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










Intervalo de ano de publicação
1.
Adv Cancer Res ; 132: 45-71, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27613129

RESUMO

The majority of cancer deaths are due to metastases that can occur years or decades after primary tumor diagnosis and treatment. Disseminated tumor cells (DTCs) surviving in a dormant state in target organs appear to explain the timing of this phenomenon. Knowledge on this process is important as it might provide a window of opportunity to prevent recurrences by eradicating dormant DTCs and/or by maintaining DTCs in a dormant state. Importantly, this research might offer markers of dormancy for early monitoring of metastatic relapse. However, our understanding of the mechanisms underlying the regulation of entry into and exit from dormancy is still limited and crippling any therapeutic opportunity. While cancer cell-intrinsic signaling pathways have been linked to dormancy regulation, it is likely that these pathways and the switch controlling reactivation from dormancy are regulated by microenvironmental cues. Here we review and discuss recent findings on how the microenvironment regulates cancer dormancy and raise new questions that may help advance the field.


Assuntos
Neoplasia Residual/patologia , Neoplasias/patologia , Microambiente Tumoral , Animais , Humanos
2.
Leukemia ; 29(12): 2307-16, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26104660

RESUMO

P38α/ß has been described as a tumor-suppressor controlling cell cycle checkpoints and senescence in epithelial malignancies. However, p38α/ß also regulates other cellular processes. Here, we describe a role of p38α/ß as a regulator of acute lymphoblastic leukemia (ALL) proliferation and survival in experimental ALL models. We also report first evidence that p38α/ß phosphorylation is associated with the occurrence of relapses in TEL-AML1-positive leukemia. First, in vitro experiments show that p38α/ß signaling is induced in a cyclical manner upon initiation of proliferation and remains activated during log-phase of cell growth. Next, we provide evidence that growth-permissive signals in the bone marrow activate p38α/ß in a novel avian ALL model, in which therapeutic targeting can be tested. We further demonstrate that p38α/ß inhibition by small molecules can suppress leukemic expansion and prolong survival of mice bearing ALL cell lines and primary cells. Knockdown of p38α strongly delays leukemogenesis in mice xenografted with cell lines. Finally, we show that in xenografted TEL-AML1 patients, ex vivo p38α/ß phosphorylation is associated with an inferior long-term relapse-free survival. We propose p38α/ß as a mediator of proliferation and survival in ALL and show first preclinical evidence for p38α/ß inhibition as an adjunct approach to conventional therapies.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Adolescente , Animais , Proliferação de Células , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Camundongos , Fosforilação , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidade , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia
3.
Oncogene ; 33(48): 5483-90, 2014 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-24413078

RESUMO

The postnatal mammary gland develops extensively through cycles of proliferation, branching, involution and remodeling. We review recent advances made in the field of stress signaling pathways and its roles in mammary gland organogenesis, how they contribute to normal organ specification and homeostasis and how its subversion by oncogenes leads to cancer. We analyze stress signaling in mammary gland biology taking into account the interrelationship with the extracellular matrix and adhesion signaling during morphogenesis. By integrating the information gathered from in vivo and three dimensional in vitro organogenesis studies, we review the novel contribution of p38(SAPK), c-Jun NH2-terminal kinase and PKR-like endoplasmic reticulum kinase (PERK) signaling pathways to the timely activation of cell death, correct establishment of polarity and growth arrest and autophagy, respectively. We also review the evidence supporting that the activation of the aforementioned stress kinases maintain breast acinar structures as part of a tumor suppressive program and that its deregulation is commonplace during breast cancer initiation.


Assuntos
Neoplasias da Mama/patologia , Glândulas Mamárias Humanas , Transdução de Sinais/fisiologia , Animais , Feminino , Humanos , Morfogênese
4.
Oncogene ; 32(41): 4932-40, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23160380

RESUMO

Adhesion to the extracellular matrix (ECM) is critical for epithelial tissue homeostasis and function. ECM detachment induces metabolic stress and programmed cell death via anoikis. ECM-detached mammary epithelial cells are able to rapidly activate autophagy allowing for survival and an opportunity for re-attachment. However, the mechanisms controlling detachment-induced autophagy remain unclear. Here we uncover that the kinase PERK rapidly promotes autophagy in ECM-detached cells by activating AMP-activated protein kinase (AMPK), resulting in downstream inhibition of mTORC1-p70(S6K) signaling. LKB1 and TSC2, but not TSC1, are required for PERK-mediated inhibition of mammalian target of rapamycinin MCF10A cells and mouse embryo fibroblast cells. Importantly, this pathway shows fast kinetics, is transcription-independent and is exclusively activated during ECM detachment, but not by canonical endoplasmic reticulum stressors. Moreover, enforced PERK or AMPK activation upregulates autophagy and causes luminal filling during acinar morphogenesis by perpetuating a population of surviving autophagic luminal cells that resist anoikis. Hence, we identify a novel pathway in which suspension-activated PERK promotes the activation of LKB1, AMPK and TSC2, leading to the rapid induction of detachment-induced autophagy. We propose that increased autophagy, secondary to persistent PERK and LKB1-AMPK signaling, can robustly protect cells from anoikis and promote luminal filling during early carcinoma progression.


Assuntos
Autofagia , Matriz Extracelular/metabolismo , Complexos Multiproteicos/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , eIF-2 Quinase/metabolismo , Proteínas Quinases Ativadas por AMP , Animais , Adesão Celular , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Homeostase , Humanos , Lactação , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Animais/fisiologia , Neoplasias Mamárias Animais/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/metabolismo , Organogênese , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo
5.
Biochem Biophys Res Commun ; 283(4): 854-61, 2001 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-11350063

RESUMO

Oncogenic transformation of fibroblasts by v-Src and v-Ras is often associated with downregulation of fibronectin (FN) and increased expression of CD44, a receptor for hyaluronan. Both v-Src and v-Ras as well as v-Raf activate phospholipase D through the small GTPase, RalA, an important mediator of transformation and tumorigenesis in vivo. We have therefore investigated whether RalA is involved in the downregulation of FN and overproduction of CD44 upon oncogenic transformation. We report here that compared to untransfected cells NIH3T3 cells transformed by v-Src, v-Ras, or v-Raf have reduced levels of FN and increased levels of CD44. Moreover, the ability to form extracellular FN fibrils was significantly reduced in the oncogene-transformed cells compared to parental controls. Coexpression of the dominant negative S28N-RalA mutant restored the levels of CD44 and FN and the capacity of v-Src-, v-Ras-, and v-Raf-expressing cells to form extracellular FN fibrils, to those observed in NIH3T3 cells. The data presented here show a novel regulatory role for RalA, which is required for tumor formation in transformed NIH3T3 cells, in mediating the signal transduction pathway activated by v-Src, v-Ras, and v-Raf, that leads to FN downregulation and CD44 overexpression.


Assuntos
Fibronectinas/metabolismo , GTP Fosfo-Hidrolases/fisiologia , Receptores de Hialuronatos/metabolismo , Proteína Oncogênica p21(ras)/metabolismo , Proteína Oncogênica pp60(v-src)/metabolismo , Proteínas ral de Ligação ao GTP , Células 3T3 , Animais , Linhagem Celular Transformada , Camundongos
6.
Mol Biol Cell ; 12(4): 863-79, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11294892

RESUMO

We discovered that a shift between the state of tumorigenicity and dormancy in human carcinoma (HEp3) is attained through regulation of the balance between two classical mitogen-activated protein kinase (MAPK)-signaling pathways, the mitogenic extracellular regulated kinase (ERK) and the apoptotic/growth suppressive stress-activated protein kinase 2 (p38(MAPK)), and that urokinase plasminogen activator receptor (uPAR) is an important regulator of these events. This is a novel function for uPAR whereby, when expressed at high level, it enters into frequent, activating interactions with the alpha5beta1-integrin, which facilitates the formation of insoluble fibronectin (FN) fibrils. Activation of alpha5beta1-integrin by uPAR generates persistently high level of active ERK necessary for tumor growth in vivo. Our results show that ERK activation is generated through a convergence of two pathways: a positive signal through uPAR-activated alpha5beta1, which activates ERK, and a signal generated by the presence of FN fibrils that suppresses p38 activity. When fibrils are removed or their assembly is blocked, p38 activity increases. Low uPAR derivatives of HEp3 cells, which are growth arrested (dormant) in vivo, have a high p38/ERK activity ratio, but in spite of a similar level of alpha5beta1-integrin, they do not assemble FN fibrils. However, when p38 activity is inhibited by pharmacological (SB203580) or genetic (dominant negative-p38) approaches, their ERK becomes activated, uPAR is overexpressed, alpha5beta1-integrins are activated, and dormancy is interrupted. Restoration of these properties in dormant cells can be mimicked by a direct re-expression of uPAR through transfection with a uPAR-coding plasmid. We conclude that overexpression of uPAR and its interaction with the integrin are responsible for generating two feedback loops; one increases the ERK activity that feeds back by increasing the expression of uPAR. The second loop, through the presence of FN fibrils, suppresses p38 activity, further increasing ERK activity. Together these results indicate that uPAR and its interaction with the integrin should be considered important targets for induction of tumor dormancy.


Assuntos
Fibronectinas/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores de Superfície Celular/fisiologia , Transdução de Sinais/fisiologia , Divisão Celular , Humanos , Integrina beta1/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Fibronectina/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno
7.
Curr Opin Cell Biol ; 12(5): 613-20, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10978898

RESUMO

Urokinase-type plasminogen activator receptor (uPAR) binds the urokinase-type plasminogen activator (uPA) and facilitates a proteolytic cascade focused at the cell surface. More recently, uPAR was recognized as a multifunctional protein that, through its interactions with integrins, initiates signaling events that alter cell adhesion, migration and proliferation. Results obtained recently have led to new insights into the structural aspects of uPAR interaction with integrins, provided a more detailed description of the signaling pathway they induce, and determined that uPAR signaling plays a role in cell migration and tumorigenicity.


Assuntos
Integrinas/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Adesão Celular/fisiologia , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Humanos , Ligação Proteica/fisiologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Transdução de Sinais
8.
Medicina (B Aires) ; 60 Suppl 2: 34-40, 2000.
Artigo em Espanhol | MEDLINE | ID: mdl-11188928

RESUMO

Deregulation of several signaling pathways have been found to be critical for the development of different types of tumors, both in transgenic and spontaneous models. The role of proteases and adhesion molecules during the early stages of tumor progression induced by oncogenes in epithelial and mesenchymal tumors has remained relatively unexplored. This review summarizes recent work showing that different but overlapping signaling effector modules (PKC, v-Ras-RalA-PLD1 or v-Src-RalA-PLD1) induce changes in the production of proteases (uPA and MMPs) and adhesion molecules (fibronectin, CD44, beta 1-integrin) in normal epithelial or mesenchymal cell lines, associated with tumor development in vivo. Overexpression of PKC gamma in normal mammary epithelial cells or of v-Src and v-Ras in NIH3T3 fibroblasts induced in all cases overproduction of uPA and MMPs and a tumorigenic phenotype. Proteases production and tumorigenicity in transformed NIH3T3 cells were dependent on the GTPase RalA. In contrast to the common outcome in protease production by the different tumor promoting stimuli, fibronectin production was high in PKC-overexpressing mammary epithelial cells and it was organized into a rich fibrillar matrix, while oncogene transformed fibroblasts displayed reduced fibronectin production and a total loss of FN fibrillogenesis, an effect also dependent on RalA. These results show that protease overexpression is a common denominator in the acquisition of a malignant phenotype both in mesenchymal and epithelial cells. In contrast there is a dramatic difference in the expression and function of adhesion molecules like fibronectin between these two cell types, suggesting different regulatory roles for this glycoprotein during tumor progression, in cells of different tissular origin.


Assuntos
Metaloproteinases da Matriz/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimologia , Proteína Quinase C/metabolismo , Transdução de Sinais/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Células 3T3 , Animais , Transformação Celular Neoplásica/metabolismo , Progressão da Doença , Humanos , Neoplasias Mamárias Experimentais/enzimologia , Camundongos , Invasividade Neoplásica , Proteína Quinase C/fisiologia
9.
Medicina [B Aires] ; 60 Suppl 2: 34-40, 2000.
Artigo em Espanhol | BINACIS | ID: bin-39706

RESUMO

Deregulation of several signaling pathways have been found to be critical for the development of different types of tumors, both in transgenic and spontaneous models. The role of proteases and adhesion molecules during the early stages of tumor progression induced by oncogenes in epithelial and mesenchymal tumors has remained relatively unexplored. This review summarizes recent work showing that different but overlapping signaling effector modules (PKC, v-Ras-RalA-PLD1 or v-Src-RalA-PLD1) induce changes in the production of proteases (uPA and MMPs) and adhesion molecules (fibronectin, CD44, beta 1-integrin) in normal epithelial or mesenchymal cell lines, associated with tumor development in vivo. Overexpression of PKC gamma in normal mammary epithelial cells or of v-Src and v-Ras in NIH3T3 fibroblasts induced in all cases overproduction of uPA and MMPs and a tumorigenic phenotype. Proteases production and tumorigenicity in transformed NIH3T3 cells were dependent on the GTPase RalA. In contrast to the common outcome in protease production by the different tumor promoting stimuli, fibronectin production was high in PKC-overexpressing mammary epithelial cells and it was organized into a rich fibrillar matrix, while oncogene transformed fibroblasts displayed reduced fibronectin production and a total loss of FN fibrillogenesis, an effect also dependent on RalA. These results show that protease overexpression is a common denominator in the acquisition of a malignant phenotype both in mesenchymal and epithelial cells. In contrast there is a dramatic difference in the expression and function of adhesion molecules like fibronectin between these two cell types, suggesting different regulatory roles for this glycoprotein during tumor progression, in cells of different tissular origin.

10.
J Cell Biol ; 147(1): 89-104, 1999 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-10508858

RESUMO

Mechanisms that regulate the transition of metastases from clinically undetectable and dormant to progressively growing are the least understood aspects of cancer biology. Here, we show that a large ( approximately 70%) reduction in the urokinase plasminogen activator receptor (uPAR) level in human carcinoma HEp3 cells, while not affecting their in vitro growth, induced a protracted state of tumor dormancy in vivo, with G(0)/G(1) arrest. We have now identified the mechanism responsible for the induction of dormancy. We found that uPA/uPAR proteins were physically associated with alpha5beta1, and that in cells with low uPAR the frequency of this association was significantly reduced, leading to a reduced avidity of alpha5beta1 and a lower adhesion of cells to the fibronectin (FN). Adhesion to FN resulted in a robust and persistent ERK1/2 activation and serum-independent growth stimulation of only uPAR-rich cells. Compared with uPAR-rich tumorigenic cells, the basal level of active extracellular regulated kinase (ERK) was four to sixfold reduced in uPAR-poor dormant cells and its stimulation by single chain uPA (scuPA) was weak and showed slow kinetics. The high basal level of active ERK in uPAR-rich cells could be strongly and rapidly stimulated by scuPA. Disruption of uPAR-alpha5beta1 complexes in uPAR-rich cells with antibodies or a peptide that disrupts uPAR-beta1 interactions, reduced the FN-dependent ERK1/2 activation. These results indicate that dormancy of low uPAR cells may be the consequence of insufficient uPA/uPAR/alpha5beta1 complexes, which cannot induce ERK1/2 activity above a threshold needed to sustain tumor growth in vivo. In support of this conclusion we found that treatment of uPAR-rich cells, which maintain high ERK activity in vivo, with reagents interfering with the uPAR/beta1 signal to ERK activation, mimic the in vivo dormancy induced by downregulation of uPAR.


Assuntos
Regulação para Baixo , Integrinas/metabolismo , Sistema de Sinalização das MAP Quinases , Neoplasias/patologia , Proteínas Serina-Treonina Quinases , Receptores de Superfície Celular/metabolismo , Animais , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Divisão Celular , Embrião de Galinha , Ativação Enzimática , Fibronectinas/metabolismo , Humanos , Cinética , MAP Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias/enzimologia , Neoplasias/metabolismo , Fosforilação , Testes de Precipitina , Ligação Proteica , Receptores de Superfície Celular/genética , Receptores de Fibronectina/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Células Tumorais Cultivadas , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
11.
Oncogene ; 18(33): 4718-25, 1999 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-10467419

RESUMO

Overproduction of urokinase-type plasminogen activator (uPA) and metalloproteases (MMPs) is strongly correlated with tumorigenicity and with invasive and metastatic phenotypes of human and experimental tumors. We demonstrated previously that overproduction of uPA in tumor cells is mediated by a phospholipase D (PLD)- and protein kinase C-dependent mechanism. The oncogenic stimulus of v-Src and v-Ras results in the activation of PLD, which is dependent upon the monomeric GTPase RalA. We have therefore investigated whether RalA plays a role in uPA and MMP overproduction that is observed in response to oncogenic signals. We report here that NIH3T3 cells transformed by both v-Src and v-Ras, constitutively overproduce uPA and that expression of a dominant negative RalA mutant (S28N) blocks overproduction of uPA in both the v-Src-and v-Ras-transformed cells. v-Src and v-Ras also induced an upregulation of the activity of MMP-2 and MMP-9 as detected by zymograms, however only the v-Src induction correlated with MMP protein levels detected by Western blot analysis. The dominant negative RalA mutant blocked increased MMP-2 and 9 overproduction induced by v-Src, but not the increased activity of MMP-2 and 9 induced by v-Ras. And, consistent with a role for the RalA/PLD pathway in mitogenesis and tumor development, the dominant negative RalA mutant completely blocked tumor formation by v-Src- and v-Ras-transformed NIH3T3 cells injected subcutaneously in syngeneic mice. The data presented here implicate RalA and PLD as signaling mediators for tumor formation and protease production by transformed cells.


Assuntos
Transformação Celular Neoplásica/genética , GTP Fosfo-Hidrolases/genética , Genes ras , Genes src , Metaloendopeptidases/biossíntese , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Proteínas ral de Ligação ao GTP , Células 3T3 , Animais , Colagenases/biossíntese , Gelatinases/biossíntese , Regulação Neoplásica da Expressão Gênica , Metaloproteinase 2 da Matriz , Metaloproteinase 9 da Matriz , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Regulação para Cima
12.
Int J Cancer ; 82(5): 748-53, 1999 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-10417775

RESUMO

Tumor invasion and metastasis development is a multistep process involving adhesion molecules as well as tumor proteases. It has been reported that tumor cells lacking fibronectin (FN) expression and engineered to re-express FN showed a marked reduction in metastatic ability. Besides its effects on cell adhesion and migration, FN could be modulating other cellular events associated with the metastatic cascade. To test this hypothesis, we analyzed the production of urokinase-type plasminogen activator (uPA), and its receptor (uPAR), 2 molecules involved in the invasive phenotype, in cells over-expressing RGD wild-type FN (FNwt clones) or RGD-mutated FN (FN RGD-minus clones). Secreted uPA activity and antigen were significantly up-regulated in FN-expressing clones, although RGD-minus cells secreted approximately 50% less uPA than the FNwt ones. Interestingly, while control and FN RGD-minus clones were able to readily bind uPA to their surface, FNwt clones exhibited impaired uPA binding. Furthermore, treatment of the parental cell line as well as the control and FN-expressing clones with exogenous purified FN or RGD peptides induced up-regulation of uPA production and the reduction of uPA membrane binding, which was associated with lower expression of uPAR. This modulation by FN was found to be dependent on RGD sequence and beta1 integrin. These results strongly suggest a novel activity for the multifunctional glycoprotein FN regarding the regulation of uPA production as well as the capacity of tumor cells to bind uPA.


Assuntos
Antineoplásicos/metabolismo , Fibronectinas/metabolismo , Neoplasias Mamárias Animais/enzimologia , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Animais , Movimento Celular , Indução Enzimática , Fibronectinas/genética , Integrina beta1/fisiologia , Neoplasias Mamárias Animais/patologia , Camundongos , Oligopeptídeos/fisiologia , Ligação Proteica , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/fisiologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Células Tumorais Cultivadas , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
13.
Eur J Biochem ; 263(2): 295-304, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10406935

RESUMO

We review the evidence in support of the notion that, upon experimental oncogenic transformation or in spontaneous human cancers, mitogenesis and expression of urokinase (uPA) and its receptor (uPAR) are activated through common signaling complexes and pathways. It is well documented that uPA, uPAR or metalloproteinases (MMPs) are overexpressed in tumor cells of mesenchymal or epithelial origin and these molecules are required for tumor invasion and metastasis. Furthermore, oncogenic stimuli, which may render the transformed cells tumorigenic and metastatic in vivo, activate, in a constitutive fashion, the extracellular-regulated kinases (Erk 1 and 2) classical mitogenic pathway and others such as the NH(2)-Jun-kinase (Jnk). Cells from human tumors or oncogene-transformed cells overexpress uPA and uPAR, and also show a sustained activation of the above-mentioned signaling modules. In this paper we show that the classical mitogenic pathway involving Ras-Erk, PKC-Erk or Rac-JNK, among others, is activated by growth factors or endogenously by oncogenes, and constitutively activates uPA and uPAR expression. All the data obtained from human tumors or experimental systems, incorporated into a general model, indicate that oncogenic stimuli lead to the constitutive activation of mitogenesis and uPA and its receptor expression, through the activation of the same classical and nonclassical signaling complexes and pathways that regulate cell proliferation. We also discuss contrasting points of view. For instance, what governs the differential regulation of mitogenesis and the signal that leads to protease overexpression in a way that allows normal cells during physiological events to respond to growth factors, and proliferate without overexpressing extracellular matrix (ECM) proteases? Or how can cells remodel their microenvironment without proliferating? What restrains benign tumors from overexpressing tumor-associated proteases when they certainly have the mitogenic signal fully activated? This may occur by the differential regulation of transcriptional programs and recent reports reviewed in this paper may provide an insight into how this occurs at the signaling and transcriptional levels.


Assuntos
Transdução de Sinais , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Ativador de Plasminogênio Tipo Uroquinase/genética , Transformação Celular Neoplásica , Regulação Neoplásica da Expressão Gênica , Humanos , Modelos Biológicos , Proteína Supressora de Tumor p53/metabolismo
14.
Medicina (B Aires) ; 59(5 Pt 2): 547-52, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10684157

RESUMO

Urokinase plasminogen activator receptor (uPAR) has been identified some 15 years ago and the anticipation was that its presence on the cell surface will provide a focus for anchoring uPA and possibly protect the enzyme from native inhibitors. The studies of the last decade have shown that uPA localized to the surface of cells by uPAR is indeed an important factor in the process of cancer cell invasion and metastasis. We developed a chick embryo model in which we showed that uPAR is crucial in invasion of stroma and in intravasation (breaching of the blood vessels walls). More recently and unexpectedly, uPAR--a protein anchored in the outer leaf-let of the plasma membrane, has been shown to initiate signal transduction events and affect cell migration. We have shown that uPAR co-associates with fibronectin binding integrin, alpha 5 beta 1, activates them and that this interaction leads to a greatly increased level of active ERK. When the association between uPAR and integrin or integrin and fibronectin are interrupted either by reduction of surface uPAR expression, or by other means, human carcinoma cells enter a state of protracted dormancy. We show that very high levels of active ERK are required to keep cancer cells proliferating in vivo.


Assuntos
Invasividade Neoplásica , Neoplasias/fisiopatologia , Ativadores de Plasminogênio/fisiologia , Receptores de Superfície Celular/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Animais , Embrião de Galinha , Humanos , Metástase Neoplásica , Neoplasias/metabolismo , Transdução de Sinais , Fatores de Tempo
15.
Int J Cancer ; 78(6): 727-34, 1998 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-9833766

RESUMO

The invasion and metastasis process involves degradation of the extracellular matrix mediated by tumor- and host-produced proteolytic enzymes. The main enzymes involved in this process are urokinase-type plasminogen activator (uPA) and the matrix metalloproteinases (MMPs). Calcium is a main co-factor in the signaling pathways that regulate cell proliferation and protease production. We have studied here the effect of verapamil, a calcium channel blocker widely used to treat hypertensive diseases, on local tumor growth, spontaneous and experimental metastasis development, tumor-associated protease production and circulating MMP activity in tumor-bearing mice. BALB/c mice treated for 45 days with verapamil showed no toxic effects. Oral administration of verapamil to mice injected with F311 tumor cells, either pre-treated or not with verapamil, showed a significant decrease of local tumor invasion and both spontaneous and experimental metastasis development (51.3% inhibition of metastasis in both cases, p < 0.01). uPA and MMP-9 production by tumor cells in vitro was significantly inhibited by verapamil in a dose-dependent manner, showing a long-term inhibition after removal of the drug. Verapamil also exhibited a marked cytostatic effect on F311 cell proliferation in vitro. In addition, circulating MMP activity, usually enhanced in tumor-bearing mice, diminished significantly with all verapamil treatments. Our results suggest that modulation of the calcium-dependent signaling pathways that regulate tumor- or host-dependent production of proteases and tumor cell proliferation could contribute to the inhibition of metastasis development. Finally, we describe the inhibitory effects of a commonly used hypotensor in humans, verapamil, on the invasive and metastatic capacity of mammary tumor cells.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Endopeptidases/metabolismo , Verapamil/farmacologia , Animais , Divisão Celular , Relação Dose-Resposta a Droga , Neoplasias Pulmonares/secundário , Masculino , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Metástase Neoplásica , Células Tumorais Cultivadas , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
16.
Exp Cell Res ; 242(2): 515-27, 1998 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9683538

RESUMO

The cell surface glycoprotein CD44 is proposed as a main participant in cell adhesion and migration. We studied the function, expression, and distribution of CD44 in the invasive and metastatic F3II murine carcinoma cell line during adhesion, spreading, migration, and invasion. A mAb anti-CD44 (KM 201) dramatically blocked F3II cell adhesion on both plastic and hyaluronic acid coatings, as well as spreading on uncoated plastic surfaces (P < 0.01). KM201 mAb significantly inhibited F3II cell migration and invasion in Transwell chambers. Immunocytochemistry of spreading cells revealed that CD44 distributed in bands on the cell surface, particularly in the tip of leading edges and in the perinuclear zones of the cell membrane. CD44 antigen was never detected in filopodia or lamellipodia nor in focal adhesion-like structures, but was also detectable as strong interlamellar bands. Fully spread cells showed a decreased CD44 signal compared to cells in early stages of spreading. This decrease correlated with a reduced expression of CD44 as detected by Western blot. We also investigated the signals that may regulate CD44 expression in F3II cells. Treatment of F3II cells, with phorbol myristate acetate (PMA) or phosphatidic acid (PA, the product of PLD-dependent hydrolysis of phosphatidylcholine), significantly enhanced CD44 expression. Conversely, the treatment of F3II cells with H7, a specific PKC inhibitor, or propranolol, which blocks PA conversion to DAG, significantly decreased CD44 expression levels. These results suggest the involvement of PKC and PLD pathways in CD44 expression. These results demonstrate that CD44 plays an important role during F3II cells adhesion, spreading, migration, and invasion. In addition we provide information linking the PLD- and PKC-dependent pathways with the regulation of CD44 expression.


Assuntos
Movimento Celular/fisiologia , Receptores de Hialuronatos/biossíntese , Receptores de Hialuronatos/fisiologia , Neoplasias Mamárias Animais/patologia , Células Tumorais Cultivadas/citologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Carcinógenos/farmacologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Receptores de Hialuronatos/imunologia , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/fisiopatologia , Camundongos , Ácidos Fosfatídicos/farmacologia , Fosfolipase D/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
17.
Int J Cancer ; 76(3): 362-7, 1998 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-9579573

RESUMO

Urokinase-type plasminogen activator (uPA) is a key serine protease involved in invasion and metastasis. We had shown that overproduction of uPA in tumor cells is controlled by a phospholipase D-protein kinase C-dependent pathway. Now we studied whether other signaling pathways participate in the regulation of constitutive uPA and metalloproteinase (MMP) overproduction in tumor cells. Staurosporine, a protein kinase inhibitor, stimulated uPA and MMP-9 secretion as measured by radial caseinolysis, zymography and Western blotting. Genistein, a specific tyrosine kinase inhibitor, reduced the constitutive and staurosporine-induced uPA and MMP-9 secretion. Interestingly, the phosphatase inhibitor vanadate stimulated uPA secretion. Verapamil, a calcium channel blocker, inhibited both endogenous and PMA-stimulated secretion of uPA but was unable to inhibit staurosporine-induced secretion. The alcohol n-butanol, a phospholipase D and protein kinase C inhibitor, besides inhibiting constitutive uPA secretion, blocked staurosporine-induced secretion. Our results suggest that constitutive and staurosporine-induced uPA and MMP-9 secretion by LM3 murine mammary tumor cells is controlled by an endogenous tyrosine kinase pathway and probably involves protein phosphatases. In addition, the staurosporine-induced signal regulating urokinase secretion is independent of extracellular calcium but dependent on phospholipase D.


Assuntos
Inibidores Enzimáticos/farmacologia , Neoplasias Mamárias Animais/enzimologia , Metaloendopeptidases/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Estaurosporina/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/efeitos dos fármacos , Animais , Cálcio/metabolismo , Feminino , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ativadores de Plasminogênio/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
18.
Int J Cancer ; 71(5): 881-90, 1997 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-9180160

RESUMO

Spreading is a critical process involved in motility and growth of tumor cells during the metastatic cascade. Focal adhesion kinase, src-proteins and PKC have been reported to participate in the regulation of cytoskeleton organization in both normal and transformed cells during spreading. The role of other signaling enzymes such as PLD and PAP has not been studied during spreading in tumor cells. We now show that the spreading of murine mammary adenocarcinoma LM3 cells was significantly reduced by n-butanol, a PLD and PKC inhibitor, with a maximal inhibition of 54% (p < 0.001) in both the presence and absence of serum, as measured by phase-contrast microscopy. PMA only stimulated cell spreading over the control in the absence of serum and n-butanol inhibition was completely reversed by PMA treatment in both conditions. PA, the product of PLD activity, stimulated LM3 cell spreading and the same effect was observed with staurosporine. Spreading was enhanced when cells were seeded on collagen-IV- or fibronectin-coated surfaces and n-butanol could inhibit both integrin-derived signals. Cell spreading inhibition correlated with the absence of f-actin bundles and fewer beta1-integrin point contacts as determined by double immunofluorescence microscopy. In addition, n-butanol inhibited the proliferation of LM3 cells in the presence of serum (p < 0.01). These results suggest that beta1-integrin and f-actin/point contact assembly, involved in spreading and proliferation, require the participation of PLD-PKC regulatory pathways in LM3 cells.


Assuntos
Actinas/análise , Adenocarcinoma/patologia , Integrina beta1/análise , Neoplasias Mamárias Experimentais/patologia , Fosfolipase D/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , 1-Butanol , Animais , Butanóis/farmacologia , Adesão Celular , Divisão Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Genisteína , Isoflavonas/farmacologia , Camundongos , Microscopia de Fluorescência , Ácidos Fosfatídicos/farmacologia , Transdução de Sinais , Estaurosporina/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas
19.
Biochim Biophys Acta ; 1356(2): 171-84, 1997 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-9150275

RESUMO

Urokinase-type plasminogen activator (uPA) initiates a proteolytic cascade with which invasive cells eliminate barriers to movement. The signaling pathways regulating uPA production in tumor cells remain unclear. We first studied the effects of n-butanol, a phospholipase D (PLD) and protein kinase C (PKC) inhibitor, on the production of uPA in murine mammary adenocarcinoma cells. Tumor cell monolayers treated during 24 h with 0.3% v/v n-butanol, secreted 45-50% less uPA to the culture medium than control monolayers (P < 0.001) as determined by radial caseinolysis, zymography and western blot. This inhibition occurred also with 5-h treatments and remained up to 5 h after the removal of the alcohol. Treatment with the phorbol ester PMA or with EGF, strongly increased uPA production (P < 0.001). Interestingly, a mild inhibition of uPA production was observed when PMA stimulation was assayed in cotreatments with n-butanol. In contrast EGF was unable to reverse the inhibition induced by n-butanol. H7 significantly inhibited uPA activity (P < 0.001) secreted to the culture media. Furthermore, phosphatidic acid significantly stimulated uPA production meanwhile propranolol, which blocks phosphatidic acid availability, reduced it, suggesting a main regulatory role for this intermediary metabolite. These results suggest for the first time that uPA production is regulated by PLD and PKC signal transduction pathways in murine mammary adenocarcinoma cells.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Fosfolipase D/metabolismo , Proteína Quinase C/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , 1-Butanol , Animais , Butanóis/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Feminino , Camundongos , Ácidos Fosfatídicos/farmacologia , Fosfolipase D/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , Transdução de Sinais , Temperatura , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas/metabolismo , Regulação para Cima/efeitos dos fármacos
20.
Tumour Biol ; 18(1): 41-52, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-8989924

RESUMO

We studied the effect of tumoral microenvironments on metastatic phenotypes. Therefore, murine mammary adenocarcinoma cells cultured in vivo in diffusion chambers (DC) were implanted intraperitoneally in BALB/c mice. The behavior of DC-cultured cells was compared with that of cells obtained from tumors growing subcutaneously or intraperitoneally and from primary cultures in vitro of the former. DC-cultured and control cells were inoculated into normal mice to evaluate their tumorigenicity and metastasizing ability. We found that DC-cultured cells were less tumorigenic and metastatic both in spontaneous and in experimental metastasis assays. The host response to tumor progression resulted in an early leukocytosis, probably due to the overproduction of a hematopoietic factor by the tumor cells. Finally, it was found that DC-cultured cells produced lower levels of urokinase-type plasminogen activator activity, while no differences were found in the metalloproteinase production compared to cells obtained from a tumor growing subcutaneously.


Assuntos
Adenocarcinoma/fisiopatologia , Neoplasias Mamárias Experimentais/fisiopatologia , Adenocarcinoma/patologia , Adenocarcinoma/secundário , Animais , Contagem de Células Sanguíneas , Cultura em Câmaras de Difusão/métodos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Metaloendopeptidases/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Fenótipo , Células Tumorais Cultivadas , Ativador de Plasminogênio Tipo Uroquinase/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...