Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 14(5)2022 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-35267539

RESUMO

Cancer-associated fibroblasts (CAFs) are a heterogenous population of stromal cells found in solid malignancies that coexist with the growing tumor mass and other immune/nonimmune cellular elements. In certain neoplasms (e.g., desmoplastic tumors), CAFs are the prominent mesenchymal cell type in the tumor microenvironment, where their presence and abundance signal a poor prognosis in multiple cancers. CAFs play a major role in the progression of various malignancies by remodeling the supporting stromal matrix into a dense, fibrotic structure while secreting factors that lead to the acquisition of cancer stem-like characteristics and promoting tumor cell survival, reduced sensitivity to chemotherapeutics, aggressive growth and metastasis. Tumors with high stromal fibrotic signatures are more likely to be associated with drug resistance and eventual relapse. Clarifying the molecular basis for such multidirectional crosstalk among the various normal and neoplastic cell types present in the tumor microenvironment may yield novel targets and new opportunities for therapeutic intervention. This review highlights the most recent concepts regarding the complexity of CAF biology including CAF heterogeneity, functionality in drug resistance, contribution to a progressively fibrotic tumor stroma, the involved signaling pathways and the participating genes.

2.
Adv Wound Care (New Rochelle) ; 3(3): 281-290, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24669362

RESUMO

Significance: A highly interactive serine protease/plasmin/matrix metalloproteinase axis regulates stromal remodeling in the wound microenvironment. Current findings highlight the importance of stringent controls on protease expression and their topographic activities in cell proliferation, migration, and tissue homeostasis. Targeting elements in this cascading network may lead to novel therapeutic approaches for fibrotic diseases and chronic wounds. Recent Advances: Matrix-active proteases and their inhibitors orchestrate wound site tissue remodeling, cell migration, and proliferation. Indeed, the serine proteases urokinase plasminogen activator and tissue-type plasminogen activator (uPA/tPA) and their major phsyiological inhibitor, plasminogen activator inhibitor-1 (PAI-1; serine protease inhibitor clade E member 1 [SERPINE1]), are upregulated in several cell types during injury repair. Coordinate expression of proteolytic enzymes and their inhibitors in the wound bed provides a mechanism for fine control of focal proteolysis to facilitate matrix restructuring and cell motility in complex environments. Critical Issues: Cosmetic and tissue functional consequences of wound repair anomalies affect the quality of life of millions of patients in the United States alone. The development of novel therapeutics to manage individuals most affected by healing anomalies will likely derive from the identification of critical, translationally accessible, control elements in the wound site microenvironment. Future Directions: Activation of the PAI-1 gene early after wounding, its prominence in the repair transcriptome and varied functions suggest a key role in the global cutaneous injury response program. Targeting PAI-1 gene expression and/or PAI-1 function with molecular genetic constructs, neutralizing antibodies or small molecule inhibitors may provide a novel, therapeutically relevant approach, to manage the pathophysiology of wound healing disorders associated with deficient or excessive PAI-1 levels.

3.
Mol Med Ther ; 1(2): 106, 2012 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-23847772

RESUMO

Accumulation of neurotoxic amyloid peptides (Aß) in the brain, generated by ß-site proteolytic processing of the amyloid precursor protein (APP), is the hallmark pathophysiologic feature of Alzheimer's disease. The plasmin-activating cascade, in which urokinase (uPA) and tissue-type (tPA) plasminogen activators convert plasminogen to the broad-spectrum protease plasmin, appears to serve a protective, Aß-clearing, role in the central nervous system. Plasmin degrades Aß and catalyzes α- site APP proteolysis generating nontoxic peptides. Plasmin activation in the brain is negatively regulated by the fast-acting clade E serine protease inhibitor (SERPIN) plasminogen activator inhibitor type-1 (PAI-1; SERPINE1) resulting in Aß accumulation. PAI-1 and its major physiological inducer TGF-ß1, moreover, are both increased in Alzheimer's disease models and implicated in the etiology and progression of human neurodegenerative disorders. Current findings support the hypothesis that targeting of PAI-1 function (by small molecule drugs) and/or gene expression (by histone deacetylase inhibitors) may constitute a clinically-relevant molecular approach to the therapy of neurodegenerative diseases associated with increased PAI-1 levels.

4.
Exp Cell Res ; 312(7): 1093-105, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16457817

RESUMO

Transforming growth factor-beta1 (TGF-beta1) transcriptionally regulates the expression of genes that encode specific proteins (e.g., plasminogen activator inhibitor-1; PAI-1) important in stromal remodeling and cellular invasion. Definition of molecular events underlying TGF-beta1-initiated PAI-1 transcription, therefore, may lead to the identification of new therapeutic targets for diseases associated with elevated PAI-1 synthesis (e.g., tissue fibrosis, vascular disorders, tumor progression). An intact upstream stimulatory factor (USF)-binding E box motif (5'-(-165)CACGTG(-160)-3') at the HRE-2 site in the rat PAI-1 gene was required for PAI-1 transcription in TGF-beta1-treated cells. Mutation of the CA dinucleotide to TC at position -165/-164 in a reporter construct driven by 764 bp of PAI-1 promoter sequence decreased TGF-beta1-dependent CAT activity by >80% indicating the necessity for a consensus hexanucleotide E box motif in induced expression. The same CA --> TC substitution eliminated USF binding to an 18-bp HRE-2 DNA target highlighting the importance of site occupancy to transcriptional activation. Transfection of a dominant-negative USF construct, moreover, completely inhibited formation of USF/HRE-2 probe complexes, attenuated PAI-1 promoter-driven luciferase activity and reduced the response of the endogenous PAI-1 gene to TGF-beta1 (to that approximating quiescent controls). Maximal immediate-early PAI-1 induction upon exposure to TGF-beta1 required EGFR, p21ras, MEK and pp60(c-src) signaling as pharmacologic or dominant-negative inhibition of any of the four intermediates (EGFR, p21ras, MEK, pp60(c-src)) virtually eliminated TGF-beta1-augmented PAI-1 levels. U0126 titering experiments, furthermore, revealed that the same MEK inhibitor concentration that blocked the TGF-beta1 increase in ERK1/2 phosphorylation (20 microM) also effectively attenuated the PAI-1 inductive response suggesting a requirement for stimulated ERK signaling in TGF-beta1-mediated PAI-1 expression. These data suggest a model whereby TGF-beta1 activates a complex signaling cascade to affect PAI-1 gene control and involves USF occupancy of a critical E box motif at the HRE-2 site in the PAI-1 gene.


Assuntos
Elementos E-Box/fisiologia , Receptores ErbB/fisiologia , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Humanos , MAP Quinase Quinase Quinases/fisiologia , Mutagênese Sítio-Dirigida , Inibidor 1 de Ativador de Plasminogênio/genética , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Quinazolinas , Ratos , Fator de Crescimento Transformador beta1 , Tirfostinas/farmacologia
5.
J Cell Physiol ; 204(1): 236-46, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15622520

RESUMO

Transforming growth factor-beta1 (TGF-beta1) stimulates expression of plasminogen activator inhibitor type-1 (PAI-1), a serine protease inhibitor (SERPIN) important in the control of stromal barrier proteolysis and cell-to-matrix adhesion. Pharmacologic agents that target MEK (PD98059, U0126) or src family (PP1) kinases attenuated TGF-beta1-dependent PAI-1 transcription in R22 aortic smooth muscle cells. Pretreatment with PP1 at concentrations that inhibited TGF-beta1-dependent PAI-1 expression also blocked ERK1/2 activation/nuclear accumulation suggesting that the required src kinase activity is upstream of ERK1/2 in the TGF-beta1-initiated signaling cascade. The IC(50) of the PP1-sensitive kinase, furthermore, specifically implied involvement of pp60(c-src) in PAI-1 induction. Indeed, addition of TGF-beta1 to quiescent R22 cells resulted in a 3-fold increase in pp60(c-src) autophosphorylation and kinase activity. Transfection of a dominant-negative pp60(c-src) construct, moreover, reduced TGF-beta1-induced PAI-1 expression levels to that of unstimulated controls or PP1-pretreated cells. A >/=170 kDa protein that co-immunoprecipitated with TGF-beta1-activated pp60(c-src) was also phosphorylated transiently in response to TGF-beta1. TGF-beta1 is known to transactivate the 170 kDa EGF receptor (EGFR) by autocrine HB-EGF or TGF-alpha mechanisms suggesting involvement of EGFR activation in certain TGF-beta1-initiated responses. Incubation of quiescent R22 cells with the EGFR-specific inhibitor AG1478 prior to growth factor (EGF or TGF-beta1) addition effectively blocked EGFR activation as determined by direct visualization of receptor internalization. AG1478 suppressed (in a dose-dependent fashion) EGF-induced PAI-1 protein levels and, at a final concentration of 2.5 muM, virtually eliminated EGF-dependent PAI-1 synthesis. More importantly, AG1478 similarly repressed inducible PAI-1 levels in TGF-beta1-stimulated R22 cultures. PP1, PD98059, and U0126 also inhibited TGF-beta1-dependent cell motility at concentrations that significantly attenuated PAI-1 expression. Consistent with the AG1478-associated reductions in EGF- and TGF-beta1-stimulated PAI-1 expression, pretreatment of R22 cell cultures with AG1478 effectively suppressed growth factor-stimulated cell motility. These data indicate that two major phenotypic characteristics of TGF-beta1-exposure (i.e., transcription of specific target genes [e.g., PAI-1], increased cell motility) are linked in the R22 vascular smooth muscle cell system, require pp60(c-src) kinase activity and MEK signaling and involve activation of an AG1478-sensitive (likely EGFR-dependent) pathway.


Assuntos
Movimento Celular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Miócitos de Músculo Liso/fisiologia , Inibidor 1 de Ativador de Plasminogênio/genética , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Aorta/citologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Receptores ErbB/metabolismo , Expressão Gênica/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Proteínas Proto-Oncogênicas pp60(c-src)/antagonistas & inibidores , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Pirazóis/farmacologia , Pirimidinas/farmacologia , Quinazolinas , Fator de Crescimento Transformador beta1 , Tirfostinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...