Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Opin Organ Transplant ; 22(1): 55-63, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27898465

RESUMO

PURPOSE OF REVIEW: Chronic rejection is associated with persistent mononuclear cell recruitment, endothelial activation and proliferation, local tissue hypoxia and related biology that enhance effector immune responses. In contrast, the tumor microenvironment elicits signals/factors that inhibit effector T cell responses and rather promote immunoregulation locally within the tissue itself. The identification of immunoregulatory check points and/or secreted factors that are deficient within allografts is of great importance in the understanding and prevention of chronic rejection. RECENT FINDINGS: The relative deficiency of immunomodulatory molecules (cell surface and secreted) on microvascular endothelial cells within the intragraft microenvironment, is of functional importance in shaping the phenotype of rejection. These regulatory molecules include coinhibitory and/or intracellular regulatory signals/factors that enhance local activation of T regulatory cells. For example, semaphorins may interact with endothelial cells and CD4 T cells to promote local tolerance. Additionally, metabolites and electrolytes within the allograft microenvironment may regulate local effector and regulatory cell responses. SUMMARY: Multiple factors within allografts shape the microenvironment either towards local immunoregulation or proinflammation. Promoting the expression of intragraft cell surface or secreted molecules that support immunoregulation will be critical for long-term graft survival and/or alloimmune tolerance.


Assuntos
Rejeição de Enxerto/imunologia , Tolerância Imunológica/imunologia , Humanos , Microambiente Tumoral
2.
Am J Pathol ; 186(11): 2803-2812, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27751443

RESUMO

The vasculature influences the progression and resolution of tissue inflammation. Capillaries express vascular endothelial growth factor (VEGF) receptors, including neuropilins (NRPs), which regulate interstitial fluid flow. NRP2, a receptor of VEGFA and semaphorin (SEMA) 3F ligands, is expressed in the vascular and lymphatic endothelia. Previous studies have demonstrated that blocking VEGF receptor 2 attenuates VEGFA-induced vascular permeability. The inhibition of NRP2 was hypothesized to decrease vascular permeability as well. Unexpectedly, massive tissue swelling and edema were observed in Nrp2-/- mice compared with wild-type littermates after delayed-type hypersensitivity reactions. Vascular permeability was twofold greater in inflamed blood vessels in Nrp2-deficient mice compared to those in Nrp2-intact littermates. The addition of exogenous SEMA3F protein inhibited vascular permeability in Balb/cJ mice, suggesting that the loss of endogenous Sema3F activity in the Nrp2-deficient mice was responsible for the enhanced vessel leakage. Functional lymphatic capillaries are necessary for draining excess fluid after inflammation; however, Nrp2-mutant mice lacked superficial lymphatic capillaries, leading to 2.5-fold greater fluid retention and severe lymphedema after inflammation. In conclusion, Nrp2 deficiency increased blood vessel permeability and decreased lymphatic vessel drainage during inflammation, highlighting the importance of the NRP2/SEMA3F pathway in the modulation of tissue swelling and resolution of postinflammatory edema.


Assuntos
Linfedema/genética , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuropilina-2/deficiência , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Vasos Sanguíneos/fisiopatologia , Permeabilidade Capilar , Feminino , Humanos , Inflamação/genética , Inflamação/fisiopatologia , Vasos Linfáticos/fisiopatologia , Linfedema/fisiopatologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Tecido Nervoso/genética , Neuropilina-2/genética , Neuropilina-2/metabolismo , Organismos Livres de Patógenos Específicos , Fator A de Crescimento do Endotélio Vascular/genética
3.
J Immunol ; 197(4): 1389-98, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27430720

RESUMO

Netrin-1 is a neuronal guidance cue that regulates cellular activation, migration, and cytoskeleton rearrangement in multiple cell types. It is a chemotropic protein that is expressed within tissues and elicits both attractive and repulsive migratory responses. Netrin-1 has recently been found to modulate the immune response via the inhibition of neutrophil and macrophage migration. However, the ability of Netrin-1 to interact with lymphocytes and its in-depth effects on leukocyte migration are poorly understood. In this study, we profiled the mRNA and protein expression of known Netrin-1 receptors on human CD4(+) T cells. Neogenin, uncoordinated-5 (UNC5)A, and UNC5B were expressed at low levels in unstimulated cells, but they increased following mitogen-dependent activation. By immunofluorescence, we observed a cytoplasmic staining pattern of neogenin and UNC5A/B that also increased following activation. Using a novel microfluidic assay, we found that Netrin-1 stimulated bidirectional migration and enhanced the size of migratory subpopulations of mitogen-activated CD4(+) T cells, but it had no demonstrable effects on the migration of purified CD4(+)CD25(+)CD127(dim) T regulatory cells. Furthermore, using a short hairpin RNA knockdown approach, we observed that the promigratory effects of Netrin-1 on T effectors is dependent on its interactions with neogenin. In the humanized SCID mouse, local injection of Netrin-1 into skin enhanced inflammation and the number of neogenin-expressing CD3(+) T cell infiltrates. Neogenin was also observed on CD3(+) T cell infiltrates within human cardiac allograft biopsies with evidence of rejection. Collectively, our findings demonstrate that Netrin-1/neogenin interactions augment CD4(+) T cell chemokinesis and promote cellular infiltration in association with acute inflammation in vivo.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Quimiotaxia de Leucócito/fisiologia , Fatores de Crescimento Neural/metabolismo , Receptores de Superfície Celular/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Western Blotting , Células Cultivadas , Citometria de Fluxo , Imunofluorescência , Humanos , Técnicas In Vitro , Camundongos , Camundongos SCID , Técnicas Analíticas Microfluídicas , Receptores de Netrina , Netrina-1 , Reação em Cadeia da Polimerase em Tempo Real
4.
Sci Rep ; 6: 26794, 2016 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-27243144

RESUMO

Heparin binding epidermal growth factor-like growth factor (HB-EGF) is an angiogenic factor mediating radial migration of the developing forebrain, while vascular endothelial growth factor (VEGF) is known to influence rostral migratory stream in rodents. Cell migratory defects have been identified in animal models of hydrocephalus; however, the relationship between HB-EGF and hydrocephalus is unclear. We show that mice overexpressing human HB-EGF with ß-galactosidase reporter exhibit an elevated VEGF, localization of ß-galactosidase outside the subventricular zone (SVZ), subarachnoid hemorrhage, and ventriculomegaly. In Wistar polycystic kidney rats with hydrocephalus, alteration of migratory trajectory is detected. Furthermore, VEGF infusions into the rats result in ventriculomegaly with an increase of SVZ neuroblast in rostral migratory stream, whereas VEGF ligand inhibition prevents it. Our results support the idea that excess HB-EGF leads to a significant elevation of VEGF and ventricular dilatation. These data suggest a potential pathophysiological mechanism that elevated HB-EGF can elicit VEGF induction and hydrocephalus.


Assuntos
Ventrículos Cerebrais/patologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Hidrocefalia/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Movimento Celular , Células Endoteliais/metabolismo , Humanos , Hidrocefalia/patologia , Ventrículos Laterais/metabolismo , Masculino , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Ratos Wistar , Transdução de Sinais
5.
Biol Open ; 4(9): 1063-76, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26209534

RESUMO

Neuropilin-1 (NRP1) has been identified as a VEGF-A receptor. DJM-1, a human skin cancer cell line, expresses endogenous VEGF-A and NRP1. In the present study, the RNA interference of VEGF-A or NRP1 suppressed DJM-1 cell proliferation. Furthermore, the overexpression of the NRP1 wild type restored shNRP1-treated DJM-1 cell proliferation, whereas NRP1 cytoplasmic deletion mutants did not. A co-immunoprecipitation analysis revealed that VEGF-A induced interactions between NRP1 and GIPC1, a scaffold protein, and complex formation between GIPC1 and Syx, a RhoGEF. The knockdown of GIPC1 or Syx reduced active RhoA and DJM-1 cell proliferation without affecting the MAPK or Akt pathway. C3 exoenzyme or Y27632 inhibited the VEGF-A-induced proliferation of DJM-1 cells. Conversely, the overexpression of the constitutively active form of RhoA restored the proliferation of siVEGF-A-treated DJM-1 cells. Furthermore, the inhibition of VEGF-A/NRP1 signaling upregulated p27, a CDK inhibitor. A cell-penetrating oligopeptide that targeted GIPC1/Syx complex formation inhibited the VEGF-A-induced activation of RhoA and suppressed DJM-1 cell proliferation. In conclusion, this new signaling pathway of VEGF-A/NRP1 induced cancer cell proliferation by forming a GIPC1/Syx complex that activated RhoA to degrade the p27 protein.

6.
Sci Rep ; 5: 11789, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-26156437

RESUMO

Semaphorin 3F (SEMA3F) provides neuronal guidance cues via its ability to bind neuropilin 2 (NRP2) and Plexin A family molecules. Recent studies indicate that SEMA3F has biological effects in other cell types, however its mechanism(s) of function is poorly understood. Here, we analyze SEMA3F-NRP2 signaling responses in human endothelial, T cell and tumor cells using phosphokinase arrays, immunoprecipitation and Western blot analyses. Consistently, SEMA3F inhibits PI-3K and Akt activity, and responses are associated with the disruption of mTOR/rictor assembly and mTOR-dependent activation of the RhoA GTPase. We also find that the expression of vascular endothelial growth factor, as well as mTOR-inducible cellular activation responses and cytoskeleton stability are inhibited by SEMA3F-NRP2 interactions in vitro. In vivo, local and systemic overproduction of SEMA3F reduces tumor growth in NRP2-expressing xenografts. Taken together, SEMA3F regulates mTOR signaling in diverse human cell types, suggesting that it has broad therapeutic implications.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuropilina-2/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Xenoenxertos , Humanos , Hipóxia/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Proteínas de Membrana/genética , Camundongos , Modelos Biológicos , Complexos Multiproteicos/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Proteínas do Tecido Nervoso/genética , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Carga Tumoral , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Biochem Biophys Res Commun ; 464(1): 126-32, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26086095

RESUMO

Class 3 semaphorins were discovered as a family of axon guidance molecules, but are now known to be involved in diverse biologic processes. In this study, we investigated the anti-angiogenic potential of SEMA3E and SEMA3F (SEMA3E&F) in infantile hemangioma (IH). IH is a common vascular tumor that involves both vasculogenesis and angiogenesis. Our lab has identified and isolated hemangioma stem cells (HemSC), glucose transporter 1 positive (GLUT1(+)) endothelial cells (designated as GLUT1(sel) cells) based on anti-GLUT1 magnetic beads selection and GLUT1-negative endothelial cells (named HemEC). We have shown that these types of cells play important roles in hemangiogenesis. We report here that SEMA3E inhibited HemEC migration and proliferation while SEMA3F was able to suppress the migration and proliferation in all three types of cells. Confocal microscopy showed that stress fibers in HemEC were reduced by SEMA3E&F and that stress fibers in HemSC were decreased by SEMA3F, which led to cytoskeletal collapse and loss of cell motility in both cell types. Additionally, SEMA3E&F were able to inhibit vascular endothelial growth factor (VEGF)-induced sprouts in all three types of cells. Further, SEMA3E&F reduced the level of p-VEGFR2 and its downstream p-ERK in HemEC. These results demonstrate that SEMA3E&F inhibit IH cell proliferation and suppress the angiogenic activities of migration and sprout formation. SEMA3E&F may have therapeutic potential to treat or prevent growth of highly proliferative IH.


Assuntos
Células Endoteliais/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Proteínas de Membrana/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Neovascularização Patológica/prevenção & controle , Proteínas do Tecido Nervoso/farmacologia , Semaforinas/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hemangioma/irrigação sanguínea , Hemangioma/genética , Hemangioma/patologia , Humanos , Lactente , Neoplasias de Tecido Vascular/irrigação sanguínea , Neoplasias de Tecido Vascular/genética , Neoplasias de Tecido Vascular/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Br J Pharmacol ; 172(16): 4107-18, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25989290

RESUMO

BACKGROUND AND PURPOSE: Highly vascularized ovarian carcinoma secretes the putative endocannabinoid and GPR55 agonist, L-α-lysophosphatidylinositol (LPI), into the circulation. We aimed to assess the involvement of this agonist and its receptor in ovarian cancer angiogenesis. EXPERIMENTAL APPROACH: Secretion of LPI by three ovarian cancer cell lines (OVCAR-3, OVCAR-5 and COV-362) was tested by mass spectrometry. Involvement of cancer cell-derived LPI on angiogenesis was tested in the in vivo chicken chorioallantoic membrane (CAM) assay along with the assessment of the effect of LPI on proliferation, network formation, and migration of neonatal and adult human endothelial colony-forming cells (ECFCs). Engagement of GPR55 was verified by using its pharmacological inhibitor CID16020046 and diminution of GPR55 expression by four different target-specific siRNAs. To study underlying signal transduction, Western blot analysis was performed. KEY RESULTS: Ovarian carcinoma cell-derived LPI stimulated angiogenesis in the CAM assay. Applied LPI stimulated proliferation, network formation, and migration of neonatal ECFCs in vitro and angiogenesis in the in vivo CAM. The pharmacological GPR55 inhibitor CID16020046 inhibited LPI-stimulated ECFC proliferation, network formation and migration in vitro as well as ovarian carcinoma cell- and LPI-induced angiogenesis in vivo. Four target-specific siRNAs against GPR55 prevented these effects of LPI on angiogenesis. These pro-angiogenic effects of LPI were transduced by GPR55-dependent phosphorylation of ERK1/2 and p38 kinase. CONCLUSIONS AND IMPLICATIONS: We conclude that inhibiting the pro-angiogenic LPI/GPR55 pathway appears a promising target against angiogenesis in ovarian carcinoma.


Assuntos
Lisofosfolipídeos/metabolismo , Neovascularização Patológica/metabolismo , Neoplasias Ovarianas/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Galinhas , Membrana Corioalantoide/efeitos dos fármacos , Membrana Corioalantoide/fisiologia , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Receptores de Canabinoides , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Stem Cells ; 33(1): 133-45, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25187207

RESUMO

Endothelial glucose transporter 1 (GLUT1) is a definitive and diagnostic marker for infantile hemangioma (IH), a vascular tumor of infancy. To date, GLUT1-positive endothelial cells in IH have not been quantified nor directly isolated and studied. We isolated GLUT1-positive and GLUT1-negative endothelial cells from IH specimens and characterized their proliferation, differentiation, and response to propranolol, a first-line therapy for IH, and to rapamycin, an mTOR pathway inhibitor used to treat an increasingly wide array of proliferative disorders. Although freshly isolated GLUT1-positive cells, selected using anti-GLUT1 magnetic beads, expressed endothelial markers CD31, VE-Cadherin, and vascular endothelial growth factor receptor 2, they converted to a mesenchymal phenotype after 3 weeks in culture. In contrast, GLUT1-negative endothelial cells exhibited a stable endothelial phenotype in vitro. GLUT1-selected cells were clonogenic when plated as single cells and could be induced to redifferentiate into endothelial cells, or into pericytes/smooth muscle cells or into adipocytes, indicating a stem cell-like phenotype. These data demonstrate that, although they appear and function in the tumor as bona fide endothelial cells, the GLUT1-positive endothelial cells display properties of facultative stem cells. Pretreatment with rapamycin for 4 days significantly slowed proliferation of GLUT1-selected cells, whereas propranolol pretreatment had no effect. These results reveal for the first time the facultative nature of GLUT1-positive endothelial cells in IH.


Assuntos
Células Endoteliais/patologia , Transportador de Glucose Tipo 1/metabolismo , Hemangioma/patologia , Células-Tronco Neoplásicas/patologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Criança , Pré-Escolar , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Hemangioma/tratamento farmacológico , Hemangioma/metabolismo , Humanos , Lactente , Masculino , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Propranolol/farmacologia , Sirolimo/farmacologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/patologia
10.
Angiogenesis ; 18(2): 151-62, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25424831

RESUMO

Lymphatic malformations (LM) are characterized by abnormal formation of lymphatic vessels and tissue overgrowth. The lymphatic vessels present in LM lesions may become blocked and enlarged as lymphatic fluid collects, forming a mass or cyst. Lesions are typically diagnosed during childhood and are often disfiguring and life threatening. Available treatments consist of sclerotherapy, surgical removal and therapies to diminish complications. We isolated lymphatic endothelial cells (LM-LEC) from a surgically removed microcystic LM lesion. LM-LEC and normal human dermal-LEC (HD-LEC) expressed endothelial (CD31, VE-Cadherin) as well as lymphatic endothelial (Podoplanin, PROX1, LYVE1)-specific markers. Targeted gene sequencing analysis in patient-derived LM-LEC revealed the presence of two mutations in class I phosphoinositide 3-kinases (PI3K) genes. One is an inherited, premature stop codon in the PI3K regulatory subunit PIK3R3. The second is a somatic missense mutation in the PI3K catalytic subunit PIK3CA; this mutation has been found in association with overgrowth syndromes and cancer growth. LM-LEC exhibited angiogenic properties: both cellular proliferation and sprouting in collagen were significantly increased compared with HD-LEC. AKT-Thr308 was constitutively hyper-phosphorylated in LM-LEC. Treatment of LM-LEC with PI3-Kinase inhibitors Wortmannin and LY294 decreased cellular proliferation and prevented the phosphorylation of AKT-Thr308 in both HD-LEC and LM-LEC. Treatment with the mTOR inhibitor rapamycin also diminished cellular proliferation, sprouting and AKT phosphorylation, but only in LM-LEC. Our results implicate disrupted PI3K-AKT signaling in LEC isolated from a human lymphatic malformation lesion.


Assuntos
Endotélio/enzimologia , Vasos Linfáticos/anormalidades , Mutação , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Endotélio/patologia , Feminino , Humanos , Masculino , Fosforilação , Sirolimo/farmacologia
11.
Pathology ; 46(5): 416-23, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24977735

RESUMO

Neuropilin-1 (NRP1) is a receptor for vascular endothelial growth factor (VEGF). A soluble isoform of Nrp1 (sNrp1) has not been described in the mouse. Our goal was to examine the expression of mouse sNrp1 during liver development and regeneration.sNrp1 was cloned from mouse liver. The expression of sNrp1 and VEGF was examined in mouse liver during post-natal development and regeneration using northern blot, western blot, in situ hybridisation, and immunohistochemical analyses. HGF/NRP1 binding was examined in vitro.A novel 588-amino acid sNrp1 isoform was found to contain the ligand binding regions of Nrp1. The adult liver expressed more sNrp1 than full-length Nrp1. In vivo, hepatocytes constitutively expressed VEGF and sNrp1 in the quiescent state. sNrp1 was highly up-regulated at P20, a time point coinciding with a plateau in liver and body weights. Following hepatectomy, endogenous levels of sNrp1 decreased during the rapid growth phase, and VEGF levels were highest just prior to and during the angiogenic phase. sNrp1 levels again rose 5-10 days post-hepatectomy, presumably to control regeneration. HGF protein bound NRP1 and binding was competed with sNRP1.We cloned a novel mouse sNrp1 isoform from liver and provide evidence that this endogenous angiogenesis inhibitor may regulate VEGF or HGF bioavailability during normal physiological growth and development as well as during liver regeneration.


Assuntos
Regeneração Hepática/fisiologia , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Neovascularização Fisiológica/fisiologia , Neuropilina-1/biossíntese , Animais , Sequência de Bases , Northern Blotting , Western Blotting , Humanos , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Homologia de Sequência do Ácido Nucleico , Fator A de Crescimento do Endotélio Vascular/biossíntese
12.
Cancer Res ; 74(14): 3716-26, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24812271

RESUMO

Invasion and dissemination of medulloblastoma within the central nervous system is the principal factor predicting medulloblastoma treatment failure and death. Netrin-1 is an axon guidance factor implicated in tumor and vascular biology, including in invasive behaviors. We found that exogenous netrin-1 stimulated invasion of human medulloblastoma cells and endothelial cells in contrast to VEGF-A, which promoted invasion of endothelial cells but not medulloblastoma cells. Furthermore, medulloblastoma cells expressed endogenous netrin-1 along with its receptors, neogenin and UNC5B. Blockades in endogenous netrin-1, neogenin, or UNC5B reduced medulloblastoma invasiveness. Neogenin blockade inhibited netrin-1-induced endothelial cells tube formation and recruitment of endothelial cells into Matrigel plugs, two hallmarks of angiogenesis. In patients with pediatric medulloblastoma, netrin-1 mRNA levels were increased 1.7-fold in medulloblastoma tumor specimens compared with control specimens from the same patient. Immunohistochemical analyses showed that netrin-1 was elevated in medulloblastoma tumors versus cerebellum controls. Notably, urinary levels of netrin-1 were 9-fold higher in patients with medulloblastoma compared with control individuals. Moreover, urinary netrin-1 levels were higher in patients with invasive medulloblastoma compared with patients with noninvasive medulloblastoma. Finally, we noted that urinary netrin-1 levels diminished after medulloblastoma resection in patients. Our results suggest netrin-1 is a candidate biomarker capable of detecting an invasive, disseminated phenotype in patients with medulloblastoma and predicting their disease status.


Assuntos
Meduloblastoma/genética , Meduloblastoma/patologia , Neovascularização Patológica/genética , Fatores de Crescimento Neural/genética , Proteínas Supressoras de Tumor/genética , Adolescente , Biomarcadores/metabolismo , Biomarcadores/urina , Encéfalo/patologia , Linhagem Celular Tumoral , Criança , Células Endoteliais/metabolismo , Ativação Enzimática/efeitos dos fármacos , Expressão Gênica , Humanos , Imageamento por Ressonância Magnética , Meduloblastoma/diagnóstico , Meduloblastoma/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica , Neovascularização Patológica/metabolismo , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/urina , Receptores de Netrina , Netrina-1 , Prognóstico , Receptores de Superfície Celular/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/farmacologia , Proteínas Supressoras de Tumor/urina
13.
Biochem Biophys Res Commun ; 448(2): 134-8, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24759231

RESUMO

Class 3 semaphorins are axonal guidance mediators and regulators of angiogenesis and tumor progression. Semaphorin 3A and 3F (SEMA3A&F) act by depolymerizing F-actin, resulting in cytoskeleton collapse. A key signaling step is that SEMA3A&F activates ABL2 tyrosine kinase, which activates p190RhoGAP, which in turn inactivates RhoA, thereby diminishing stress fiber formation and ensuing cell migration. We now demonstrate that Gleevec (imatinib, STI571), an ABL2 tyrosine kinase inhibitor, abrogates SEMA3A&F-induced stress fiber loss in glioblastoma cells and endothelial cells and diminishes their ability to inhibit migration. On the other hand, Sutent (sunitinib), a receptor tyrosine kinase inhibitor, did not rescue SEMA3A&F-induced collapsing activity. These results describe a novel property of Gleevec, its ability to antagonize semaphorins.


Assuntos
Benzamidas/farmacologia , Citoesqueleto/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas/farmacologia , Semaforina-3A/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Citoesqueleto/metabolismo , Células Endoteliais/citologia , Proteínas Ativadoras de GTPase/metabolismo , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Mesilato de Imatinib , Proteínas Tirosina Quinases/metabolismo , Semaforina-3A/genética
14.
Nat Commun ; 4: 2824, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24280686

RESUMO

Mechanisms governing the distinct temporal dynamics that characterize post-natal angiogenesis and lymphangiogenesis elicited by cutaneous wounds and inflammation remain unclear. RhoB, a stress-induced small GTPase, modulates cellular responses to growth factors, genotoxic stress and neoplastic transformation. Here we show, using RhoB null mice, that loss of RhoB decreases pathological angiogenesis in the ischaemic retina and reduces angiogenesis in response to cutaneous wounding, but enhances lymphangiogenesis following both dermal wounding and inflammatory challenge. We link these unique and opposing roles of RhoB in blood versus lymphatic vasculatures to the RhoB-mediated differential regulation of sprouting and proliferation in primary human blood versus lymphatic endothelial cells. We demonstrate that nuclear RhoB-GTP controls expression of distinct gene sets in each endothelial lineage by regulating VEZF1-mediated transcription. Finally, we identify a small-molecule inhibitor of VEZF1-DNA interaction that recapitulates RhoB loss in ischaemic retinopathy. Our findings establish the first intra-endothelial molecular pathway governing the phased response of angiogenesis and lymphangiogenesis following injury.


Assuntos
Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Linfangiogênese , Neovascularização Patológica , Doenças Retinianas/fisiopatologia , Proteína rhoB de Ligação ao GTP/fisiologia , Animais , Animais Recém-Nascidos , Linhagem da Célula/genética , Proteínas de Ligação a DNA , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Feminino , Regulação da Expressão Gênica , Inflamação/genética , Inflamação/fisiopatologia , Linfangiogênese/genética , Masculino , Camundongos , Neovascularização Patológica/genética , Doenças Retinianas/genética , Doenças Retinianas/patologia , Fatores de Transcrição , Cicatrização/genética , Cicatrização/fisiologia , Proteína rhoB de Ligação ao GTP/genética
15.
Angiogenesis ; 16(4): 939-52, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23892628

RESUMO

GATA-binding protein 2 (GATA2) and LIM domain only 2 (Lmo2) form common transcription complexes during hematopoietic differentiation. Here we show that these two transcription factors also play a key role in endothelial cells (EC) and lymphatic EC (LEC) function. Primary EC and tumor-associated blood vessels expressed GATA2 and Lmo2. VEGF-induced sprouting angiogenesis in both differentiating embryonic stem cells (embryoid bodies) and primary EC increased GATA2 and Lmo2 levels. Conversely, silencing of GATA2 and Lmo2 expression in primary EC inhibited VEGF-induced angiogenic activity, including EC migration and sprouting in vitro, two key steps of angiogenesis in vivo. This inhibition of EC function was associated with downregulated expression of neuropilin-2 (NRP2), a co-receptor of VEGFRs for VEGF, at the protein, mRNA and promoter levels. NRP2 overexpression partially rescued the impaired angiogenic sprouting in the GATA2/Lmo2 knockdown EC, confirming that GATA2 and Lmo2 mediated EC function, at least in part, by directly regulating NRP2 gene expression. Furthermore, it was found that primary LEC expressed GATA2 and Lmo2 as well. Silencing of GATA2 and Lmo2 expression in LEC inhibited VEGF-induced LEC sprouting, also in a NRP2-dependent manner. In conclusion, our results demonstrate that GATA2 and Lmo2 cooperatively regulate VEGF-induced angiogenesis and lymphangiogenesis via NRP2.


Assuntos
Células Endoteliais/fisiologia , Fator de Transcrição GATA2/fisiologia , Proteínas com Domínio LIM/fisiologia , Linfangiogênese/fisiologia , Neovascularização Fisiológica/fisiologia , Neuropilina-2/fisiologia , Transcrição Gênica , Animais , Linhagem Celular Tumoral , Corpos Embrioides , Células Endoteliais/metabolismo , Feminino , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica , Glioblastoma/irrigação sanguínea , Glioblastoma/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteínas com Domínio LIM/genética , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/patologia , Camundongos , Camundongos Nus , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Neuropilina-2/genética , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia
16.
FEBS Lett ; 587(11): 1650-5, 2013 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-23603393

RESUMO

Semaphorins are known modulators of axonal sprouting and angiogenesis. In the retina, we identified a distinct and almost exclusive expression of Semaphorin 3F in the outer layers. Interestingly, these outer retinal layers are physiologically avascular. Using functional in vitro models, we report potent anti-angiogenic effects of Semaphorin 3F on both retinal and choroidal vessels. In addition, human retinal pigment epithelium isolates from patients with pathologic neovascularization of the outer retina displayed reduced Semaphorin 3F expression in 10 out of 15 patients. Combined, these results elucidate a functional role for Semaphorin 3F in the outer retina where it acts as a vasorepulsive cue to maintain physiologic avascularity.


Assuntos
Proteínas Angiostáticas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Epitélio Pigmentado da Retina/metabolismo , Vasos Retinianos/metabolismo , Esferoides Celulares , Fator A de Crescimento do Endotélio Vascular/fisiologia
17.
Exp Neurol ; 247: 703-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23518418

RESUMO

Hydrocephalus is a condition characterized primarily by excessive accumulation of fluid in the ventricles of the brain for which there is currently no effective pharmacological treatment. Surgery, often accompanied by complications, is the only current treatment. Extensive research in our laboratory along with work from others has suggested a link between hydrocephalus and vascular function. We hypothesized that vascular endothelial growth factor (VEGF), the major angiogenic factor, could play a role in the pathogenesis of hydrocephalus. We tested this hypothesis by examining two predictions of such a link: first, that VEGF is present in many cases of clinical hydrocephalus; and second, that exogenous VEGF in an animal model could cause ventricular enlargement and tissue changes associated with hydrocephalus. Our results support the idea that VEGF elevation can potentiate hydrocephalus. The clinical relevance of this work is that anti-angiogenic drugs may be useful in patients with hydrocephalus, either alone or in combination with the currently available surgical treatments.


Assuntos
Epêndima/efeitos dos fármacos , Hidrocefalia/líquido cefalorraquidiano , Hidrocefalia/induzido quimicamente , Fator A de Crescimento do Endotélio Vascular/líquido cefalorraquidiano , Fator A de Crescimento do Endotélio Vascular/toxicidade , Adolescente , Adulto , Inibidores da Angiogênese/uso terapêutico , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Bevacizumab , Caderinas/metabolismo , Criança , Pré-Escolar , Modelos Animais de Doenças , Feminino , Humanos , Hidrocefalia/tratamento farmacológico , Lactente , Masculino , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem , beta Catenina/metabolismo
18.
Cell ; 152(5): 1065-76, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-23452854

RESUMO

Medulloblastoma is the most common pediatric malignant brain tumor. Although current therapies improve survival, these regimens are highly toxic and are associated with significant morbidity. Here, we report that placental growth factor (PlGF) is expressed in the majority of medulloblastomas, independent of their subtype. Moreover, high expression of PlGF receptor neuropilin 1 (Nrp1) correlates with poor overall survival in patients. We demonstrate that PlGF and Nrp1 are required for the growth and spread of medulloblastoma: PlGF/Nrp1 blockade results in direct antitumor effects in vivo, resulting in medulloblastoma regression, decreased metastasis, and increased mouse survival. We reveal that PlGF is produced in the cerebellar stroma via tumor-derived Sonic hedgehog (Shh) and show that PlGF acts through Nrp1-and not vascular endothelial growth factor receptor 1-to promote tumor cell survival. This critical tumor-stroma interaction-mediated by Shh, PlGF, and Nrp1 across medulloblastoma subtypes-supports the development of therapies targeting PlGF/Nrp1 pathway.


Assuntos
Neoplasias Cerebelares/patologia , Cerebelo/metabolismo , Meduloblastoma/patologia , Neuropilina-1/metabolismo , Proteínas da Gravidez/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Neoplasias Cerebelares/metabolismo , Humanos , Meduloblastoma/metabolismo , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Comunicação Parácrina , Fator de Crescimento Placentário , Transplante Heterólogo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
J Biol Chem ; 288(4): 2210-22, 2013 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-23195957

RESUMO

Glioblastomas are very difficult tumors to treat because they are highly invasive and disseminate within the normal brain, resulting in newly growing tumors. We have identified netrin-1 as a molecule that promotes glioblastoma invasiveness. As evidence, netrin-1 stimulates glioblastoma cell invasion directly through Matrigel-coated transwells, promotes tumor cell sprouting and enhances metastasis to lymph nodes in vivo. Furthermore, netrin-1 regulates angiogenesis as shown in specific angiogenesis assays such as enhanced capillary endothelial cells (EC) sprouting and by increased EC infiltration into Matrigel plugs in vivo, as does VEGF-A. This netrin-1 signaling pathway in glioblastoma cells includes activation of RhoA and cyclic AMP response element-binding protein (CREB). A novel finding is that netrin-1-induced glioblastoma invasiveness and angiogenesis are mediated by activated cathepsin B (CatB), a cysteine protease that translocates to the cell surface as an active enzyme and co-localizes with cell surface annexin A2 (ANXA2). The specific CatB inhibitor CA-074Me inhibits netrin-1-induced cell invasion, sprouting, and Matrigel plug angiogenesis. Silencing of CREB suppresses netrin-1-induced glioblastoma cell invasion, sprouting, and CatB expression. It is concluded that netrin-1 plays an important dual role in glioblastoma progression by promoting both glioblastoma cell invasiveness and angiogenesis in a RhoA-, CREB-, and CatB-dependent manner. Targeting netrin-1 pathways may be a promising strategy for brain cancer therapy.


Assuntos
Catepsina B/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Neovascularização Patológica , Fatores de Crescimento Neural/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Anexina A2/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular , Colágeno/química , Combinação de Medicamentos , Feminino , Inativação Gênica , Células HEK293 , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Laminina/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Modelos Biológicos , Invasividade Neoplásica , Netrina-1 , Ligação Proteica , Proteoglicanas/química , Proteínas Recombinantes/metabolismo
20.
Am J Pathol ; 181(5): 1573-84, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23022210

RESUMO

Oncosomes are tumor-derived microvesicles that transmit signaling complexes between cell and tissue compartments. Herein, we show that amoeboid tumor cells export large (1- to 10-µm diameter) vesicles, derived from bulky cellular protrusions, that contain metalloproteinases, RNA, caveolin-1, and the GTPase ADP-ribosylation factor 6, and are biologically active toward tumor cells, endothelial cells, and fibroblasts. We describe methods by which large oncosomes can be selectively sorted by flow cytometry and analyzed independently of vesicles <1 µm. Structures resembling large oncosomes were identified in the circulation of different mouse models of prostate cancer, and their abundance correlated with tumor progression. Similar large vesicles were also identified in human tumor tissues, but they were not detected in the benign compartment. They were more abundant in metastases. Our results suggest that tumor microvesicles substantially larger than exosome-sized particles can be visualized and quantified in tissues and in the circulation, and isolated and characterized using clinically adaptable methods. These findings also suggest a mechanism by which migrating tumor cells condition the tumor microenvironment and distant sites, thereby potentiating advanced disease.


Assuntos
Micropartículas Derivadas de Células/patologia , Neoplasias da Próstata/sangue , Neoplasias da Próstata/patologia , Fator 6 de Ribosilação do ADP , Animais , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Micropartículas Derivadas de Células/ultraestrutura , Citometria de Fluxo , Humanos , Masculino , Camundongos , Modelos Biológicos , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias da Próstata/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...