Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Ecology ; 98(6): 1548-1559, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28266031

RESUMO

The persistence and geographic expansion of dryland forests in the 21st century will be influenced by how climate change supports the demographic processes associated with tree regeneration. Yet, the way that climate change may alter regeneration is unclear. We developed a quantitative framework that estimates forest regeneration potential (RP) as a function of key environmental conditions for ponderosa pine, a key dryland forest species. We integrated meteorological data and climate projections for 47 ponderosa pine forest sites across the western United States, and evaluated RP using an ecosystem water balance model. Our primary goal was to contrast conditions supporting regeneration among historical, mid-21st century and late-21st century time frames. Future climatic conditions supported 50% higher RP in 2020-2059 relative to 1910-2014. As temperatures increased more substantially in 2060-2099, seedling survival decreased, RP declined by 50%, and the frequency of years with very low RP increased from 25% to 58%. Thus, climate change may initially support higher RP and increase the likelihood of successful regeneration events, yet will ultimately reduce average RP and the frequency of years with moderate climate support of regeneration. Our results suggest that climate change alone may begin to restrict the persistence and expansion of dryland forests by limiting seedling survival in the late 21st century.


Assuntos
Mudança Climática , Florestas , Ecossistema , Pinus ponderosa , Árvores
2.
Appl Opt ; 53(24): 5344-52, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25321104

RESUMO

The generation of well-calibrated radiometric measurements from imaging spectrometer data requires careful consideration of all influencing factors, as well as an instrument calibration based on a detailed sensor model. Deviations of ambient parameters (i.e., pressure, humidity, temperature) from standard laboratory conditions during airborne operations can lead to biases that should be accounted for and properly compensated by using dedicated instrument models. This study introduces a model for the airborne imaging spectrometer airborne prism experiment (APEX), describing the impact of spectral shifts as well as polarization effects on the radiometric system response due to changing ambient parameters. Key issues are related to changing properties of the dichroic coating applied to the dispersing elements within the optical path. We present a model based on discrete numerical simulations. With the improved modeling approach, we predict radiometric biases with an root mean square error (RMSE) below 1%, leading to a substantial improvement of radiometric stability and predictability of system behavior.

3.
Curr Mol Med ; 14(2): 221-34, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24467206

RESUMO

Rho guanine exchange factors (GEFs) are a large, diverse family of proteins defined by their ability to catalyze the exchange of GDP for GTP on small GTPase proteins such as Rho family members. GEFs act as integrators from varied intra- and extracellular sources to promote spatiotemporal activity of Rho GTPases that control signaling pathways regulating cell proliferation and movement. Here we review recent studies elucidating roles of RhoGEF proteins in cell motility. Emphasis is placed on Dbl-family GEFs and connections to development, integrin signaling to Rho GTPases regulating cell adhesion and movement, and how these signals may enhance tumor progression. Moreover, RhoGEFs have additional domains that confer distinctive functions or specificity. We will focus on a unique interaction between Rgnef (also termed Arhgef28 or p190RhoGEF) and focal adhesion kinase (FAK), a non-receptor tyrosine kinase that controls migration properties of normal and tumor cells. This Rgnef-FAK interaction activates canonical GEF-dependent RhoA GTPase activity to govern contractility and also functions as a scaffold in a GEF-independent manner to enhance FAK activation. Recent studies have also brought to light the importance of specific regions within the Rgnef pleckstrin homology (PH) domain for targeting the membrane. As revealed by ongoing Rgnef-FAK investigations, exploring GEF roles in cancer will yield fundamental new information on the molecular mechanisms promoting tumor spread and metastasis.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Animais , Movimento Celular/genética , Movimento Celular/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/genética , Humanos , Modelos Biológicos , Fatores de Troca de Nucleotídeo Guanina Rho/genética
4.
Oncogene ; 31(22): 2783-93, 2012 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-21963850

RESUMO

Tyrosine kinase receptors have an essential role in various aspects of tumor progression. In particular, epidermal growth factor receptor (EGFR) and its ligands have been implicated in the growth and dissemination of a wide array of human carcinomas. Here, we describe an EGFR-mediated signaling pathway that regulates human pancreatic carcinoma cell invasion and metastasis, yet does not influence the growth of primary tumors. In fact, ligation/activation of EGFR induces Src-dependent phosphorylation of two critical tyrosine residues of p130CAS, leading to the assembly of a Crk-associated substrate (CAS)/Nck1 complex that promotes Ras-associated protein-1 (Rap1) signaling. Importantly, GTP loading of Rap1 is specifically required for pancreatic carcinoma cell migration on vitronectin but not on collagen. Furthermore, Rap1 activation is required for EGFR-mediated metastasis in vivo without impacting primary tumor growth. These findings identify a molecular pathway that promotes the invasive/metastatic properties of human pancreatic carcinomas driven by EGFR.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Movimento Celular , Receptores ErbB/metabolismo , Proteínas Oncogênicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/secundário , Proteínas de Ligação a Telômeros/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Western Blotting , Proliferação de Células , Células Cultivadas , Embrião de Galinha , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Metástase Neoplásica , Proteínas Oncogênicas/antagonistas & inibidores , Proteínas Oncogênicas/genética , RNA Interferente Pequeno/genética , Complexo Shelterina , Proteínas de Ligação a Telômeros/antagonistas & inibidores , Proteínas de Ligação a Telômeros/genética
5.
Oncogene ; 27(10): 1439-48, 2008 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-17828307

RESUMO

Signals from fibronectin-binding integrins promote neural crest cell motility during development in part through protein-tyrosine kinase (PTK) activation. Neuroblastoma (NB) is a neural crest malignancy with high metastatic potential. We find that alpha4 and alpha5 integrins are present in late-stage NB tumors and cell lines derived thereof. To determine the signaling connections promoting either alpha4beta1- or alpha5beta1-initiated NB cell motility, pharmacological, dominant negative and short-hairpin RNA (shRNA) inhibitory approaches were undertaken. shRNA knockdown revealed that alpha5beta1-stimulated NB motility is dependent upon focal adhesion kinase (FAK) PTK, Src PTK and p130Cas adapter protein expression. Cell reconstitution showed that FAK catalytic activity is required for alpha5beta1-stimulated Src activation in part through direct FAK phosphorylation of Src at Tyr-418. Alternatively, alpha4beta1-stimulated NB cell motility is dependent upon Src and p130Cas but FAK is not essential. Catalytically inactive receptor protein-tyrosine phosphatase-alpha overexpression inhibited alpha4beta1-stimulated NB motility and Src activation consistent with alpha4-regulated Src activity occurring through Src Tyr-529 dephosphorylation. In alpha4 shRNA-expressing NB cells, alpha4beta1-stimulated Src activation and NB cell motility were rescued by wild type but not cytoplasmic domain-truncated alpha4 re-expression. These studies, supported by results using reconstituted fibroblasts, reveal that alpha4beta1-mediated Src activation is mechanistically distinct from FAK-mediated Src activation during alpha5beta1-mediated NB migration and support the evaluation of inhibitors to alpha4, Src and FAK in the control of NB tumor progression.


Assuntos
Movimento Celular/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Integrina alfa4beta1/fisiologia , Integrina alfa5beta1/fisiologia , Neuroblastoma/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Ativação Enzimática/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Humanos , Integrina alfa4beta1/biossíntese , Integrina alfa4beta1/genética , Integrina alfa5beta1/biossíntese , Integrina alfa5beta1/genética , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Estrutura Terciária de Proteína/fisiologia , Células Tumorais Cultivadas
6.
Oncogene ; 25(44): 5969-84, 2006 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-16682956

RESUMO

Elevated focal adhesion kinase (FAK) expression occurs in advanced cancers, yet a signaling role for FAK in tumor progression remains undefined. Here, we suppressed FAK activity in 4T1 breast carcinoma cells resulting in reduced FAK Y925 phosphorylation, Grb2 adaptor protein binding to FAK, and signaling to mitogen-activated protein (MAP) kinase (MAPK). Loss of a FAK-Grb2-MAPK linkage did not affect 4T1 cell proliferation or survival in culture, yet FAK inhibition reduced vascular endothelial growth factor (VEGF) expression and resulted in small avascular tumors in mice. This FAK-Grb2-MAPK linkage was essential in promoting angiogenesis as reconstitution experiments using Src-transformed FAK-null fibroblasts revealed that point mutations affecting FAK catalytic activity (R454) or Y925 phosphorylation (F925) disrupted the ability of FAK to promote MAPK- and VEGF-associated tumor growth. Notably, in both FAK-inhibited 4T1 and Src-transformed FAK-null cells, constitutively activated (CA) mitogen-activated protein kinase kinase 1 (MEK1) restored VEGF production and CA-MEK1 or added VEGF rescued tumor growth and angiogenesis. These studies provide the first biological support for Y925 FAK phosphorylation and define a novel role for FAK activity in promoting a MAPK-associated angiogenic switch during tumor progression.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Neoplasias Mamárias Animais/enzimologia , Neovascularização Patológica/enzimologia , Tirosina/metabolismo , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/enzimologia , Carcinoma/irrigação sanguínea , Carcinoma/enzimologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Células Cultivadas , Células Clonais , Feminino , Proteína-Tirosina Quinases de Adesão Focal/deficiência , Proteína-Tirosina Quinases de Adesão Focal/genética , Humanos , Neoplasias Mamárias Animais/irrigação sanguínea , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação , Proteínas Tirosina Quinases/fisiologia , Transdução de Sinais/genética
7.
Oncogene ; 25(32): 4429-40, 2006 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-16547501

RESUMO

Expression of focal adhesion kinase (FAK) is elevated in malignant breast cancer, yet the role of intrinsic FAK activity in promoting tumor progression remains undefined. Here, we have inhibited FAK activity or expression in murine 4T1 breast carcinoma cells via dominant-negative focal adhesion kinase-related non-kinase (FRNK) or anti-FAK short hairpin RNA (shRNA) expression, respectively. Neither FRNK nor FAK shRNA ( approximately 80% reduced FAK levels) affected 4T1 proliferation in culture, whereas reduced FAK activity or expression blocked 4T1 cell invasion through Matrigel and resulted in 2-3-fold lower urokinase plasminogen activator (uPA) expression. Control 4T1 cells implanted into mammary fat pads of BALB/c mice exhibited spontaneous metastasis to the lungs, to the peritoneal cavity, and resulted in 90% lethality within 21 days. Whereas FAK shRNA-expressing 4T1 cells formed tumors in mice with low levels of apoptosis, when mammary-injected, these cells did not exhibit lung metastasis after 21 days and caused only 40% lethality up to 60 days. Transient re-expression of wild-type but not kinase-dead FAK in 4T1 FAK shRNA cells promoted uPA production and mammary to lung metastasis within 7 days. In fact, stable human uPA overexpression in 4T1 FAK shRNA cells promoted Matrigel invasion and lung metastasis equal to 4T1 controls. Conversely, treatment with plasminogen activator inhibitor-1 or neutralizing antibody to uPA blocked Matrigel invasion of 4T1 control cells. These studies provide the first direct proof that FAK catalytic activity can facilitate metastatic breast cancer progression by regulating uPA expression.


Assuntos
Neoplasias da Mama/enzimologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/secundário , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Ativador de Plasminogênio Tipo Uroquinase/genética
8.
Cancer Res ; 61(19): 7079-90, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11585739

RESUMO

Elevated focal adhesion kinase (FAK) expression in human tumor cells has been correlated with an increased cell invasion potential. In cell culture, studies with FAK-null fibroblasts have shown that FAK function is required for cell migration. To determine the role of elevated FAK expression in facilitating epidermal growth factor (EGF)-stimulated human adenocarcinoma (A549) cell motility, antisense oligonucleotides were used to reduce FAK protein expression >75%. Treatment of A549 cells with FAK antisense (ISIS 15421) but not a mismatched control (ISIS 17636) oligonucleotide resulted in reduced EGF-stimulated p130(Cas)-Src complex formation, c-Jun NH(2)-terminal kinase (JNK) activation, directed cell motility, and serum-stimulated cell invasion through Matrigel. Because residual FAK protein in ISIS 15421-treated A549 cells was highly phosphorylated at the Tyr-397/Src homology (SH)2 binding site, expression of the FAK COOH-terminal domain (FRNK) was also used as an inhibitor of FAK function. Adenoviral-mediated infection and expression of FRNK promoted FAK dephosphorylation at Tyr-397, resulted in reduced EGF-stimulated JNK as well as extracellular-regulated kinase 2 (ERK2) kinase activation, inhibited matrix metalloproteinase-9 (MMP-9) secretion, and potently blocked both random and EGF-stimulated A549 cell motility. Equivalent expression of a FRNK (S-1034) point-mutant that did not promote FAK dephosphorylation also did not affect EGF-stimulated signaling or cell motility. Dose-dependent reduction in EGF-stimulated A549 motility was observed with the PD98059 MEK1 inhibitor and the batimastat (BB-94) inhibitor of MMP activity, but not with the SB203580 inhibitor of p38 kinase. Finally, comparisons between normal, FAK-null, and FAK-reconstituted fibroblasts revealed that FAK enhanced EGF-stimulated JNK and ERK2 kinase activation that was required for cell motility. These data indicate that FAK functions as an important signaling platform to coordinate EGF-stimulated cell migration in human tumor cells and support a role for inhibitors of FAK expression or activity in the control of neoplastic cell invasion.


Assuntos
Adenocarcinoma/enzimologia , Movimento Celular/fisiologia , Fator de Crescimento Epidérmico/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Adenocarcinoma/patologia , Movimento Celular/efeitos dos fármacos , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , Fosforilação , Proteínas Tirosina Quinases/biossíntese , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/fisiologia , Células Tumorais Cultivadas
9.
J Biol Chem ; 276(44): 40903-9, 2001 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-11533025

RESUMO

alpha4 integrins are essential for embryogenesis, hematopoiesis, inflammation, and immune response possibly because alpha4 integrins have distinct signaling properties from other integrins. Specifically, the alpha4 cytoplasmic domain binds tightly to paxillin, a signaling adaptor protein, leading to increased cell migration and an altered cytoskeletal organization that results in reduced cell spreading. The alpha4 tail contains potential phosphorylation sites clustered in its core paxillin binding region. We now report that the alpha4 tail is phosphorylated in vitro and in vivo. Furthermore, Ser(988) is a major phosphorylation site. Using antibodies specific for Ser(988)-phosphorylated alpha4, we found the stoichiometry of alpha4 phosphorylation varied in different cells. However, >60% of alpha4 was phosphorylated in Jurkat T cells. Phosphorylation at Ser(988) blocked paxillin binding to the alpha4 tail. A phosphorylation-mimicking mutant of alpha4 (alpha4S988D) blocked paxillin binding and reversed the inhibitory effect of alpha4 on cell spreading. Consequently, alpha4 phosphorylation is a biochemical mechanism to modulate paxillin binding to alpha4 integrins with consequent regulation of alpha4 integrin-dependent cellular functions.


Assuntos
Antígenos CD/metabolismo , Citoplasma/metabolismo , Proteínas do Citoesqueleto/metabolismo , Fosfoproteínas/metabolismo , Sequência de Aminoácidos , Antígenos CD/química , Antígenos CD/genética , Western Blotting , Humanos , Integrina alfa4 , Células Jurkat , Dados de Sequência Molecular , Paxilina , Mapeamento de Peptídeos , Fosforilação , Testes de Precipitina , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina/metabolismo
10.
J Biol Chem ; 276(31): 28676-85, 2001 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-11369769

RESUMO

alpha-Actinin is tyrosine-phosphorylated in activated human platelets (Izaguirre, G., Aguirre, L., Ji, P., Aneskievich, B., and Haimovich, B. (1999) J. Biol. Chem. 274, 37012--37020). Analysis of platelet RNA by reverse transcription-polymerase chain reaction revealed that alpha-actinin expressed in platelets is identical to the cytoskeletal/non-muscle isoform. A construct of this isoform containing a His(6) tag at the amino terminus was generated. Robust tyrosine phosphorylation of the recombinant protein was detected in cells treated with the tyrosine phosphatase inhibitor vanadate. The tyrosine phosphorylation site was localized to the amino-terminal domain by proteolytic digestion. A recombinant alpha-actinin protein containing a Tyr --> Phe mutation at position 12 (Y12F) was no longer phosphorylated when expressed in vanadate-treated cells, indicating that tyrosine 12 is the site of phosphorylation. The wild type recombinant protein was not phosphorylated in cells lacking the focal adhesion kinase (FAK). Re-expression of FAK in these cells restored alpha-actinin phosphorylation. Purified wild type alpha-actinin, but not the Y12F mutant, was phosphorylated in vitro by wild type as well as a Phe-397 mutant of FAK. In contrast, no phosphorylation was detected in the presence of a kinase-dead FAK. Tyrosine phosphorylation reduced the amount of alpha-actinin that cosedimented with actin filaments. These results establish that alpha-actinin is a direct substrate for FAK and suggest that alpha-actinin mediates FAK-dependent signals that could impact the physical properties of the cytoskeleton.


Assuntos
Actinina/metabolismo , Actinas/metabolismo , Citoesqueleto/metabolismo , Proteínas Tirosina Quinases/metabolismo , Actinina/sangue , Actinina/química , Actinas/química , Substituição de Aminoácidos , Sítios de Ligação , Plaquetas/metabolismo , Clonagem Molecular , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Cinética , Mutagênese Sítio-Dirigida , Fenilalanina , Fosforilação , Isoformas de Proteínas/metabolismo , RNA/sangue , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tirosina
11.
J Biol Chem ; 276(21): 17653-62, 2001 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-11278488

RESUMO

Integrins facilitate cell attachment to the extracellular matrix, and these interactions generate cell survival, proliferation, and motility signals. Integrin signals are relayed in part by focal adhesion kinase (FAK) activation and the formation of a transient signaling complex initiated by Src homology 2 (SH2)-dependent binding of Src family protein-tyrosine kinases to the FAK Tyr-397 autophosphorylation site. Here we show that in viral Src (v-Src)-transformed NIH3T3 fibroblasts, an adhesion-independent FAK-Src signaling complex occurs. Co-expression studies in human 293T cells showed that v-Src could associate with and phosphorylate a Phe-397 FAK mutant at Tyr-925 promoting Grb2 binding to FAK in suspended cells. In vitro, glutathione S-transferase fusion proteins of the v-Src SH3 but not c-Src SH3 domain bound to FAK in lysates of NIH3T3 fibroblasts. The v-Src SH3-binding sites were mapped to known proline-X-X-proline (PXXP) SH3-binding motifs in the FAK N- (residues 371-377) and C-terminal domains (residues 712-718 and 871-882) by in vitro pull-down assays, and these sites are composed of a PXXPXXPhi (where Phi is a hydrophobic residue) v-Src SH3 binding consensus. Sequence comparisons show that residues in the RT loop region of the c-Src and v-Src SH3 domains differ. Substitution of c-Src RT loop residues (Arg-97 and Thr-98) for those found in the v-Src SH3 domain (Trp-97 and Ile-98) enhanced the binding of distinct NIH3T3 cellular proteins to a glutathione S-transferase fusion protein of the c-Src (Trp-97 + Ile-98) SH3 domain. FAK was identified as a c-Src (Trp-97 + Ile-98) SH3 domain target in fibroblasts, and co-expression studies in 293T cells showed that full-length c-Src (Trp-97 + Ile-98) could associate in vivo with Phe-397 FAK in an SH2-independent manner. These studies establish a functional role for the v-Src SH3 domain in stabilizing an adhesion-independent signaling complex with FAK.


Assuntos
Transformação Celular Viral , Genes src/fisiologia , Proteínas Tirosina Quinases/fisiologia , Células 3T3 , Animais , Adesão Celular/fisiologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Camundongos , Domínios de Homologia de src/fisiologia
12.
J Cell Biol ; 152(1): 97-110, 2001 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-11149924

RESUMO

Focal adhesion kinase-null (FAK(-/-) fibroblasts exhibit morphological and motility defects that are reversed by focal adhesion kinase (FAK) reexpression. The FAK-related kinase, proline-rich tyrosine kinase 2 (Pyk2), is expressed in FAK(-/-) cells, yet it exhibits a perinuclear distribution and does not functionally substitute for FAK. Chimeric Pyk2/FAK proteins were created and expressed in FAK(-/-) cells to determine the impact of Pyk2 localization to focal contacts. Whereas an FAK/Pyk2 COOH-terminal (CT) domain chimera was perinuclear distributed, stable expression of a Pyk2 chimera with the FAK-CT domain (Pyk2/FAK-CT) localized to focal contact sites and enhanced fibronectin (FN)-stimulated haptotactic cell migration equal to FAK-reconstituted cells. Disruption of paxillin binding to the FAK-CT domain (S-1034) inhibited Pyk2/FAK-CT localization to focal contacts and its capacity to promote cell motility. Paxillin binding to the FAK-CT was necessary but not sufficient to mediate the indirect association of FAK or Pyk2/FAK-CT with a beta 1-integrin-containing complex. Both FAK and Pyk2/FAK-CT but not Pyk2/FAK-CT S-1034 reconstituted FAK(-/-) cells, exhibit elevated FN-stimulated extracellular signal-regulated kinase 2 (ERK2) and c-Jun NH(2)-terminal kinase (JNK) kinase activation. FN-stimulated FAK or Pyk2/FAK-CT activation enhanced both the extent and duration of FN-stimulated ERK2 activity which was necessary for cell motility. Transient overexpression of the FAK-CT but not FAK-CT S-1034 domain inhibited both FN-stimulated ERK2 and JNK activation as well as FN-stimulated motility of Pyk2/FAK-CT reconstituted cells. These gain-of-function studies show that the NH(2)-terminal and kinase domains of Pyk2 can functionally substitute for FAK in promoting FN-stimulated signaling and motility events when localized to beta-integrin-containing focal contact sites via interactions mediated by the FAK-CT domain.


Assuntos
Movimento Celular/fisiologia , Fibronectinas/metabolismo , Integrina beta1/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Quinase 1 de Adesão Focal , Quinase 2 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Expressão Gênica , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Paxilina , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
13.
Infect Immun ; 68(11): 6423-30, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11035755

RESUMO

Escherichia coli K1 traversal across the blood-brain barrier is an essential step in the pathogenesis of neonatal meningitis. We have previously shown that invasive E. coli promotes the actin rearrangement of brain microvascular endothelial cells (BMEC), which constitute a lining of the blood-brain barrier, for invasion. However, signal transduction mechanisms involved in E. coli invasion are not defined. In this report we show that tyrosine kinases play a major role in E. coli invasion of human BMEC (HBMEC). E. coli induced tyrosine phosphorylation of HBMEC cytoskeletal proteins, focal adhesion kinase (FAK), and paxillin, with a concomitant increase in the association of paxillin with FAK. Overexpression of a dominant interfering form of the FAK C-terminal domain, FRNK (FAK-related nonkinase), significantly inhibited E. coli invasion of HBMEC. Furthermore, we found that FAK kinase activity and the autophosphorylation site (Tyr397) are important in E. coli invasion of HBMEC, whereas the Grb2 binding site (Tyr925) is not required. Immunocytochemical studies demonstrated that FAK is recruited to focal plaques at the site of bacterial entry. Consistent with the invasion results, overexpression of FRNK, a kinase-negative mutant (Arg454 FAK), and a Src binding mutant (Phe397 FAK) inhibited the accumulation of FAK at the bacterial entry site. The overexpression of FAK mutants in HBMEC also blocked the E. coli-induced tyrosine phosphorylation of FAK and its association with paxillin. These observations provide evidence that FAK tyrosine phosphorylation and its recruitment to the cytoskeleton play a key role in E. coli invasion of HBMEC.


Assuntos
Encéfalo/microbiologia , Endotélio Vascular/microbiologia , Escherichia coli/patogenicidade , Proteínas Tirosina Quinases/fisiologia , Encéfalo/irrigação sanguínea , Células Cultivadas , Ativação Enzimática , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Genisteína/farmacologia , Humanos , Fosforilação , Tirosina/metabolismo
14.
J Cell Sci ; 113 ( Pt 20): 3673-8, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11017882

RESUMO

Focal adhesion kinase (FAK) is activated and localized at focal adhesions upon cell adhesion to extracellular matrices. Cells lacking FAK show increased focal adhesion number and decreased cell migration, functions that are regulated by the small GTPase Rho. We now report that fibroblasts from FAK-/- mice failed to transiently inhibit Rho activity when plated on fibronectin. Re-expression of FAK restored normal Rho regulation. Turnover of focal adhesions correlated inversely with Rho activity. The presence or absence of FAK was mimicked by inhibiting or activating Rho, respectively. These data suggest that loss of FAK resulting in constitutive activation of Rho and inhibition of focal adhesion turnover can account for deficiencies in cell migration and embryonic lethality of the FAK knockout.


Assuntos
Adesões Focais , Proteínas Tirosina Quinases/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Movimento Celular , Tamanho Celular , Células Cultivadas , Citoesqueleto/ultraestrutura , Ativação Enzimática , Fibroblastos , Fibronectinas , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Camundongos , Proteínas rho de Ligação ao GTP/antagonistas & inibidores
15.
J Biol Chem ; 275(52): 41092-9, 2000 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-10998418

RESUMO

The focal adhesion (FAK) non-receptor protein-tyrosine kinase (PTK) links both extracellular matrix/integrin and growth factor stimulation to intracellular signals promoting cell migration. Here we show that both transient and stable overexpression of the FAK C-terminal domain termed FRNK (FAK-related non-kinase) inhibits serum and platelet-derived growth factor (PDGF)-BB-induced vascular smooth muscle cell (SMC) migration in wound healing and in vitro Boyden Chamber chemotaxis assays, respectively. Expression of FRNK, but not a point mutant of FRNK (FRNK L1034S), disrupted the formation of a complex containing both FAK and the activated PDGF-beta receptor and resulted in reduced tyrosine phosphorylation of endogenous FAK at the Tyr-397 binding site for Src family PTKs. As demonstrated using FAK-deficient and FAK-reconstituted fibroblasts, FAK positively contributed to PDGF-BB-stimulated ERK2/MAP kinase activity, and in SMCs, ERK2/MAP kinase activity was required for PDGF-BB-stimulated chemotaxis. Stable expression of FRNK but not FRNK L1034S expression in SMCs lowered the extent and duration of stimulated ERK2/MAP kinase activation at low but not at high PDGF-BB concentrations. Importantly, stable expression of FRNK in SMCs did not affect SMC morphology or proliferation in culture. Because the increased migration of vascular SMCs in response to extracellular matrix proteins and growth factors contributes to neointima formation, our results show that FAK inhibition by FRNK expression may provide a novel approach to regulate abnormal vascular SMC migration in vivo.


Assuntos
Quimiotaxia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Tirosina Quinases/fisiologia , Animais , Becaplermina , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular , Ativação Enzimática , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Fosforilação , Proteínas Proto-Oncogênicas c-sis , Ratos , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Tirosina/metabolismo , Cicatrização
16.
J Biol Chem ; 275(49): 38371-7, 2000 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-10967115

RESUMO

Integrins are transmembrane receptors involved in interactions between cells and extracellular matrix proteins. Here we show that cell adhesion regulates insulin receptor substrate-1 (IRS-1) mRNA synthesis. When fibroblasts are held in suspension, lower levels of IRS-1 mRNA, but not of IRS-2 mRNA, are detected, and this effect is due to the negative regulation of IRS-1 transcription rather than to decreased mRNA stability. Upon fibronectin- or vitronectin-mediated integrin stimulation, the level of IRS-1 mRNA was restored within 4 h. The focal adhesion kinase (FAK) is known to be activated upon integrin stimulation, and we found that IRS-1 was not expressed in FAK(-)(/-) cells. Stable re-expression of epitope-tagged FAK in FAK(-)(/-) fibroblasts (DA2 cells) restored normal levels of IRS-1 expression, confirming that IRS-1 mRNA expression is regulated by FAK. It is known that integrins activate the JNK pathway. However, in adherent FAK(-)(/-) cells, we failed to detect activation of JNK, whereas JNK was stimulated in DA2 cells. This confirms the role of FAK in integrin-induced JNK stimulation. FAK-independent stimulation of JNK with anisomycin treatment both in FAK(-)(/-) cells and in suspended FAK(+/+) cells confirmed that IRS-1 mRNA transcription can be partially regulated by JNK. We suggest that integrins can modulate insulin and insulin-like growth factor-1 signaling pathways by regulating the levels of IRS-1 in cells and that FAK-mediated signaling to JNK is one pathway involved in this process.


Assuntos
Adesão Celular/fisiologia , Regulação da Expressão Gênica , Fosfoproteínas/genética , Proteínas Tirosina Quinases/metabolismo , Transcrição Gênica , Animais , Células Cultivadas , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/fisiologia , Fibronectinas/fisiologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Proteínas Substratos do Receptor de Insulina , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , RNA Mensageiro/genética , Receptor de Insulina/fisiologia , Proteínas Recombinantes/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Vitronectina/fisiologia
17.
Immunol Res ; 21(2-3): 293-303, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10852130

RESUMO

In performing host-defense functions, cells of the immune system become activated by soluble chemokine signals and must migrate through endothelial cell or solid tissue barriers to reach sites of inflammation or infection. Regulated adhesive interactions of immune cells with endothelium, extracellular matrix components, and cells of solid organs are critical control points of the overall immune response. Both the soluble chemokine and cell adhesion receptor-mediated migration signals must converge on common intracellular targets to engage the cell migration machinery. In this article, we focus on the role of focal adhesion kinase (FAK) and its homolog Pyk2 as cytoplasmic mediators of motility events in multiple cell types. We introduce the overall domain structure of the FAK and Pyk2 nonreceptor protein tyrosine kinases (PTKs), highlight some of the signals that activate these PTKs, and detail the molecules that functionally interact and signal transduction pathways that may mediate cell migration responses. Emphasis is placed on the knowledge gained from studies using FAK-null cells as a model system to decipher the role of this PTK in promoting cell motility.


Assuntos
Movimento Celular/fisiologia , Proteínas Tirosina Quinases/fisiologia , Animais , Quinase 1 de Adesão Focal , Quinase 2 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Transdução de Sinais/fisiologia
18.
J Cell Biol ; 149(3): 741-54, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10791986

RESUMO

Most transformed cells have lost anchorage and serum dependence for growth and survival. Previously, we established that when serum is absent, fibronectin survival signals transduced by focal adhesion kinase (FAK), suppress p53-regulated apoptosis in primary fibroblasts and endothelial cells (Ilic et al. 1998. J. Cell Biol. 143:547-560). The present goals are to identify survival sequences in FAK and signaling molecules downstream of FAK required for anchorage-dependent survival of primary fibroblasts. We report that binding of the SH3 domain of p130Cas to proline-rich region 1 of FAK is required to support survival of fibroblasts on fibronectin when serum is withdrawn. The FAK-p130Cas complex activates c-Jun NH2-terminal kinase (JNK) via a Ras/Rac1/Pak1/MAPK kinase 4 (MKK4) pathway. Activated (phospho-) JNK colocalizes with FAK in focal adhesions of fibroblasts cultured on fibronectin, which supports their survival, but not in fibroblasts cultured on collagen, which does not. Cells often survive in the absence of extracellular matrix if serum factors are provided. In that case, we confirm work of others that survival signals are transduced by FAK, phosphatidylinositol 3'-kinase (PI3-kinase), and Akt/protein kinase B (PKB). However, when serum is absent, PI3-kinase and Akt/PKB are not involved in the fibronectin-FAK-JNK survival pathway documented herein. Thus, survival signals from extracellular matrix and serum are transduced by FAK via two distinct pathways.


Assuntos
Adesão Celular , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases/metabolismo , Proteínas , Animais , Sobrevivência Celular , Meios de Cultura Livres de Soro , Fibroblastos , Proteína-Tirosina Quinases de Adesão Focal , Imuno-Histoquímica , Proteínas Quinases JNK Ativadas por Mitógeno , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Coelhos , Proteína p130 Retinoblastoma-Like , Transdução de Sinais , Transfecção , Domínios de Homologia de src
19.
Nat Cell Biol ; 2(5): 249-56, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10806474

RESUMO

Here we show that cells lacking focal adhesion kinase (FAK) are refractory to motility signals from platelet-derived and epidermal growth factors (PDGF and EGF respectively), and that stable re-expression of FAK rescues these defects. FAK associates with activated PDGF- and EGF-receptor (PDGFR and EGFR) signalling complexes, and expression of the band-4.1-like domain at the FAK amino terminus is sufficient to mediate an interaction with activated EGFR. However, efficient EGF-stimulated cell migration also requires FAK to be targeted, by its carboxy-terminal domain, to sites of integrin-receptor clustering. Although the kinase activity of FAK is not needed to promote PDGF- or EGF-stimulated cell motility, kinase-inactive FAK is transphosphorylated at the indispensable Src-kinase-binding site, FAK Y397, after EGF stimulation of cells. Our results establish that FAK is an important receptor-proximal link between growth-factor-receptor and integrin signalling pathways.


Assuntos
Movimento Celular/fisiologia , Integrinas/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Tirosina Quinases/metabolismo , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/fisiologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Quinase 1 de Adesão Focal , Quinase 2 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mutagênese/fisiologia , Estrutura Terciária de Proteína , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/genética , Receptores do Fator de Crescimento Derivado de Plaquetas/fisiologia , Quinases da Família src/metabolismo
20.
J Biol Chem ; 275(19): 14729-35, 2000 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-10799562

RESUMO

The Crk-associated substrate, p130(CAS), has been implicated in the regulation of the actin cytoskeleton following ligation of cell integrins with the extracellular matrix. Integrin-mediated cell adhesion involves p130(CAS) association with focal adhesion kinase (p125(FAK)). Internalization/cell entry of type 2 and type 5 adenoviruses (Ad) is also mediated by alpha(v) integrins. However, expression of dominant negative forms of p125(FAK) does not alter virus entry, and Ad entry occurs normally in p125(FAK)-deficient fibroblasts. We now provide evidence that Ad internalization, a process which is mediated by alpha(v) integrins, also requires p130(CAS) and phosphatidylinositol-3-OH kinase (PI 3-kinase). Ad induces p130(CAS) phosphorylation and inhibition of p130(CAS) phosphorylation by tyrphostin and genistein, or expression of the substrate domain deleted p130(CAS) blocks Ad internalization. p130(CAS) was also found to associate with the p85 subunit of PI 3-kinase through its proline-rich domain during virus internalization and expression of p130(CAS) containing a deleted proline-rich domain (PRD) inhibited adenovirus cell entry. We showed further that the RPLPSPP motif in the proline-rich region of p130(CAS) interacts with the SH3 domain of p85/PI 3-kinase. These studies reveal the molecular basis by which p130(CAS) coordinates the signaling pathways involved in integrin-mediated Ad endocytosis.


Assuntos
Adenoviridae/fisiologia , Fusão de Membrana , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Proteínas , Receptores de Vitronectina , Motivos de Aminoácidos , Sequência de Aminoácidos , Linhagem Celular , Proteína Substrato Associada a Crk , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Integrinas/fisiologia , Fosfatidilinositol 3-Quinases/química , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Proteína p130 Retinoblastoma-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA