(4,5)P(2) was necessary for cytoskeletal polymerization, as expression of the phosphatase synaptojanin blocked this effect. We noted phosphorylation and activation of PLCγ and an increase in intracellular calcium upon treatment of HUVEC with Sema4D, responses that were necessary for a pro-angiogenic phenotype observed in vitro. Taken together, these results suggest that Plexin-B1 promotes angiogenesis in endothelial cells by signaling through PI(4)P5Kα and generating lipid second messengers.
Collapse
|
79
|
Maier V, Jolicoeur C, Rayburn H, Takegahara N, Kumanogoh A, Kikutani H, Tessier-Lavigne M, Wurst W, Friedel RH. Semaphorin 4C and 4G are ligands of Plexin-B2 required in cerebellar development. Mol Cell Neurosci 2011; 46:419-31. [PMID: 21122816 PMCID: PMC3030677 DOI: 10.1016/j.mcn.2010.11.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Revised: 10/27/2010] [Accepted: 11/15/2010] [Indexed: 01/15/2023] Open
Abstract
Semaphorins and Plexins are cognate ligand-receptor families that regulate important steps during nervous system development. The Plexin-B2 receptor is critically involved in neural tube closure and cerebellar granule cell development, however, its specific ligands have only been suggested by in vitro studies. Here, we show by in vivo and in vitro analyses that the two Semaphorin-4 family members Sema4C and Sema4G are likely to be in vivo ligands of Plexin-B2. The Sema4C and Sema4G genes are expressed in the developing cerebellar cortex, and Sema4C and Sema4G proteins specifically bind to Plexin-B2 expressing cerebellar granule cells. To further elucidate their in vivo function, we have generated and analyzed Sema4C and Sema4G knockout mouse mutants. Like Plexin-B2-/- mutants, Sema4C-/- mutants reveal exencephaly and subsequent neonatal lethality with partial penetrance. Sema4C-/- mutants that bypass exencephaly are viable and fertile, but display distinctive defects of the cerebellar granule cell layer, including gaps in rostral lobules, fusions of caudal lobules, and ectopic granule cells in the molecular layer. In addition to neuronal defects, we observed in Sema4C-/- mutants also ventral skin pigmentation defects that are similar to those found in Plexin-B2-/- mutants. The Sema4G gene deletion causes no overt phenotype by itself, but combined deletion of Sema4C and Sema4G revealed an enhanced cerebellar phenotype. However, Sema4C/Sema4G double mutants showed overall less severe cerebellar phenotypes than Plexin-B2-/- mutants, indicating that further ligands of Plexin-B2 exist. In explant cultures of the developing cerebellar cortex, Sema4C promoted migration of cerebellar granule cell precursors in a Plexin-B2-dependent manner, supporting the model that a reduced migration rate of granule cell precursors is the basis for the cerebellar defects of Sema4C-/- and Sema4C/Sema4G mutants.
Collapse
Affiliation(s)
- Viola Maier
- Institute of Developmental Genetics, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Christine Jolicoeur
- Department of Biological Sciences, Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| | - Helen Rayburn
- Department of Biological Sciences, Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| | - Noriko Takegahara
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Hitoshi Kikutani
- Department of Molecular Immunology, Osaka University, Osaka 565-0871, Japan
| | - Marc Tessier-Lavigne
- Department of Biological Sciences, Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
- Division of Research, Genentech Inc., South San Francisco, California 94080, USA
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Roland H. Friedel
- Institute of Developmental Genetics, Helmholtz Center Munich, 85764 Neuherberg, Germany
- Corresponding author: Roland H. Friedel, , Phone: +1 (212) 241 0937, Fax: +1 (212) 860 9279, Mount Sinai School of Medicine, Department of Developmental & Regenerative Biology, 1468 Madison Avenue, Annenberg Building, room 25-70, New York, NY 10029
| |
Collapse
|
80
|
Ye S, Hao X, Zhou T, Wu M, Wei J, Wang Y, Zhou L, Jiang X, Ji L, Chen Y, You L, Zhang Y, Xu G, Zhou J, Ma D, Wang S. Plexin-B1 silencing inhibits ovarian cancer cell migration and invasion. BMC Cancer 2010; 10:611. [PMID: 21059203 PMCID: PMC2991310 DOI: 10.1186/1471-2407-10-611] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Accepted: 11/08/2010] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Elevated Plexin-B1 expression has been found in diverse human cancers and in non-neoplastic tissues, and it mediates diverse biological and pathological activities. However, whether or not Plexin-B1 expression is involved in human ovarian tumors remains unclear. In the present study, Plexin-B1 expression was explored in benign and malignant human ovarian tumor tissues. In addition, the impact of Plexin-B1 expression on ovarian cancer cell proliferation, migration and invasion were investigated in vitro. METHODS Plexin-B1 expression was analyzed in normal and benign ovarian tissues and serous ovarian tumors (both borderline and malignant) by immunohistochemical staining, as well as in four human ovarian cancer cell lines (A2780, C13*, SKOV3, and OV2008) by RT-PCR and western blot analyses. Furthermore, endogenous Plexin-B1 expression was suppressed by Plexin-B1 siRNA in SKOV3 cells, which overexpress Plexin-B1. Protein levels of Plexin-B1, AKT and AKTSer473 were examined by western blot analysis. Cell proliferation, migration and invasion were measured with MTT, wound healing and boyden chamber assays, respectively, and the cytoskeleton was monitored via F-actin staining. RESULTS Expression levels of Plexin-B1 protein were significantly higher in serous ovarian carcinomas than in normal ovaries or benign ovarian neoplasms, and in the former, Plexin-B1 expression was positively correlated with lymphatic metastasis, and the membrane and cytoplasm of cancer cells stained positively. SKOV3 cells displayed the highest Plexin-B1 expression at both the mRNA and protein levels among the four tested human ovarian cancer cell lines and was selected as a cell model for further in vitro experiments. Plexin-B1 siRNA significantly suppressed phosphorylation of AKT at Ser473 in SKOV3 cells, but it did not alter total AKT expression. In addition, silencing of Plexin-B1 in SKOV3 cells inhibited cell migration and invasion and reorganized the cytoskeleton, whereas cell proliferation was not affected. CONCLUSION Plexin-B1 expression correlates with malignant phenotypes of serous ovarian tumors, probably via phosphorylation of AKT at Ser473, suggesting that Plexin-B1 might be a useful biomarker and/or a novel therapeutic target.
Collapse
Affiliation(s)
- Shuangmei Ye
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Melani M, Weinstein BM. Common factors regulating patterning of the nervous and vascular systems. Annu Rev Cell Dev Biol 2010; 26:639-65. [PMID: 19575651 DOI: 10.1146/annurev.cellbio.093008.093324] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The vascular and the nervous systems of vertebrates share many features with similar and often overlapping anatomy. The parallels between these two systems extend to the molecular level, where recent work has identified ever-increasing similarities between the molecular mechanisms employed in the specification, differentiation, and patterning of both systems. This review discusses some of the most recent literature on this subject, with particular emphasis on the roles that the Ephrin, Semaphorin, Netrin, and Slit signaling pathways play in vascular development.
Collapse
Affiliation(s)
- Mariana Melani
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
82
|
Ch'ng ES, Kumanogoh A. Roles of Sema4D and Plexin-B1 in tumor progression. Mol Cancer 2010; 9:251. [PMID: 20858260 PMCID: PMC2955613 DOI: 10.1186/1476-4598-9-251] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Accepted: 09/21/2010] [Indexed: 12/20/2022] Open
Abstract
Sema4D, also known as CD100, is a protein belonging to class IV semaphorin. Its physiologic roles in the immune and nervous systems have been extensively explored. However, the roles of Sema4D have extended beyond these traditionally studied territories. Via interaction with its high affinity receptor Plexin-B1, Sema4D-Plexin-B1 involvement in tumor progression is strongly implied. Here, we critically review and delineate the Sema4D-Plexin-B1 interaction in many facets of tumor progression: tumor angiogenesis, regulation of tumor-associated macrophages and control of invasive growth. We correlate the in vitro and in vivo experimental data with the clinical study outcomes, and present a molecular mechanistic basis accounting for the intriguingly contradicting results from these recent studies.
Collapse
Affiliation(s)
- Ewe Seng Ch'ng
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Malaysia.
| | | |
Collapse
|
83
|
Zielonka M, Xia J, Friedel RH, Offermanns S, Worzfeld T. A systematic expression analysis implicates Plexin-B2 and its ligand Sema4C in the regulation of the vascular and endocrine system. Exp Cell Res 2010; 316:2477-86. [PMID: 20478304 DOI: 10.1016/j.yexcr.2010.05.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 05/04/2010] [Accepted: 05/06/2010] [Indexed: 12/15/2022]
Abstract
Plexins serve as receptors for semaphorins and play important roles in the developing nervous system. Plexin-B2 controls decisive developmental programs in the neural tube and cerebellum. However, whether Plexin-B2 also regulates biological functions in adult nonneuronal tissues is unknown. Here we show by two methodologically independent approaches that Plexin-B2 is expressed in discrete cell types of several nonneuronal tissues in the adult mouse. In the vasculature, Plexin-B2 is selectively expressed in functionally specialized endothelial cells. In endocrine organs, Plexin-B2 localizes to the pancreatic islets of Langerhans and to both cortex and medulla of the adrenal gland. Plexin-B2 expression is also detected in certain types of immune and epithelial cells. In addition, we report on a systematic comparison of the expression patterns of Plexin-B2 and its ligand Sema4C, which show complementarity or overlap in some but not all tissues. Furthermore, we demonstrate that Plexin-B2 and its family member Plexin-B1 display largely nonredundant expression patterns. This work establishes Plexin-B2 and Sema4C as potential regulators of the vascular and endocrine system and provides an anatomical basis to understand the biological functions of this ligand-receptor pair.
Collapse
Affiliation(s)
- Matthias Zielonka
- Max-Planck-Institute for Heart and Lung Research, Department of Pharmacology, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | | | | | | | | |
Collapse
|
84
|
Cain RJ, Vanhaesebroeck B, Ridley AJ. The PI3K p110alpha isoform regulates endothelial adherens junctions via Pyk2 and Rac1. ACTA ACUST UNITED AC 2010; 188:863-76. [PMID: 20308428 PMCID: PMC2845076 DOI: 10.1083/jcb.200907135] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Only the p110α isoform of PI3K mediates the association of VE-cadherin with Pyk2, a Rac GEF and the p85 PI3K regulatory subunit, to reduce junctional integrity in response to TNF. Endothelial cell–cell junctions control efflux of small molecules and leukocyte transendothelial migration (TEM) between blood and tissues. Inhibitors of phosphoinositide 3-kinases (PI3Ks) increase endothelial barrier function, but the roles of different PI3K isoforms have not been addressed. In this study, we determine the contribution of each of the four class I PI3K isoforms (p110α, -β, -γ, and -δ) to endothelial permeability and leukocyte TEM. We find that depletion of p110α but not other p110 isoforms decreases TNF-induced endothelial permeability, Tyr phosphorylation of the adherens junction protein vascular endothelial cadherin (VE-cadherin), and leukocyte TEM. p110α selectively mediates activation of the Tyr kinase Pyk2 and GTPase Rac1 to regulate barrier function. Additionally, p110α mediates the association of VE-cadherin with Pyk2, the Rac guanine nucleotide exchange factor Tiam-1 and the p85 regulatory subunit of PI3K. We propose that p110α regulates endothelial barrier function by inducing the formation of a VE-cadherin–associated protein complex that coordinates changes to adherens junctions with the actin cytoskeleton.
Collapse
Affiliation(s)
- Robert J Cain
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, England, UK
| | | | | |
Collapse
|
85
|
Malan D, Wenzel D, Schmidt A, Geisen C, Raible A, Bölck B, Fleischmann BK, Bloch W. Endothelial beta1 integrins regulate sprouting and network formation during vascular development. Development 2010; 137:993-1002. [PMID: 20179098 DOI: 10.1242/dev.045377] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
beta1 integrins are important regulators of vascular differentiation and development, as their endothelial-specific deletion results in embryonic lethality. In the present study, we investigated the molecular mechanisms underlying the prominent vascular abnormalities that occur in the absence of beta1 integrins. Because of the early embryonic lethality of knockout mice, we studied endothelial cell and vessel development in beta1-integrin-deficient murine embryonic stem cells to gain novel insights into the role of beta1 integrins in vasculo-angiogenesis. We found that vessel development was strongly defective in the mutant embryoid bodies (EBs), as only primitive and short sprouts developed from clusters of vascular precursors in beta1 integrin(-/-) EBs, whereas complex network formation of endothelial tubes was observed in wild-type EBs. The vascular defect was due to deficient beta1 integrin expression in endothelial cells, as its endothelial-specific re-expression rescued the phenotype entirely. The mechanism responsible for defective vessel formation was found to be reduced endothelial cell maturation, migration and elongation. Moreover, the lower number of endothelial cells in beta1 integrin(-/-) EBs was due to an increased apoptosis versus proliferation rate. The enhanced apoptosis and proliferation of beta1 integrin(-/-) endothelial cells was related to the elevation of peNOS and pAKT signaling molecules, respectively. Our data demonstrate that endothelial beta1 integrins are determinants of vessel formation and that this effect is mediated via different signaling pathways.
Collapse
Affiliation(s)
- Daniela Malan
- Institute of Physiology I, University of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Wegiel B, Gallo DJ, Raman KG, Karlsson JM, Ozanich B, Chin BY, Tzeng E, Ahmad S, Ahmed A, Baty CJ, Otterbein LE. Nitric oxide-dependent bone marrow progenitor mobilization by carbon monoxide enhances endothelial repair after vascular injury. Circulation 2010; 121:537-48. [PMID: 20083679 DOI: 10.1161/circulationaha.109.887695] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Carbon monoxide (CO) has emerged as a vascular homeostatic molecule that prevents balloon angioplasty-induced stenosis via antiproliferative effects on vascular smooth muscle cells. The effects of CO on reendothelialization have not been evaluated. METHODS AND RESULTS Exposure to CO has diametrically opposite effects on endothelial cell (EC) and vascular smooth muscle cell proliferation in rodent models of carotid injury. In contrast to its effect of blocking vascular smooth muscle cell growth, CO administered as a gas or as a CO-releasing molecule enhances proliferation and motility of ECs in vitro by >50% versus air controls, and in vivo, it accelerates reendothelialization of the denuded artery by day 4 after injury versus day 6 in air-treated animals. CO enhanced EC proliferation via rapid activation of RhoA (Ras homolog gene family, member A), followed by downstream phosphorylation of Akt, endothelial nitric oxide (NO) synthase phosphorylation, and a 60% increase in NO generation by ECs. CO drives cell cycle progression through phosphorylation of retinoblastoma, which is dependent in part on endothelial NO synthase-generated NO. Similarly, endothelial repair in vivo requires NO-dependent mobilization of bone marrow-derived EC progenitors, and CO yielded a 4-fold increase in the number of mobilized green fluorescent protein-Tie2-positive endothelial progenitor cells versus controls, with a corresponding accelerated deposition of differentiated green fluorescent protein-Tie2-positive ECs at the site of injury. CO was ineffective in augmenting EC repair and the ensuing development of intimal hyperplasia in eNOS(-/-) mice. CONCLUSIONS Collectively, the present data demonstrate that CO accelerates EC proliferation and vessel repair in a manner dependent on NO generation and enhanced recruitment of bone marrow-derived endothelial progenitor cells.
Collapse
Affiliation(s)
- Barbara Wegiel
- Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Okuno T, Nakatsuji Y, Moriya M, Takamatsu H, Nojima S, Takegahara N, Toyofuku T, Nakagawa Y, Kang S, Friedel RH, Sakoda S, Kikutani H, Kumanogoh A. Roles of Sema4D-plexin-B1 interactions in the central nervous system for pathogenesis of experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2009; 184:1499-506. [PMID: 20038643 DOI: 10.4049/jimmunol.0903302] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Although semaphorins were originally identified as axonal guidance molecules during neuronal development, it is emerging that several semaphorins play crucial roles in various phases of immune responses. Sema4D/CD100, a class IV semaphorin, has been shown to be involved in the nervous and immune systems through its receptors plexin-B1 and CD72, respectively. However, the involvement of Sema4D in neuroinflammation still remains unclear. We found that Sema4D promoted inducible NO synthase expression by primary mouse microglia, the effects of which were abolished in plexin-B1-deficient but not in CD72-deficient microglia. In addition, during the development of experimental autoimmune encephalomyelitis (EAE), which was induced by immunization with myelin oligodendrocyte glycoprotein-derived peptides, we observed that the expression of Sema4D and plexin-B1 was induced in infiltrating mononuclear cells and microglia, respectively. Consistent with these expression profiles, when myelin oligodendrocyte glycoprotein-specific T cells derived from wild-type mice were adoptively transferred into plexin-B1-deficient mice or bone marrow chimera mice with plexin-B1-deficient CNS resident cells, the development of EAE was considerably attenuated. Furthermore, blocking Abs against Sema4D significantly inhibited neuroinflammation during EAE development. Collectively, our findings demonstrate the role of Sema4D-plexin-B1 interactions in the activation of microglia and provide their pathologic significance in neuroinflammation.
Collapse
Affiliation(s)
- Tatsusada Okuno
- Department of Immunopathology, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Vodrazka P, Korostylev A, Hirschberg A, Swiercz JM, Worzfeld T, Deng S, Fazzari P, Tamagnone L, Offermanns S, Kuner R. The semaphorin 4D-plexin-B signalling complex regulates dendritic and axonal complexity in developing neurons via diverse pathways. Eur J Neurosci 2009; 30:1193-208. [PMID: 19788569 DOI: 10.1111/j.1460-9568.2009.06934.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Semaphorins and their receptors, plexins, have emerged as key regulators of various aspects of neuronal development. In contrast to the Plexin-A family, the cellular functions of Plexin-B family proteins in developing neurons are only poorly understood. An activation of Plexin-B1 via its ligand, semaphorin 4D (Sema4D), produces an acute collapse of axonal growth cones in hippocampal and retinal neurons over the early stages of neurite outgrowth. However, the functional role of Sema4D-Plexin-B interactions over subsequent stages of neurite development, differentiation and maturation has not been characterized. Here we addressed this question using morphogenetic assays and time-lapse imaging on developing rat hippocampal neurons as a model system. Interestingly, Sema4D treatment over several hours was observed to promote branching and complexity in hippocampal neurons via the activation of Plexin-B1. The activation of receptor tyrosine kinases and the Rho kinase following Sema4D treatment was found to control dendritic and axonal morphogenesis by differentially regulating branching and extension. Phosphoinositide-3-kinase, but not extracellular signal-regulated kinase 1/2, was observed to be important for the stimulatory effects of Sema4D on dendritic branching. Furthermore, we observed that the mammalian target of rapamycin is activated downstream of Plexin-B1 and contributes to Sema4D-induced effects on dendritic branching. In contrast, glycogen synthase kinase-3 beta, another effector of phosphoinositide-3-kinase signalling, was not involved. Thus, our results show that Sema4D-Plexin-B interactions modulate dendritic and axonal arborizations of developing neurons by co-ordinated and concerted activation of diverse signalling pathways.
Collapse
Affiliation(s)
- Peter Vodrazka
- Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Sun XZ, Li ZF, Liu Y, Fang P, Li MX. Inhibition of cGMP phosphodiesterase 5 suppresses matrix metalloproteinase-2 production in pulmonary artery smooth muscles cells. Clin Exp Pharmacol Physiol 2009; 37:362-7. [PMID: 19793106 DOI: 10.1111/j.1440-1681.2009.05304.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
1. It has been shown that the beneficial effects of phosphodiesterase (PDE) 5 inhibition on pulmonary hypertension (PH) are associated with the induction of vascular relaxation and suppression of the proliferation of pulmonary artery smooth muscle cells (PASMC). In the present study, we investigated whether PDE5 inhibition affects the production and/or secretion of matrix metalloproteinases (MMPs) in PASMC, resulting in extracellular matrix remodelling in the pulmonary vasculature and, thus, the development of PH. 2. Primary cultured PASMC were stimulated with endothelin (ET)-1 and MMP-2 production and RhoA activation were then determinded using gelatin zymography and a GTP-bound RhoA assay, respectively. The effects of the selective PDE5 inhibitor sildenafil and subsequent protein kinase G-specific inhibitor Rp-8Br-cGMPs on MMP-2 production and RhoA activation were further exmamined. 3. Endothelin-1 (1-1000 nmol/L) concentration-dependently stimulated MMP-2 production and/or secretion in primary cultured PASMC, with 100 nmol/L ET-1 causing a 2.41-fold increase in MMP-2 production compared with control (P < 0.01). This increase in MMP-2 production was accompanied by RhoA activation, which was abolished by preincubation of cells with 10 micromol/L Y27632, an inhibitor of Rho-associated kinase (ROCK). Furthermore, 10 micromol/L Y27632 abolished the ET-1-induced production of MMP-2. 4. The selective PDE5 inhibitor sildenafil (0.1-1 micromol/L) concentration-dependently reduced the increased MMP-2 production induced by 100 nmol/L ET-1. Specifically, in the presence of 1 micromol/L sildenafil, the 100 nmol/L ET-1-induced increase in MMP-2 production was only increased 1.3-fold over that of the control (P < 0.01 vs 100 nmol/L ET-1-stimulated cells). 5. Suppression of RhoA activation was found to mediate the inhibitory effect of sildenafil on ET-1-induced increases in MMP-2 production. Furthermore, the protein kinase G-specific inhibitor Rp-8Br-cGMPs reversed the inhibitory effects of sildenafil on RhoA activation and MMP-2 production. 6. The results of the present study indicate that PDE5 inhibition suppresses RhoA/ROCK-mediated MMP-2 production by PASMC, which may contribute to the regulation of pulmonary vascular remodelling. Thus, PDE5 inhibition may benefit patients with PH through multiple mechanisms of action.
Collapse
Affiliation(s)
- Xiu-Zhen Sun
- Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | | | | | | | | |
Collapse
|
90
|
Capparuccia L, Tamagnone L. Semaphorin signaling in cancer cells and in cells of the tumor microenvironment--two sides of a coin. J Cell Sci 2009; 122:1723-36. [PMID: 19461072 DOI: 10.1242/jcs.030197] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Semaphorins are a large family of secreted and membrane-bound molecules that were initially implicated in the development of the nervous system and in axon guidance. More recently, they have been found to regulate cell adhesion and motility, angiogenesis, immune responses, and tumor progression. Semaphorin receptors, the neuropilins and the plexins, are expressed by a wide variety of cell types, including endothelial cells, bone-marrow-derived cells and cancer cells. Interestingly, a growing body of evidence indicates that semaphorins also have an important role in cancer. It is now known that cancer progression, invasion and metastasis involve not only genetic changes in the tumor cells but also crosstalk between tumor cells and their surrounding non-tumor cells. Through the recruitment of endothelial cells, leukocytes, pericytes and fibroblasts, and the local release of growth factors and cytokines, the tumor microenvironment can mediate tumor-cell survival, tumor proliferation and regulation of the immune response. Moreover, by conferring cancer cells with an enhanced ability to migrate and invade adjacent tissues, extracellular regulatory signals can play a major role in the metastatic process. In this Commentary, we focus on the emerging role of semaphorins in mediating the crosstalk between tumor cells and multiple stromal cell types in the surrounding microenvironment.
Collapse
Affiliation(s)
- Lorena Capparuccia
- Institute for Cancer Research and Treatment (IRCC), University of Turin, S.P. 142, 10060, Candiolo (TO), Italy
| | | |
Collapse
|
91
|
Plexin B1 is repressed by oncogenic B-Raf signaling and functions as a tumor suppressor in melanoma cells. Oncogene 2009; 28:2697-709. [PMID: 19483722 PMCID: PMC3238492 DOI: 10.1038/onc.2009.133] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human melanomas show oncogenic B-Raf mutations which activate the B-Raf/MKK/ERK cascade. We screened microarrays to identify cellular targets of this pathway, and found that genes upregulated by B-Raf/MKK/ERK showed highest association with cell cycle regulators, whereas genes downregulated were most highly associated with axon guidance genes, including plexin-semaphorin family members. Plexin B1 was strongly inhibited by MAP kinase signaling in melanoma cells and melanocytes. In primary melanoma cells, plexin B1 blocked tumorigenesis as measured by growth of colonies in soft agar, spheroids in extracellular matrix, and xenograft tumors. Tumor suppression depended on residues in the C-terminal domain of plexin B1 which mediate receptor GAP activity, and also correlated with AKT inhibition. Interestingly, the inhibitory response to plexin B1 was reduced or absent in cells from a matched metastatic tumor, suggesting that changes occur in metastatic cells which bypass the tumor suppressor mechanisms. Plexin B1 also inhibited cell migration, but this was seen in metastatic cells and not in matched primary cells. Thus, plexin B1 has tumor suppressor function in early-stage cells, while suppressing migration in late-stage cells. Our findings suggest that B-Raf/MKK/ERK provides a permissive environment for melanoma genesis by modulating plexin B1.
Collapse
|
92
|
Abstract
Semaphorins belong to a large family of proteins well-conserved along evolution from viruses to mammalians. Secreted and membrane-bound semaphorins participate in a wide range of biological phenomena including development and regeneration of nervous system, cardiovascular development, and immune system activities. Different classes of semaphorins are bifunctional and often exert opposite effects (i.e., repellent or attractive) by acting through the plexin receptor family. However, some classes use other membrane receptors and the same plexin-mediated signals may be modulated by co-receptors, in particular neuropilins or some tyrosine kinase receptors. In cancer, semaphorins have both tumor-suppressor and tumor-promoting functions, by acting on both tumor and stromal components. Here, we review the role of semaphorins in tumor angiogenesis and propose that an unbalance between autocrine loops respectively involving angiogenic inducers and class 3 semaphorin is instrumental for structural and functional abnormalities observed in tumor vasculature.
Collapse
|
93
|
Guidance from above: common cues direct distinct signaling outcomes in vascular and neural patterning. Trends Cell Biol 2009; 19:99-110. [PMID: 19200729 DOI: 10.1016/j.tcb.2009.01.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Revised: 12/19/2008] [Accepted: 01/05/2009] [Indexed: 01/13/2023]
Abstract
The nervous and vascular systems are both exquisitely branched and complicated systems and their proper development requires careful guidance of nerves and vessels. The recent realization that common ligand-receptor pairs are used in guiding the patterning of both systems has prompted the question of whether similar signaling pathways are used in both systems. This review highlights recent progress in our understanding of the similarities and differences in the intracellular signaling mechanisms downstream of semaphorins, ephrins and vascular endothelial growth factor in neurons and endothelial cells during neural and vascular development. We present evidence that similar intracellular signaling principles underlying cytoskeletal regulation are used to control neural and vascular guidance, although the specific molecules used in neurons and endothelial cells are often different.
Collapse
|
94
|
Korostylev A, Worzfeld T, Deng S, Friedel RH, Swiercz JM, Vodrazka P, Maier V, Hirschberg A, Ohoka Y, Inagaki S, Offermanns S, Kuner R. A functional role for semaphorin 4D/plexin B1 interactions in epithelial branching morphogenesis during organogenesis. Development 2008; 135:3333-43. [PMID: 18799546 DOI: 10.1242/dev.019760] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Semaphorins and their receptors, plexins, carry out important functions during development and disease. In contrast to the well-characterized plexin A family, however, very little is known about the functional relevance of B-type plexins in organogenesis, particularly outside the nervous system. Here, we demonstrate that plexin B1 and its ligand Sema4d are selectively expressed in epithelial and mesenchymal compartments during key steps in the genesis of some organs. This selective expression suggests a role in epithelial-mesenchymal interactions. Importantly, using the developing metanephros as a model system, we have observed that endogenously expressed and exogenously supplemented Sema4d inhibits branching morphogenesis during early stages of development of the ureteric collecting duct system. Our results further suggest that the RhoA-ROCK pathway, which is activated downstream of plexin B1, mediates these inhibitory morphogenetic effects of Sema4d and suppresses branch-promoting signalling effectors of the plexin B1 signalling complex. Finally, mice that lack plexin B1 show early anomalies in kidney development in vivo. These results identify a novel function for plexin B1 as a negative regulator of branching morphogenesis during kidney development, and suggest that the Sema4d-plexin B1 ligand-receptor pair contributes to epithelial-mesenchymal interactions during organogenesis via modulation of RhoA signalling.
Collapse
Affiliation(s)
- Alexander Korostylev
- Pharmacology Institute, Im Neuenheimer Feld 366, University of Heidelberg, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Gavard J, Gutkind JS. Protein kinase C-related kinase and ROCK are required for thrombin-induced endothelial cell permeability downstream from Galpha12/13 and Galpha11/q. J Biol Chem 2008; 283:29888-96. [PMID: 18713748 DOI: 10.1074/jbc.m803880200] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Increase in vascular permeability occurs under many physiological conditions such as wound repair, inflammation, and thrombotic reactions and is central in diverse human pathologies, including tumor-induced angiogenesis, ocular diseases, and septic shock. Thrombin is a pro-coagulant serine protease, which causes the local loss of endothelial barrier integrity thereby enabling the rapid extravasation of plasma proteins and the local formation of fibrin-containing clots. Available information suggests that thrombin induces endothelial permeability by promoting actomyosin contractility through the Rho/ROCK signaling pathway. Here we took advantage of pharmacological inhibitors, knockdown approaches, and the emerging knowledge on how permeability factors affect endothelial junctions to investigate in detail the mechanism underlying thrombin-induced endothelial permeability. We show that thrombin signals through PAR-1 and its coupled G proteins Galpha(12/13) and Galpha(11/q) to induce RhoA activation and intracellular calcium elevation, and that these events are interrelated. In turn, this leads to the stimulation of ROCK, which causes actin stress-fiber formation. However, this alone is not sufficient to account for thrombin-induced permeability. Instead, we found that protein kinase C-related kinase, a Rho-dependent serine/threonine kinase, is activated in endothelial cells upon thrombin stimulation and that its expression is required for endothelial permeability and the remodeling of cell-extracellular matrix and cell-cell adhesions. Our results demonstrate that the signal initiated by thrombin bifurcates at the level of RhoA to promote changes in the cytoskeletal architecture through ROCK, and the remodeling of focal adhesion components through protein kinase C-related kinase. Ultimately, both pathways converge to cause cell-cell junction disruption and provoke vascular leakage.
Collapse
Affiliation(s)
- Julie Gavard
- Oral and Pharyngeal Cancer Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
96
|
Neufeld G, Kessler O. The semaphorins: versatile regulators of tumour progression and tumour angiogenesis. Nat Rev Cancer 2008; 8:632-45. [PMID: 18580951 DOI: 10.1038/nrc2404] [Citation(s) in RCA: 303] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The semaphorins and their receptors, the neuropilins and the plexins, were originally characterized as constituents of the complex regulatory system responsible for the guidance of axons during the development of the central nervous system. However, a growing body of evidence indicates that various semaphorins can either promote or inhibit tumour progression through the promotion or inhibition of processes such as tumour angiogenesis, tumour metastasis and tumour cell survival. This Review focuses on the emerging role of the semaphorins in cancer.
Collapse
Affiliation(s)
- Gera Neufeld
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, P.O. Box 9679, 1 Efron Street, Haifa, 31096, Israel.
| | | |
Collapse
|
97
|
Tyrosine phosphorylation in semaphorin signalling: shifting into overdrive. EMBO Rep 2008; 9:865-71. [PMID: 18660749 DOI: 10.1038/embor.2008.139] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 06/24/2008] [Indexed: 01/15/2023] Open
Abstract
The semaphorins constitute a large family of molecular signals with regulatory functions in neuronal development, angiogenesis, cancer progression and immune responses. Accumulating data indicate that semaphorins might trigger multiple signalling pathways, and mediate different and sometimes opposing effects, depending on the cellular context and the particular plexin-associated subunits of the receptor complex, which can include receptor-type or cytoplasmic tyrosine kinases such as MET, ERBB2, VEGFR2, FYN, FES, PYK2 and SRC. It has also been shown that a specific plexin can alternatively associate with different tyrosine kinase receptors, eliciting divergent signalling pathways and functional outcomes. Tyrosine phosphorylation is a pivotal post-translational protein modification that regulates intracellular signalling. Therefore, phosphorylation of tyrosines in the intracellular domain of plexins could determine or modify their interactions with additional signal transducers. Here, we discuss the potential relevance of tyrosine phosphorylation in semaphorin-induced signalling, with an emphasis on its probable role in dictating the choice between multiple pathways and functional outcomes. The identification of implicated tyrosine kinases will pave the way to target individual semaphorin-mediated functions.
Collapse
|
98
|
Abstract
Contractile forces mediated by RhoA and Rho kinase (ROCK) are required for a variety of cellular processes, including cell adhesion. In this study, we show that RhoA-dependent ROCKII activation is negatively regulated by phosphorylation at a conserved tyrosine residue (Y722) in the coiled-coil domain of ROCKII. Tyrosine phosphorylation of ROCKII is increased with cell adhesion, and loss of Y722 phosphorylation delays adhesion and spreading on fibronectin, suggesting that this modification is critical for restricting ROCKII-mediated contractility during these processes. Further, we provide evidence that Shp2 mediates dephosphorylation of ROCKII and, therefore, regulates RhoA-induced cell rounding, indicating that Shp2 couples with RhoA signaling to control ROCKII activation during deadhesion. Thus, reversible tyrosine phosphorylation confers an additional layer of control to fine-tune RhoA-dependent activation of ROCKII.
Collapse
Affiliation(s)
- Hsiao-Hui Lee
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | | |
Collapse
|
99
|
Zhou Y, Gunput RAF, Pasterkamp RJ. Semaphorin signaling: progress made and promises ahead. Trends Biochem Sci 2008; 33:161-70. [PMID: 18374575 DOI: 10.1016/j.tibs.2008.01.006] [Citation(s) in RCA: 236] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 01/16/2008] [Accepted: 01/18/2008] [Indexed: 12/31/2022]
Abstract
Semaphorins were initially characterized according to their role in repulsive axon guidance but are now recognized as crucial regulators of morphogenesis and homeostasis over a wide range of organ systems. The pleiotropic nature of semaphorin signaling and its implication in human disease has triggered an enormous interest in the receptor and intracellular signaling mechanisms that direct the cell-type-specific and diverse biological effects of semaphorins. Recent breakthroughs in our understanding of semaphorin signaling link integrin and semaphorin signaling pathways, identify novel ligand-receptor interactions and provide insight into the cellular and molecular bases of bifunctional and reverse signaling events. These discoveries could lead to therapeutic advances in axonal regeneration, cancer and other diseases.
Collapse
Affiliation(s)
- Yeping Zhou
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Universiteitsweg 100, Utrecht, The Netherlands
| | | | | |
Collapse
|
100
|
Abstract
PURPOSE OF REVIEW Understanding the role of integrins in the formation of vascular bed is important for designing new therapeutic approaches to ameliorate or inhibit pathological vascularization. Besides regulating cell adhesion and migration, integrins dynamically participate in a network with soluble molecules and their receptors. This study summarizes recent progress in the understanding of the reciprocal interactions between integrins, tyrosine kinase, and semaphorin receptors. RECENT FINDINGS During angiogenic remodeling, endothelial cells that line blood vessel walls dynamically modify their integrin-mediated adhesive contacts with the surrounding extracellular matrix. During angiogenesis, opposing autocrine and paracrine loops of growth factors and semaphorins regulate endothelial integrin activation and function through tyrosine kinase receptors and the neuropilin/plexins system. Moreover, proangiogenic and antiangiogenic factors can directly bind integrins and regulate endothelial cell behavior. Studies describing these intense research areas are discussed. SUMMARY Alteration in the balance between the angiogenic growth factors and semaphorins results in an impairment of integrin functions and could account for cardiovascular malformation and structural and functional abnormalities of the tumor vasculature.
Collapse
|