Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
BMC Med ; 20(1): 59, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35144591

RESUMO

INTRODUCTION: Bevacizumab improves survival outcomes in women diagnosed with epithelial ovarian cancer (EOC). Pre-clinical data showed that the c-MET/VEGFR-2 heterocomplex negates VEGF inhibition through activation of c-MET signalling, leading to a more invasive and metastatic phenotype. We evaluated the clinical significance of c-MET and VEGFR-2 co-localisation and its association with VEGF pathway-related single nucleotide polymorphisms (SNPs) in women participating in the phase 3 trial, ICON7 (ClinicalTrials.gov identifier: NCT00262847). MATERIALS AND METHODS: Patients had FIGO stage I-IIA grade 3/poorly differentiated or clear cell carcinoma or stage IIB-IV epithelial ovarian, primary peritoneal or fallopian tube cancer. Immunofluorescence staining for co-localised c-MET and VEGFR-2 on tissue microarrays and genotyping of germline DNA from peripheral blood leukocytes for VEGFA and VEGFR-2 SNPs was performed. The significance of these biomarkers was assessed against survival. RESULTS: Tissue microarrays from 178 women underwent immunofluorescence staining. Multivariable analysis showed that greater c-MET/VEGFR-2 co-localisation predicted worse OS in patients treated with bevacizumab after adjusting for FIGO stage and debulking surgery outcome (hazard ratio [HR] 1.034, 95% confidence interval [95%CI] 1.010-1.059). Women in the c-MET/VEGFR-2HIGH group treated with bevacizumab demonstrated significantly reduced OS (39.3 versus > 60 months; HR 2.00, 95%CI 1.08-3.72). Germline DNA from 449 women underwent genotyping. In the bevacizumab group, those women with the VEGFR-2 rs2305945 G/G variant had a trend towards shorter PFS compared with G/T or T/T variants (18.3 versus 23.0 months; HR 0.74, 95%CI 0.53-1.03). CONCLUSIONS: In bevacizumab-treated women diagnosed with EOC, high c-MET/VEGFR-2 co-localisation on tumour tissue and the VEGFR-2 rs2305945 G/G variant, which may be biologically related, were associated with worse survival outcomes.


Assuntos
Neoplasias Ovarianas , Receptor 2 de Fatores de Crescimento do Endotélio Vascular , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bevacizumab/uso terapêutico , Biomarcadores , Carcinoma Epitelial do Ovário/tratamento farmacológico , Carcinoma Epitelial do Ovário/genética , Feminino , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/uso terapêutico
2.
J Biol Chem ; 289(15): 10488-10501, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24563483

RESUMO

Heparan sulfate (HS) is a component of cell surface and extracellular matrix proteoglycans that regulates numerous signaling pathways by binding and activating multiple growth factors and chemokines. The amount and pattern of HS sulfation are key determinants for the assembly of the trimolecular, HS-growth factor-receptor, signaling complex. Here we demonstrate that HS 6-O-sulfotransferases 1 and 2 (HS6ST-1 and HS6ST-2), which perform sulfation at 6-O position in glucosamine in HS, impact ovarian cancer angiogenesis through the HS-dependent HB-EGF/EGFR axis that subsequently modulates the expression of multiple angiogenic cytokines. Down-regulation of HS6ST-1 or HS6ST-2 in human ovarian cancer cell lines results in 30-50% reduction in glucosamine 6-O-sulfate levels in HS, impairing HB-EGF-dependent EGFR signaling and diminishing FGF2, IL-6, and IL-8 mRNA and protein levels in cancer cells. These cancer cell-related changes reduce endothelial cell signaling and tubule formation in vitro. In vivo, the development of subcutaneous tumor nodules with reduced 6-O-sulfation is significantly delayed at the initial stages of tumor establishment with further reduction in angiogenesis occurring throughout tumor growth. Our results show that in addition to the critical role that 6-O-sulfate moieties play in angiogenic cytokine activation, HS 6-O-sulfation level, determined by the expression of HS6ST isoforms in ovarian cancer cells, is a major regulator of angiogenic program in ovarian cancer cells impacting HB-EGF signaling and subsequent expression of angiogenic cytokines by cancer cells.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Neoplasias Ovarianas/metabolismo , Sulfotransferases/metabolismo , Animais , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/química , Citocinas/metabolismo , Dissacarídeos/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glucosamina/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Transplante de Neoplasias , Neovascularização Patológica , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Transdução de Sinais
3.
Angiogenesis ; 18(1): 47-68, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25269667

RESUMO

Late outgrowth endothelial cells (OECs) that originate from peripheral blood mononuclear cells ex vivo have phenotypic and functional properties of mature endothelial cells. Given the potential therapeutic applications of OECs, understanding their biology is crucial. We have identified two distinct OEC populations based on differential expression of the cell surface marker CD34. OEC colonies lacked CD34 expression (CD34-), expressed CD34 in the majority of cells (CD34+), or showed a mixed expression pattern within a colony (CD34+/-). CD34+ and CD34- OECs were negative for hematopoietic cell marker CD45 and expressed the endothelial cell surface markers CD31, CD146, CD105, and VEGFR-2. Functionally CD34- and CD34+ OECs exhibited strikingly distinct behaviors. CD34- OECs, unlike CD34+ OECs, were capable of sprouting, formed tubes, and responded to angiogenic growth factors in vitro. In vivo, CD34- OECs formed endothelial tubes, while CD34+ OECs, despite being unable to form tubes, promoted infiltration of murine vasculature. Global gene expression profiling in CD34- and CD34+ OECs identified functional importance of the MMP-1/PAR-1 pathway in CD34- OECs. MMP-1 stimulated the expression of VEGFR-2, neuropilin-1, neuropilin-2, and CXCR4 and activated ERK1/2, whereas down-regulation of PAR-1 in CD34- OECs resulted in impaired angiogenic responses in vitro and reduced VEGFR-2 levels. In contrast, the CD34+ OEC colonies expressed high levels of the progenitor cell marker ALDH, which was absent in CD34- OECs. In summary, we show that OECs can be classified into functionally mature endothelial cells (CD34- OECs) that depend on the MMP-1/PAR-1 pathway and progenitor-like angiogenesis-promoting cells (CD34+ OECs).


Assuntos
Antígenos CD34/metabolismo , Separação Celular/métodos , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Neovascularização Fisiológica/fisiologia , Animais , Movimento Celular , Colágeno , Primers do DNA/genética , Combinação de Medicamentos , Células Endoteliais/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Immunoblotting , Laminina , Metaloproteinase 1 da Matriz/metabolismo , Camundongos , Análise em Microsséries , Proteoglicanas , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
Biochem Soc Trans ; 42(6): 1596-600, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25399576

RESUMO

Angiogenesis has emerged as a novel target for anti-cancer therapies through randomized clinical trials that tested the benefit of adding vascular endothelial growth factor (VEGF) inhibitors to conventional cytotoxic therapies. However, despite improvements in the progression-free survival, the benefit in overall survival is modest. Tumour angiogenesis is regulated by a number of angiogenic cytokines. Thus innate or acquired resistance to VEGF inhibitors can be caused, at least in part, through expression of other angiogenic cytokines, including fibroblast growth factor 2 (FGF2), interleukin 8 (IL-8) and stromal-cell-derived factor 1α (SDF-1α), which make tumours insensitive to VEGF signalling pathway inhibition. The majority of angiogenic cytokines, including VEGF-A, FGF2, IL-8 and SDF-1α, manifest an obligate dependence on heparan sulfate (HS) for their biological activity. This mandatory requirement of angiogenic cytokines for HS identifies HS as a potential target for novel anti-angiogenic therapy. Targeting multiple angiogenic cytokines with HS mimetics may represent an opportunity to inhibit tumour angiogenesis more efficiently. Our published studies and unpublished work have demonstrated the feasibility of generating synthetic HS fragments of defined structure with biological activity against a number of angiogenic cytokines.


Assuntos
Inibidores da Angiogênese/farmacologia , Heparitina Sulfato/farmacologia , Oligossacarídeos/farmacologia , Humanos
5.
Nat Rev Cancer ; 5(7): 505-15, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16069815

RESUMO

Focal-adhesion kinase (FAK) is an important mediator of growth-factor signalling, cell proliferation, cell survival and cell migration. Given that the development of malignancy is often associated with perturbations in these processes, it is not surprising that FAK activity is altered in cancer cells. Mouse models have shown that FAK is involved in tumour formation and progression, and other studies showing that FAK expression is increased in human tumours make FAK a potentially important new therapeutic target.


Assuntos
Neoplasias/fisiopatologia , Proteínas Tirosina Quinases/fisiologia , Animais , Movimento Celular/fisiologia , Proliferação de Células , Sobrevivência Celular/fisiologia , Progressão da Doença , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Camundongos , Modelos Animais , Transdução de Sinais
6.
J Biol Chem ; 287(43): 36132-46, 2012 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22927437

RESUMO

Fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor 165 (VEGF(165)) are potent pro-angiogenic growth factors that play a pivotal role in tumor angiogenesis. The activity of these growth factors is regulated by heparan sulfate (HS), which is essential for the formation of FGF2/FGF receptor (FGFR) and VEGF(165)/VEGF receptor signaling complexes. However, the structural characteristics of HS that determine activation or inhibition of such complexes are only partially defined. Here we show that ovarian tumor endothelium displays high levels of HS sequences that harbor glucosamine 6-O-sulfates when compared with normal ovarian vasculature where these sequences are also detected in perivascular area. Reduced HS 6-O-sulfotransferase 1 (HS6ST-1) or 6-O-sulfotransferase 2 (HS6ST-2) expression in endothelial cells impacts upon the prevalence of HS 6-O-sulfate moieties in HS sequences, which consist of repeating short, highly sulfated S domains interspersed by transitional N-acetylated/N-sulfated domains. 1-40% reduction in 6-O-sulfates significantly compromises FGF2- and VEGF(165)-induced endothelial cell sprouting and tube formation in vitro and FGF2-dependent angiogenesis in vivo. Moreover, HS on wild-type neighboring endothelial or smooth muscle cells fails to restore endothelial cell sprouting and tube formation. The affinity of FGF2 for HS with reduced 6-O-sulfation is preserved, although FGFR1 activation is inhibited correlating with reduced receptor internalization. These data show that 6-O-sulfate moieties in endothelial HS are of major importance in regulating FGF2- and VEGF(165)-dependent endothelial cell functions in vitro and in vivo and highlight HS6ST-1 and HS6ST-2 as potential targets of novel antiangiogenic agents.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Heparitina Sulfato/biossíntese , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Células Cultivadas , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/genética , Heparitina Sulfato/genética , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Proteínas de Neoplasias/genética , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Sulfotransferases/genética , Sulfotransferases/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
7.
Curr Opin Cell Biol ; 17(5): 542-7, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16099634

RESUMO

Src kinase controls cellular adhesions, including cadherin-based intercellular adhesions and integrin-mediated cell-matrix adhesions. In epithelial cells, Src activation, or increased signalling from migratory growth factor receptors via Src, induces an adhesion switch that enhances dynamic cell-matrix adhesions and migratory capacity while suppressing intercellular contact. Moreover, Src and the associated tyrosine kinase FAK are at the heart of the recently identified crosstalk between integrin- and cadherin-mediated adhesions of epithelial cells, particularly during the epithelial-to-mesenchymal transition.


Assuntos
Adesão Celular/fisiologia , Diferenciação Celular , Células Epiteliais/metabolismo , Mesoderma/metabolismo , Transdução de Sinais/fisiologia , Quinases da Família src/metabolismo , Animais , Caderinas/fisiologia , Endocitose/fisiologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Humanos , Integrinas/fisiologia , Mesoderma/citologia , Mesoderma/fisiologia , Modelos Biológicos , Neoplasias/metabolismo , Quinases da Família src/fisiologia
8.
J Org Chem ; 77(18): 7823-43, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22900939

RESUMO

A diastereomerically pure cyanohydrin, preparable on kilogram scale, is efficiently converted in one step into a novel L-iduronamide. A new regioselective acylation of this iduronamide and a new mild amide hydrolysis method mediated by amyl nitrite enables short, scalable syntheses of an L-iduronate diacetate C-4 acceptor, and also L-iduronate C-4 acceptor thioglycosides. Efficient conversions of these to a range of heparin-related gluco-ido disaccharide building blocks (various C-4 protection options) including efficient multigram access to key heparin-building block ido-thioglycoside donors are described. A 1-OAc disaccharide is converted into a heparin-related tetrasaccharide, via divergence to both acceptor and donor disaccharides. X-ray and NMR data of the 1,2-diacetyl iduronate methyl ester and the analogous iduronamide show that while both adopt (1)C(4) conformations in solution, the iduronate ester adopts the (4)C(1) conformation in solid state. An X-ray structure is also reported for the novel, (4)C(1)-conformationally locked bicyclic 1,6-anhydro iduronate lactone along with an X-ray structures of a novel distorted (4)C(1) iduronate 4,6-lactone. Deuterium labeling also provides mechanistic insight into the formation of lactone products during the novel amyl nitrite-mediated hydrolysis of iduronamide into the parent iduronic acid functionality.


Assuntos
Amidas/química , Dissacarídeos/química , Heparina/análogos & derivados , Heparina/química , Heparina/síntese química , Ácido Idurônico/química , Oligossacarídeos/química , Dissacarídeos/síntese química , Espectroscopia de Ressonância Magnética , Conformação Molecular , Estereoisomerismo
9.
Nat Cell Biol ; 4(8): 632-8, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12134161

RESUMO

Although Src expression and activity are often elevated in colon cancer, the precise consequences of overexpression of the non-catalytic Src homology (SH) domains, or enhanced catalytic activity, are unknown. We show that, in KM12C colon cancer cells, elevated Src activity causes the components of adherens junctions, including vinculin, to be redistributed to Src-induced integrin adhesion complexes. Specifically, elevated Src activity blocks proper assembly of cell cell contacts after cells are switched from media containing a low level of calcium to media containing a high level of calcium, and E-cadherin remains internalized. In contrast, although elevated expression of the non-catalytic domains of Src is sufficient to induce assembly of integrin adhesion complexes, it does not induce disorganization of E-cadherin-associated intercellular contacts. Surprisingly, Src-induced disruption of E-cadherin localization requires specific integrin signalling, because E-cadherin redistribution is blocked by loss of cell-matrix interaction, or by inhibitory antibodies to alpha(v) or beta(1) integrin subunits. Furthermore, phosphorylation of the integrin-regulated focal adhesion kinase (FAK) on Src-specific sites is required for Src-induced de-regulation of E-cadherin, demonstrating interdependence between integrin-induced signals and cadherin-associated adhesion changes induced by Src.


Assuntos
Caderinas/metabolismo , Neoplasias do Colo/metabolismo , Integrinas/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Anticorpos/farmacologia , Antígenos CD/metabolismo , Adesão Celular , Humanos , Integrina alfa2 , Integrina alfaV , Integrina beta1/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/química , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas , Domínios de Homologia de src
10.
Biochem J ; 415(2): 197-206, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18588508

RESUMO

Recent clinical data indicates that the emergence of mutant drug-resistant kinase alleles may be particularly relevant for targeted kinase inhibitors. In order to explore how different classes of targeted therapies impact upon resistance mutations, we performed EGFR (epidermal-growth-factor receptor) resistance mutation screens with erlotinib, lapatinib and CI-1033. Distinct mutation spectra were generated with each inhibitor and were reflective of their respective mechanisms of action. Lapatinib yielded the widest variety of mutations, whereas mutational variability was lower in the erlotinib and CI-1033 screens. Lapatinib was uniquely sensitive to mutations of residues located deep within the selectivity pocket, whereas mutation of either Gly(796) or Cys(797) resulted in a dramatic loss of CI-1033 potency. The clinically observed T790M mutation was common to all inhibitors, but occurred with varying frequencies. Importantly, the presence of C797S with T790M in the same EGFR allele conferred complete resistance to erlotinib, lapatinib and CI-1033. The combination of erlotinib and CI-1033 effectively reduced the number of drug-resistant clones, suggesting a possible clinical strategy to overcome drug resistance. Interestingly, our results also indicate that co-expression of ErbB2 (v-erb-b2 erythroblastic leukaemia viral oncogene homologue 2) has an impact upon the EGFR resistance mutations obtained, suggesting that ErbB2 may play an active role in the acquisition of drug-resistant mutations.


Assuntos
Resistência a Medicamentos/genética , Receptores ErbB/antagonistas & inibidores , Mutação , Inibidores de Proteínas Quinases/farmacologia , Animais , Linhagem Celular , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Combinação de Medicamentos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Cloridrato de Erlotinib , Immunoblotting , Imunoprecipitação , Interleucina-3/metabolismo , Lapatinib , Camundongos , Morfolinas/farmacologia , Quinazolinas/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
11.
Eur J Cell Biol ; 87(8-9): 569-79, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18562041

RESUMO

Overexpression of active Src induces invadopodia formation and associated matrix degradation in KM12C colon cancer cells. FAK is present with active Src at sites of matrix-degrading activity (invadopodia), specifically residing in rings surrounding the cortactin-containing invadopodia cores. Since FAK is a key effector protein in many aspects of Src function, we addressed whether FAK is necessary for Src-induced invadopodia formation and matrix degradation in KM12C colon cancer cells. We found that efficient knockdown of FAK expression by siRNA had no effect on invadopodia formation or matrix degradation. However, overexpression of FAK could actually suppress invadopodia formation and matrix degradation. FAK phosphorylation on the putative auto-phosphorylation tyrosine 397 and the Src-specific sites are all required for overexpressed FAK to inhibit invadopodia formation, while the kinase activity of exogenous FAK is apparently not required. These data imply that kinase activities other than FAK auto-phosphorylation may contribute to the phosphorylation of FAK tyrosine 397 in some contexts to promote an activity of FAK that can counteract invadopodia formation. Further work is required to determine how the strength of signalling through FAK suppresses invadopodia, but we propose that FAK controls the balance of adhesion types in cells, and that this is one of the determinants of whether a cancer cell can make stable matrix-degrading invadopodia.


Assuntos
Citoesqueleto de Actina/enzimologia , Neoplasias do Colo/enzimologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Citoesqueleto de Actina/ultraestrutura , Adesão Celular , Diferenciação Celular , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Humanos , Fosforilação , Transdução de Sinais , Domínios de Homologia de src
12.
Mol Biol Cell ; 15(6): 2794-803, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15075377

RESUMO

Elevated Src kinase in epithelial cancer cells induces adhesion changes that are associated with a mesenchymal-like state. We recently showed that Src induces dynamic integrin adhesions in KM12C colon cancer cells, whereas E-cadherin-dependent cell-cell contacts become disorganized. This promotes a fibroblastic-like morphology and expression of the mesenchymal marker vimentin. Furthermore, Src-induced deregulation of E-cadherin, and the associated mesenchymal transition, is dependent on integrin signaling (Avizienyte et al., Nat. Cell Biol. 2002, 4, 632-638), although the nature of downstream signals that mediate these Src- and integrin-dependent effects are unknown. Here we show that the SH2 and SH3 domains of Src mediate peripheral accumulation of phospho-myosin, leading to integrin adhesion complex assembly, whereas loss of SH2 or SH3 function restores normal regulation of E-cadherin and inhibits vimentin expression. Inhibitors of MEK, ROCK, or MLCK also suppress peripheral accumulation of phospho-myosin and Src-induced formation of integrin-dependent adhesions, whereas at the same time restoring E-cadherin redistribution to regions of cell-cell contact. Our data therefore implicate peripheral phospho-myosin activity as a point of convergence for upstream signals that regulate integrin- and E-cadherin-mediated adhesions. This further implicates spatially regulated contractile force as a determinant of epithelial cell plasticity, particularly in cancer cells that can switch between epithelial and mesenchymal-like states.


Assuntos
Caderinas/metabolismo , Diferenciação Celular , Células Epiteliais/metabolismo , Mesoderma/metabolismo , Miosinas/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Domínios de Homologia de src , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Células Epiteliais/citologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Integrinas/metabolismo , Sistema de Sinalização das MAP Quinases , Mesoderma/citologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/metabolismo , Fosforilação , Mutação Puntual , Proteínas Proto-Oncogênicas pp60(c-src)/química , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Soro , Transfecção , Vimentina/metabolismo
13.
Cancer Res ; 65(4): 1335-42, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15735019

RESUMO

Src tyrosine kinase expression and activity are elevated during colon cancer progression. How this contributes to the malignant phenotype is not fully understood. We show that in KM12C colon carcinoma cells, expression of kinase-deficient Src proteins (SrcMF and Src251) does not alter cell growth. Src kinase activity is required for turnover of cell-matrix adhesions and, in particular, the Src-dependent phosphorylation of focal adhesion kinase (FAK) is required for their disassembly. Surprisingly, we found that expression of SrcMF or Src251 resulted in increased tyrosine phosphorylation of FAK on Tyr(407), Tyr(576), Tyr(577), and Tyr(861), which are considered to be Src kinase substrates. This Src kinase-independent phosphorylation of FAK required an intact Src SH2 domain that mediates association of Src and FAK at peripheral adhesions. Use of a novel highly potent and selective Src kinase inhibitor AP23464 combined with experiments in Src/Fyn/Yes-deficient fibroblasts showed that increased phosphorylation of FAK in cells expressing SrcMF did not require Src-like kinases. However, specific phosphorylation on Tyr(925) of FAK was not evident in SrcMF- or Src251-expressing cells, and lack of Src kinase-dependent phosphorylation on this site was associated with impaired adhesion turnover. Our data show that Src kinase activity is required for adhesion turnover associated with cell migration in cancer cells and that, in addition to the catalytic activity, Src also acts as an adaptor to recruit other kinases that can phosphorylate key substrates including FAK. These studies have implications for tumor progression with respect to the use of Src kinase inhibitors.


Assuntos
Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Proteínas Tirosina Quinases/metabolismo , Quinases da Família src/metabolismo , Animais , Sítios de Ligação , Catálise , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Feminino , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Camundongos , Camundongos Nus , Fosforilação , Transdução de Sinais , Especificidade por Substrato , Tirosina/metabolismo , Domínios de Homologia de src
15.
PLoS One ; 11(8): e0159739, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27490176

RESUMO

Heparan sulphate (HS), a ubiquitously expressed glycosaminoglycan (GAG), regulates multiple cellular functions by mediating interactions between numerous growth factors and their cell surface cognate receptors. However, the structural specificity of HS in these interactions remains largely undefined. Here, we used completely synthetic, structurally defined, alternating N-sulfated glucosamine (NS) and 2-O-sulfated iduronate (IS) residues to generate dodecasaccharides ([NSIS]6) that contained no, one or six glucosamine 6-O-sulfates (6S). The aim was to address how 6S contributes to the potential of defined HS dodecasaccharides to inhibit the angiogenic growth factors FGF2 and VEGF165, in vitro and in vivo. We show that the addition of a single 6S at the non-reducing end of [NSIS]6, i.e. [NSIS6S]-[NSIS]5, significantly augments the inhibition of FGF2-dependent endothelial cell proliferation, migration and sprouting in vitro when compared to the non-6S variant. In contrast, the fully 6-O-sulfated dodecasaccharide, [NSIS6S]6, is not a potent inhibitor of FGF2. Addition of a single 6S did not significantly improve inhibitory properties of [NSIS]6 when tested against VEGF165-dependent endothelial cell functions.In vivo, [NSIS6S]-[NSIS]5 blocked FGF2-dependent blood vessel formation without affecting tumor growth. Reduction of non-FGF2-dependent ovarian tumor growth occurred when [NSIS6S]-[NSIS]5 was combined with cisplatin. The degree of inhibition by [NSIS6S]-[NSIS]5 in combination with cisplatin in vivo equated with that induced by bevacizumab and sunitinib when administered with cisplatin. Evaluation of post-treatment vasculature revealed that [NSIS6S]-[NSIS]5 treatment had the greatest impact on tumor blood vessel size and lumen formation. Our data for the first time demonstrate that synthetic, structurally defined oligosaccharides have potential to be developed as active anti-angiogenic agents that sensitize tumors to chemotherapeutic agents.


Assuntos
Inibidores da Angiogênese/síntese química , Cisplatino/uso terapêutico , Glucosamina/análogos & derivados , Oligossacarídeos/síntese química , Neoplasias Ovarianas/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Inibidores da Angiogênese/toxicidade , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cisplatino/toxicidade , Quimioterapia Combinada , Feminino , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/metabolismo , Glucosamina/química , Heparitina Sulfato/química , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oligossacarídeos/uso terapêutico , Oligossacarídeos/toxicidade , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Chem Commun (Camb) ; 51(72): 13846-9, 2015 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-26234943

RESUMO

The multigram-scale synthesis of a sulfation-site programmed heparin-like dodecasaccharide is described. Evaluation alongside dodecasaccharides lacking this single glucosamine O6-sulfation, or having per-O6-sulfation, shows that site-specific modification of the terminal glucosamine dramatically interconverts regulation of in vitro and in vivo biology mediated by the two important chemokines, CXCL12 (SDF1α) or CXCL8 (IL-8).


Assuntos
Quimiocina CXCL12/antagonistas & inibidores , Heparitina Sulfato/química , Heparitina Sulfato/farmacologia , Interleucina-8/antagonistas & inibidores , Oligossacarídeos/química , Oligossacarídeos/farmacologia , Sequência de Carboidratos , Ensaios de Migração de Leucócitos , Células Endoteliais da Veia Umbilical Humana , Humanos , Leucócitos
17.
Expert Opin Pharmacother ; 4(2): 227-34, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12562313

RESUMO

Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that plays a pivotal role in signal transduction at integrin-linked cellular adhesions, which mediate cell contact with the extracellular matrix. It has been shown to play a role in the survival of anchorage-dependent cells and to be essential for integrin-linked cell migration - processes that are likely to play important roles in the development of malignancies. FAK is upregulated in a wide variety of human epithelial cancers, with expression being closely correlated to invasive potential. Recently, evidence has emerged directly linking FAK expression to tumour development in vivo, raising the possibility that intervention strategies to block FAK function may potentially provide an opportunity for the development of anticancer therapeutics.


Assuntos
Neoplasias/metabolismo , Proteínas Tirosina Quinases/fisiologia , Animais , Movimento Celular , Inibidores Enzimáticos/farmacologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Adesões Focais , Humanos , Invasividade Neoplásica , Neoplasias/patologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/química , Transdução de Sinais , Regulação para Cima
18.
Nat Commun ; 4: 2016, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23828390

RESUMO

Heparin-like oligosaccharides mediate numerous important biological interactions, of which many are implicated in various diseases. Synthetic improvements are central to the development of such oligosaccharides as therapeutics and, in addition, there are no methods to elucidate the pharmacokinetics of structurally defined heparin-like oligosaccharides. Here we report an efficient two-cycle [4+4+4] tetrasaccharide-iteration-based approach for rapid chemical synthesis of a structurally defined heparin-related dodecasaccharide, combined with the incorporation of a latent aldehyde tag, unmasked in the final step of chemical synthesis, providing a generic end group for labelling/conjugation. We exploit this latent aldehyde tag for (3)H radiolabelling to provide the first example of this kind of agent for monitoring in vivo tissue distribution and in vivo stability of a biologically active, structurally defined heparin related dodecasaccharide. Such studies are critical for the development of related saccharide therapeutics, and the data here establish that a biologically active, synthetic, heparin-like dodecasaccharide provides good organ distribution, and serum lifetimes relevant to developing future oligosaccharide therapeutics.


Assuntos
Marcação por Isótopo , Oligossacarídeos/síntese química , Trítio/metabolismo , Animais , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Feminino , Heparina , Rim/metabolismo , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos SCID , Oligossacarídeos/química , Reprodutibilidade dos Testes , Distribuição Tecidual
19.
PLoS One ; 5(7): e11644, 2010 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-20657775

RESUMO

BACKGROUND: Heparan sulfate (HS) is an important regulator of the assembly and activity of various angiogenic signalling complexes. However, the significance of precisely defined HS structures in regulating cytokine-dependent angiogenic cellular functions and signalling through receptors regulating angiogenic responses remains unclear. Understanding such structure-activity relationships is important for the rational design of HS fragments that inhibit HS-dependent angiogenic signalling complexes. METHODOLOGY/PRINCIPAL FINDINGS: We synthesized a series of HS oligosaccharides ranging from 7 to 12 saccharide residues that contained a repeating disaccharide unit consisting of iduronate 2-O-sulfate linked to glucosamine with or without N-sulfate. The ability of oligosaccharides to compete with HS for FGF2 and VEGF165 binding significantly increased with oligosaccharide length and sulfation. Correspondingly, the inhibitory potential of oligosaccharides against FGF2- and VEGF165-induced endothelial cell responses was greater in longer oligosaccharide species that were comprised of disaccharides bearing both 2-O- and N-sulfation (2SNS). FGF2- and VEGF165-induced endothelial cell migration were inhibited by longer 2SNS oligosaccharide species with 2SNS dodecasaccharide activity being comparable to that of receptor tyrosine kinase inhibitors targeting FGFR or VEGFR-2. Moreover, the 2SNS dodecasaccharide ablated FGF2- or VEGF165-induced phosphorylation of FAK and assembly of F-actin in peripheral lamellipodia-like structures. In contrast, FGF2-induced endothelial cell proliferation was only moderately inhibited by longer 2SNS oligosaccharides. Inhibition of FGF2- and VEGF165-dependent endothelial tube formation strongly correlated with oligosaccharide length and sulfation with 10-mer and 12-mer 2SNS oligosaccharides being the most potent species. FGF2- and VEGF165-induced activation of MAPK pathway was inhibited by biologically active oligosaccharides correlating with the specific phosphorylation events in FRS2 and VEGFR-2, respectively. CONCLUSION/SIGNIFICANCE: These results demonstrate structure-function relationships for synthetic HS saccharides that suppress endothelial cell migration, tube formation and signalling induced by key angiogenic cytokines.


Assuntos
Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Heparitina Sulfato/síntese química , Heparitina Sulfato/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Oligossacarídeos/síntese química , Oligossacarídeos/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Heparitina Sulfato/química , Humanos , Immunoblotting , Microscopia de Fluorescência , Oligossacarídeos/química , Rodaminas/farmacologia , Relação Estrutura-Atividade , Cicatrização/efeitos dos fármacos
20.
Exp Cell Res ; 313(15): 3175-88, 2007 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-17651734

RESUMO

Src controls the dynamic actin cytoskeleton in fibroblasts and in cancer cells, although it is not known how direct its effects are. Using FRET/FLIM imaging, we found that wild type Src associates directly, or indirectly, with peripheral beta-actin at integrin adhesions after serum stimulation, and that an active Src kinase domain is essential. Beta-actin can be directly tyrosine-phosphorylated by Src in vitro, and in a Src-dependent manner in cells. Moreover, beta-actin dynamics are suppressed when Src is rendered kinase-inactive. Surprisingly, debilitating mutations in the Src SH2 or SH3 domains do not suppress association of Src with beta-actin. This may therefore be an example of a spatially regulated Src kinase/substrate interaction that is controlling peripheral actin dynamics. Interestingly, there is no FRET between Src and beta-actin at cadherin-mediated cell-cell contacts, despite apparent co-localization there, demonstrating precise spatial specificity of Src/beta-actin complexes.


Assuntos
Actinas/fisiologia , Quinases da Família src/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Comunicação Celular , Linhagem Celular Tumoral , Neoplasias do Colo , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Ativação Enzimática , Transferência Ressonante de Energia de Fluorescência , Humanos , Dados de Sequência Molecular , Mutação , Fosforilação , Ligação Proteica , Transdução de Sinais , Domínios de Homologia de src , Quinases da Família src/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA