Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(9): 3544-9, 2014 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-24520174

RESUMO

The coagulation system links immediate (hemostatic) and late (inflammatory, angiogenic) tissue responses to injury, a continuum that often is subverted in cancer. Here we provide evidence that tumor dormancy is influenced by tissue factor (TF), the cancer cell-associated initiator of the coagulation system and a signaling receptor. Thus, indolent human glioma cells deficient for TF remain viable but permanently dormant at the injection site for nearly a year, whereas the expression of TF leads to a step-wise transition to latent and overt tumor growth phases, a process that is preceded by recruitment of vascular (CD105(+)) and myeloid (CD11b(+) and F4/80(+)) cells. Importantly, the microenvironment orchestrated by TF expression drives permanent changes in the phenotype, gene-expression profile, DNA copy number, and DNA methylation state of the tumor cells that escape from dormancy. We postulate that procoagulant events in the tissue microenvironment (niche) may affect the fate of occult tumor cells, including their biological and genetic progression to initiate a full-blown malignancy.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Glioma/fisiopatologia , Processos Neoplásicos , Tromboplastina/metabolismo , Microambiente Tumoral/genética , Animais , Linhagem Celular Tumoral , Variações do Número de Cópias de DNA , Metilação de DNA , Perfilação da Expressão Gênica , Inativação Gênica , Glioma/metabolismo , Humanos , Camundongos , Mutação/genética , Estatísticas não Paramétricas
2.
Biochem Biophys Res Commun ; 454(2): 262-8, 2014 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-25450387

RESUMO

Glioblastoma multiforme (GBM) is an aggressive form of glial brain tumors, associated with angiogenesis, thrombosis, and upregulation of tissue factor (TF), the key cellular trigger of coagulation and signaling. Since TF is upregulated by oncogenic mutations occurring in different subsets of human brain tumors we investigated whether TF contributes to tumourigenesis driven by oncogenic activation of EGFR (EGFRvIII) and RAS pathways in the brain. Here we show that TF expression correlates with poor prognosis in glioma, but not in GBM. In situ, the TF protein expression is heterogeneously expressed in adult and pediatric gliomas. GBM cells harboring EGFRvIII (U373vIII) grow aggressively as xenografts in SCID mice and their progression is delayed by administration of monoclonal antibodies blocking coagulant (CNTO 859) and signaling (10H10) effects of TF in vivo. Mice in which TF gene is disrupted in the neuroectodermal lineage exhibit delayed progression of spontaneous brain tumors driven by oncogenic N-ras and SV40 large T antigen (SV40LT) expressed under the control of sleeping beauty transposase. Reduced host TF levels in low-TF/SCID hypomorphic mice mitigated growth of glioma subcutaneously but not in the brain. Thus, we suggest that tumor-associated TF may serve as therapeutic target in the context of oncogene-driven disease progression in a subset of glioma.


Assuntos
Neoplasias Encefálicas/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioma/genética , Oncogenes , Tromboplastina/genética , Adolescente , Adulto , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Deleção de Genes , Glioblastoma/diagnóstico , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioma/diagnóstico , Glioma/metabolismo , Glioma/patologia , Humanos , Camundongos , Camundongos SCID , Prognóstico , Tromboplastina/metabolismo
3.
Semin Thromb Hemost ; 40(3): 284-95, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24599437

RESUMO

Genetically altered cancer cells both provoke and respond to changes in their microenvironment, stroma, and vasculature. This includes local and systemic activation of the coagulation system, which is a part of the functional continuum involving inflammation, angiogenesis, and tissue repair programs, often reactivated in cancer. These responses coevolve with, and contribute to, the malignant process. Cancer coagulopathy is not only a source of comorbidity and mortality in cancer patients, but it also affects the disease biology including processes of tumor growth, initiation, dormancy, invasion, angiogenesis, metastasis, and therapeutic responsiveness. Notably, genetic and cellular differences between different cancer types are paralleled by a degree of diversity in the related coagulation system perturbations. Although some of these differences may be unspecific, iatrogenic, or indirect in nature, others are affected by oncogenic pathways (RAS, EGFR, HER2, MET, PTEN, and TP53) activated in cancer cells due to driver mutations of critical genes. Such mutations cooperate with hypoxia, cellular differentiation, and other influences to alter the expression of tissue factor, protease-activated receptors (e.g., PAR-1 and PAR-2), coagulation factors (FII and FVII), and other molecules related to the hemostatic system. Oncogenic pathways also control secretion of some of these entities from cancer cells, either as soluble proteins, or as cargo of extracellular vesicles/microparticles. Moreover, emerging evidence suggests that the expression profiles of coagulation-related genes differ between molecularly and genetically distinct subgroups of specific malignancies such as glioblastoma multiforme and medulloblastoma. Certain hereditary thrombophilias may also affect cancer pathogenesis. We suggest that mechanisms of cancer coagulopathy may be more diverse and genetically modulated than hitherto realized. If so, a possibility may exist to deliver more personalized, biologically based, anticoagulation, and thereby improve patient survival.


Assuntos
Neoplasias/complicações , Neoplasias/genética , Trombose/complicações , Trombose/genética , Animais , Humanos , Neoplasias/sangue
4.
Exp Cell Res ; 319(17): 2747-57, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23954818

RESUMO

Transitions of the cancer cell phenotype between epithelial and mesenchymal states (EMT) are likely to alter the patterns of intercellular communication. In this regard we have previously documented that EMT-like changes trigger quantitative rearrangements in exosomal vesicle emission in A431 cancer cells driven by oncogenic epidermal growth factor receptor (EGFR). Here we report that extracellular vesicles (EVs) produced by these cancer cells in their epithelial and mesenchymal states exhibit profound qualitative differences in their proteome. Thus, induction of the EMT-like state through blockade of E-cadherin and EGFR stimulation provoked a mesenchymal shift in cellular morphology and enrichment in the CD44-high/CD24-low immunophenotype, often linked to cellular stemness. This change also resulted in reprogramming of the EV-related proteome (distinct from that of corresponding cells), which contained 30 unique protein signals, and revealed enrichment in pathways related to cellular growth, cell-to-cell signaling, and cell movement. Some of the most prominent EV-related proteins were validated, including integrin α2 and tetraspanin CD9. We propose that changes in cellular differentiation status translate into unique qualitative rearrangements in the cargo of EVs, a process that may have implications for intercellular communication and could serve as source of new biomarkers to detect EMT-like processes in cancer.


Assuntos
Transição Epitelial-Mesenquimal , Exossomos/metabolismo , Proteoma/genética , Antígeno CD24/genética , Antígeno CD24/metabolismo , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Linhagem Celular Tumoral , Reprogramação Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Integrina alfa2/genética , Integrina alfa2/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteoma/metabolismo , Tetraspanina 29/genética , Tetraspanina 29/metabolismo
5.
J Biol Chem ; 287(52): 43565-72, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23118232

RESUMO

Aggressive epithelial cancer cells frequently adopt mesenchymal characteristics and exhibit aberrant interactions with their surroundings, including the vasculature. Whether the release/uptake of extracellular vesicles (EVs) plays a role during these processes has not been studied. EVs are heterogeneous membrane structures that originate either at the surface (microparticles), or within (exosomes) activated or transformed cells, and are involved in intercellular trafficking of bioactive molecules. Here, we show that epithelial cancer cells (A431, DLD-1) adopt mesenchymal features (epithelial-to-mesenchymal transition-like state) upon activation of epidermal growth factor receptor (EGFR) coupled with blockade of E-cadherin. This treatment leads to a coordinated loss of EGFR and tissue factor (TF) from the plasma membrane and coincides with a surge in emission of small, exosome-like EVs containing both receptors. TF (but not EGFR) is selectively up-regulated in EVs produced by mesenchymal-like cancer cells and can be transferred to cultured endothelial cells rendering them highly procoagulant. We postulate that epithelial-to-mesenchymal transition-like changes may alter cancer cell interactions with the vascular systems through altered vesiculation and TF shedding.


Assuntos
Comunicação Celular , Micropartículas Derivadas de Células/metabolismo , Receptores ErbB/metabolismo , Exossomos/metabolismo , Neoplasias Epiteliais e Glandulares/metabolismo , Tromboplastina/metabolismo , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Micropartículas Derivadas de Células/genética , Micropartículas Derivadas de Células/patologia , Transição Epitelial-Mesenquimal , Receptores ErbB/genética , Exossomos/genética , Exossomos/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/patologia , Tromboplastina/genética
6.
Semin Thromb Hemost ; 39(8): 881-95, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24108471

RESUMO

Brain vasculature is uniquely programmed to protect central nervous system tissues and respond to their metabolic demands. These functions are subverted during the development of primary and metastatic brain tumors, resulting in vascular perturbations that are thought to contribute to progression and comorbidities of the underlying disease, including thrombosis and hemorrhage. Chronic activation of the coagulation system is particularly obvious in glioblastoma multiforme (GBM), where intratumoral vasoocclusive thrombosis may contribute to hypoxia, pseudopalisading necrosis, and angiogenesis. GBM is also associated with spontaneous or iatrogenic bleeding, and the emission of circulating procoagulants implicated in the unusually high risk of peripheral venous thromboembolism. Tissue factor (TF) expression is elevated in several types of brain tumors, including adult and pediatric GBM, as is the production of TF-containing microparticles (TF-MPs). Both TF expression and its vesicular emission are regulated by tumor microenvironment (e.g., hypoxia), in concert with activated oncogenic and growth factor pathways (RAS, EGFR, MET), as well as the loss of tumor suppressor gene activity (PTEN). Discovery of distinct oncogenic networks led to recognition of unique molecular subtypes within brain tumors, of which GBM (proneural, neural, classical, and mesenchymal), and medulloblastoma (SHH, WNT, group 3, and group 4) exhibit subtype-specific composition of the tumor coagulome. It remains to be established whether mechanisms of thrombosis and biological effects of coagulation in brain tumors are also subtype specific. In this regard, TF pathway represents a paradigm, and its impact on tumor dormancy, inflammation, angiogenesis, formation of cancer stem cell niches, and dissemination is a subject of considerable interest. However, establishing the extent to which TF and TF-MPs contribute to pathogenesis and thromboembolic disease in the context of primary and secondary brain tumors may require molecular stratification of patient populations. We suggest that a better understanding of these molecular linkages may pave the way to a more effective (targeted) therapy, prophylaxis, adjunctive use of anticoagulants, and other agents able to modulate interactions between brain tumors and the coagulation system.


Assuntos
Coagulação Sanguínea , Neoplasias Encefálicas/irrigação sanguínea , Glioblastoma/irrigação sanguínea , Neovascularização Patológica/patologia , Adulto , Criança , Humanos , Modelos Biológicos , Neovascularização Patológica/metabolismo , Neovascularização Patológica/fisiopatologia , Transdução de Sinais , Tromboplastina/metabolismo
7.
Blood ; 116(5): 815-8, 2010 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-20462964

RESUMO

Cancer cells frequently overexpress tissue factor (TF) and become procoagulant. This conversion may be driven by genetic transformation, including through the expression of the oncogenic epidermal growth factor receptor (EGFR) and its mutant, EGFRvIII, present in glioblastoma multiforme (GBM). Here we show that the EGFRvIII-dependent GBM cell transformation is associated with the onset of the simultaneous overexpression of TF, protease-activated receptors 1 and 2 (PAR1 and PAR2), and ectopic synthesis of factor VII (FVII). Efficient generation of factor Xa by these cells still requires exogenous FVIIa. However, as a result of EGFRvIII-dependent transformation, GBM cells become hypersensitive to TF/PAR-mediated signaling and produce ample angiogenic factors (vascular endothelial growth factor and interleukin-8) on exposure to FVIIa and PAR1- or PAR2-activating peptides. Thus, oncogenes may cause complex changes in the ability of GBM cancer cells to interact with the coagulation system, thereby exacerbating its influence on angiogenesis and disease progression.


Assuntos
Neoplasias Encefálicas/genética , Receptores ErbB/fisiologia , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/genética , Mutação , Proteínas de Neoplasias/fisiologia , Oncogenes , Transdução de Sinais/genética , Tromboplastina/fisiologia , Regulação para Cima/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral/metabolismo , Genes erbB-1 , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Receptor PAR-1/biossíntese , Receptor PAR-1/genética , Receptor PAR-2/biossíntese , Receptor PAR-2/genética , Tromboplastina/biossíntese , Tromboplastina/genética
8.
Arterioscler Thromb Vasc Biol ; 29(12): 2005-14, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19628788

RESUMO

A common feature in the progression of multiple human malignancies is the protracted deregulation of the coagulation system, often referred to as cancer coagulopathy. Indeed, cancer cells and their vascular stroma often exhibit procoagulant properties, of which deregulation of tissue factor (TF) expression is a notable, although not the sole example. These changes can be traced to oncogenic influences affecting epidermal growth factor receptor (EGFR), EGFRvIII, K-ras, p53, PTEN, and probably many other proto-oncogenes and tumor suppressors in tumor parenchyma. Cancer stem cells (CSCs)/tumor initiating cells (TICs) are thought to represent the primary target and the main cellular effector through which oncogenic mutations exert their tumor-inducing effects. In so doing, CSCs/TICs depend on interactions with the tumor vasculature, which forms supportive niches for their clonal growth. We postulate that TF contributes to these interactions (directly or indirectly) through procoagulant and signaling effects, the latter executed in concert with juxtaposed protease activated receptors (mainly PAR-1 and PAR-2). TF/PAR system acts as a "blood sensing" mechanism, whereby cancer cells, including CSCs/TICs, may respond to plasma proteases (Factors VIIa, Xa, and IIa) and their related microenvironmental changes (fibrin deposition, activation of platelets). A growing body of still largely circumstantial evidence suggests that these events may contribute to the CSC/TIC niche, which could influence tumor initiation, metastasis, recurrence, and therapeutic intractability. Indeed, certain types of cancer cells harboring markers of CSCs (CD133) exhibit elevated TF expression and depend on this receptor to efficiently initiate tumor growth. We propose that both tumor cell-associated and host-related TF could influence the properties of CSCs, and that agents targeting the TF/PAR system may represent a hitherto unappreciated therapeutic opportunity to control cancer progression by influencing the CSC/TIC compartment.


Assuntos
Células-Tronco Neoplásicas/fisiologia , Tromboplastina/fisiologia , Coagulação Sanguínea , Transformação Celular Neoplásica , Genes Supressores de Tumor , Humanos , Modelos Biológicos , Neoplasias/sangue , Neoplasias/irrigação sanguínea , Neoplasias/etiologia , Neoplasias/fisiopatologia , Neovascularização Patológica , Oncogenes , Receptores Ativados por Proteinase/fisiologia , Transdução de Sinais
9.
Thromb Res ; 120 Suppl 2: S82-91, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18023719

RESUMO

Oncogenic events play an important role in cancer-related coagulopathy (Trousseau syndrome), angiogenesis and disease progression. This can, in part, be attributed to the up-regulation of tissue factor (TF) and release of TF-containing microvesicles into the pericellular milieu and the circulation. In addition, certain types of host cells (stromal cells, inflammatory cells, activated endothelium) may also express TF. At present, the relative contribution of host- vs tumor-related TF to tumor progression is not known. Our recent studies have indicated that the role of TF in tumor formation is complex and context-dependent. Genetic or pharmacological disruption of TF expression/activity in cancer cells leads to tumor growth inhibition in immunodeficient mice. This occurred even in the case of xenotransplants of human cancer cells, in which TF overexpression is driven by potent oncogenes (K-ras or EGFR). Interestingly, the expression of TF in vivo is not uniform and appears to be influenced by many factors, including the level of oncogenic transformation, tumor microenvironment, adhesion and the coexpression of markers of cancer stem cells (CSCs). Thus, minimally transformed, but tumorigenic embryonic stem (ES) cells were able to form malignant and angiogenic outgrowths in the absence of TF. However, these tumors were growth inhibited in hosts (mice) with dramatically reduced TF expression (low-TF mice). Depletion of host TF also resulted in changes affecting vascular patterning of some, but not all types of tumors. These observations suggest that TF may play different roles growth and angiogenesis of different tumors. Moreover, both tumor cell and host cell compartments may, in some circumstances, contribute to the functional TF pool. We postulate that activation of the coagulation system and TF signaling, may deliver growth-promoting stimuli (e.g. fibrin, thrombin, platelets) to dormant cancer stem cells (CSCs). Functionally, these influences may be tantamount to formation of a provisional (TF-dependent) cancer stem cell niche. As such these changes may contribute to the involvement of CSCs in tumor growth, angiogenesis and metastasis.


Assuntos
Neoplasias/patologia , Neovascularização Patológica , Oncogenes , Tromboplastina/metabolismo , Animais , Coagulação Sanguínea , Progressão da Doença , Humanos , Camundongos , Neoplasias/irrigação sanguínea , Neoplasias/genética , Células-Tronco Neoplásicas/fisiologia , Transdução de Sinais
10.
Oncotarget ; 7(32): 51991-52002, 2016 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-27437771

RESUMO

Extracellular vesicles (EVs) enable the exit of regulatory, mutant and oncogenic macromolecules (proteins, RNA and DNA) from their parental tumor cells and uptake of this material by unrelated cellular populations. Among the resulting biological effects of interest is the notion that cancer-derived EVs may mediate horizontal transformation of normal cells through transfer of mutant genes, including mutant ras. Here, we report that H-ras-mediated transformation of intestinal epithelial cells (IEC-18) results in the emission of exosome-like EVs containing genomic DNA, HRAS oncoprotein and transcript. However, EV-mediated horizontal transformation of non-transformed cells (epithelial, astrocytic, fibroblastic and endothelial) is transient, limited or absent due to barrier mechanisms that curtail the uptake, retention and function of oncogenic H-ras in recipient cells. Thus, epithelial cells and astrocytes are resistant to EV uptake, unless they undergo malignant transformation. In contrast, primary and immortalized fibroblasts are susceptible to the EV uptake, retention of H-ras DNA and phenotypic transformation, but these effects are transient and fail to produce a permanent tumorigenic conversion of these cells in vitro and in vivo, even after several months of observation. Increased exposure to EVs isolated from H-ras-transformed cancer cells, but not to those from their indolent counterparts, triggers demise of recipient fibroblasts. Uptake of H-ras-containing EVs stimulates but fails to transform primary endothelial cells. Thus, we suggest that intercellular transfer of oncogenes exerts regulatory rather than transforming influence on recipient cells, while cancer cells may often act as preferential EV recipients.


Assuntos
Comunicação Celular/fisiologia , Transformação Celular Neoplásica/genética , Vesículas Extracelulares/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Vesículas Extracelulares/genética , Vesículas Extracelulares/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Xenoenxertos , Humanos , Camundongos , Camundongos SCID , Proteínas Proto-Oncogênicas p21(ras)/genética
11.
Thromb Res ; 133 Suppl 2: S1-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24862126

RESUMO

Cancers arise and progress genetically amidst profound perturbations of the microenvironmental and systemic homeostasis. This includes the coagulation system, which is a part of the vascular milieu (niche) that remains under the control of molecular events occurring within the cancer cell genome. Thus, activation of several prototypic oncogenic pathways, such as RAS, EGFR, HER2, MET, SHH and loss of tumor suppressors (PTEN, TP53) alter the expression, activity and vesicular release of coagulation effectors, as exemplified by tissue factor (TF). The cancer-specific determinants of coagulopathy are also illustrated by the emerging link between the expression profiles of coagulation-related genes (coagulome) in glioblastoma multiforme (GBM), medulloblastoma (MB) and possibly other cancers and molecular subtypes of these respective tumors. The state of the coagulome is consequential for growth, metastasis and angiogenesis of established tumors, but could potentially also affect dormant cancer cells. For example, TF expression may trigger awakening of dormant glioma cells in mice in a manner involving recruitment of vascular and inflammatory cells, and resulting in lasting changes in the cancer cell genome and epigenome. Thus, coagulation system effectors could act as both targets and (indirect) inducers of genetic tumor progression, and a better understanding of this link may hold new diagnostic and therapeutic opportunities.


Assuntos
Coagulação Sanguínea/genética , Neoplasias Encefálicas , Glioma , Oncogenes/fisiologia , Animais , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Cerebelares/sangue , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/patologia , Glioma/sangue , Glioma/genética , Glioma/secundário , Humanos , Meduloblastoma/sangue , Meduloblastoma/genética , Meduloblastoma/secundário , Transcriptoma , Microambiente Tumoral/genética
12.
Mech Ageing Dev ; 140: 13-22, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25068886

RESUMO

Antiangiogenic therapies in cancer exert their effects in the context of age-related comorbidities, which affect the entirety of the vascular system. Among those conditions, the impact of atherosclerosis is especially prevalent, but poorly understood, and not reflected in mouse models routinely used for testing antiangiogenic therapeutics. Our earlier work suggested that these obstacles can be overcome with the use of atherosclerosis-prone ApoE-/- mice harbouring syngeneic transplantable Lewis Lung Carcinoma (LLC). Here we report that, sunitinib, the clinically approved, antiangiogenic inhibitor impedes global tumor growth to a greater extent in aged then in young mice. This activity was coupled with changes in the tumor microenvironment, which in aged mice was characterized by pronounced hypoxia, reduction in microvascular density (MVD) and lower pericyte coverage, relative to young controls. We also detected soluble VEGR2 in plasma of sunitinib treated mice. Interestingly, sunitinib modulated tumor infiltration with bone marrow-derived cells (CD45+), recruitment of M2-like macrophages (CD163+) and activation of inflammatory pathways (phospho-STAT3) in a manner that was age-dependent. We suggest that age and atherosclerosis may alter the effects of sunitinib on the tumor microenvironment, and that these considerations may also apply more broadly to other forms of antiangiogenic treatment in cancer.


Assuntos
Envelhecimento/fisiologia , Inibidores da Angiogênese/uso terapêutico , Aterosclerose/complicações , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Indóis/uso terapêutico , Pirróis/uso terapêutico , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Carcinoma Pulmonar de Lewis/patologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transplante de Neoplasias , Sunitinibe , Resultado do Tratamento
13.
Mech Ageing Dev ; 134(3-4): 118-29, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23403123

RESUMO

Ageing impacts multiple host mechanisms involved in cancer progression. Here we show that poorly metastatic Lewis lung carcinoma (LLC) cells form less bulky metastatic deposits in aged mice (>52 weeks) relative to their young (4-6 weeks) counterparts. Serial selection of LLC cells for increased metastatic capability in either young or old mice led in both cases to exaggerated growth of pulmonary nodules after only 5 cycles of in vivo passage. The respective metastatic cellular variants established in young (Y-series) or old (O-series) mice differed in cell morphology and constitutive activity of growth factor receptors, especially phospho-PDGFRa and phospho-EPHA7. These cell lines also exhibited marked differences in their time dependent profiles of cellular impedance (CI), which reflects their physical properties, such as cell shape, adhesion and interactions with substrata. In confluent monolayer culture Y-series cell lines generated high and increasing CI values, while these values remained low and constant in the O-series of cell lines. These observations suggest that the selective pressure of the metastatic microenvironment in young versus old hosts is sufficiently different to results in the enrichment of distinct, age-related metastatic phenotypes of cancer cells. Thus, age could inform therapeutic approaches to metastatic cancers.


Assuntos
Envelhecimento , Metástase Neoplásica , Neoplasias/patologia , Animais , Carcinoma Pulmonar de Lewis , Adesão Celular , Ciclo Celular , Forma Celular , Progressão da Doença , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Fosforilação , Fatores de Tempo
14.
Thromb Res ; 129 Suppl 1: S22-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22682129

RESUMO

Oncogenic events impact interactions of cancer cells with their surroundings. Amongst the most consequential, in this regard, is the influence on angiogenesis, inflammation and hemostasis. Indeed, mutant oncogenes (EGFR, HER2, RAS, MET, PML-RARα) are known to alter the expression of angiogenic and pro-inflammatory factors, as well as change the cancer cell coagulome, including the levels of tissue factor (TF) and other mediators (PAI-1, COX2). Accompanying losses of tumour suppressor genes (PTEN, p53), and changes in microRNA (miR-19b, miR-520) facilitate these effects. Transforming genes may also trigger ectopic production of coagulation factors (e.g. FVII) by cancer cells and their release and properties of procoagulant microparticles (MPs). By deregulating protease activated receptors (PAR1/2) oncogenes may also change tumour cell responses to coagulation factor signalling. These changes act in concert with microenvironmental factors (hypoxia), stress responses (therapy) and differentiation programs, including epithelial-to-mesechymal transitions (EMT) and through tumour initiating cell (TIC) compartment. In so doing, the coagulation system influences early (initiation, angiogenesis), intermediate (growth, invasion) and late stages (metastasis, relapse) of cancer progression. In fact, TF may act as a molecular switch that controls the transition between dormant, latent and progressive/metastatic disease. TIC-like cells may play a role in these effects, as they express TF and PAR-1/2, and respond to stimulation with their agonists. As major human malignancies (e.g. glioblastoma) are increasingly recognized to consist of a spectrum of molecularly distinct disease subtypes driven by specific genetic pathways, so too may their patterns of interaction differ with the coagulation system. A better understanding of these linkages may be a source of new diagnostic, prognostic and therapeutic opportunities.


Assuntos
Fatores de Coagulação Sanguínea/genética , Fatores de Coagulação Sanguínea/imunologia , Hemostasia/genética , Hemostasia/imunologia , Neoplasias/genética , Neoplasias/imunologia , Animais , Humanos
15.
Semin Immunopathol ; 33(5): 455-67, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21318413

RESUMO

Cancer cells emit a heterogeneous mixture of vesicular, organelle-like structures (microvesicles, MVs) into their surroundings including blood and body fluids. MVs are generated via diverse biological mechanisms triggered by pathways involved in oncogenic transformation, microenvironmental stimulation, cellular activation, stress, or death. Vesiculation events occur either at the plasma membrane (ectosomes, shed vesicles) or within endosomal structures (exosomes). MVs are increasingly recognized as mediators of intercellular communication due to their capacity to merge with and transfer a repertoire of bioactive molecular content (cargo) to recipient cells. Such processes may occur both locally and systemically, contributing to the formation of microenvironmental fields and niches. The bioactive cargo of MVs may include growth factors and their receptors, proteases, adhesion molecules, signalling molecules, as well as DNA, mRNA, and microRNA (miRs) sequences. Tumour cells emit large quantities of MVs containing procoagulant, growth regulatory and oncogenic cargo (oncosomes), which can be transferred throughout the cancer cell population and to non-transformed stromal cells, endothelial cells and possibly to the inflammatory infiltrates (oncogenic field effect). These events likely impact tumour invasion, angiogenesis, metastasis, drug resistance, and cancer stem cell hierarchy. Ongoing studies explore the molecular mechanisms and mediators of MV-based intercellular communication (cancer vesiculome) with the hope of using this information as a possible source of therapeutic targets and disease biomarkers in cancer.


Assuntos
Comunicação Celular , Micropartículas Derivadas de Células/metabolismo , Exossomos/metabolismo , Neoplasias/metabolismo , Animais , Transporte Biológico , Exossomos/química , Humanos , Pesquisa Translacional Biomédica
16.
Thromb Res ; 125 Suppl 2: S44-50, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20434004

RESUMO

Oncogenic transformation and aberrant cellular differentiation are regarded as key processes leading to malignancy. They produce heterogenous cellular populations including subsets of tumour initiating cells (TICs), also known as cancer stem cells (CSCs). Intracellular events involved in these changes profoundly impact the extracellular and systemic constituents of cancer progression, including those dependent on the vascular system. This includes angiogenesis, vasculogenesis, activation of the coagulation system and formation of CSC-related and premetastatic niches. Tissue factor (TF) is a unique cell-associated receptor for coagulation factor VIIa, initiator of blood coagulation, and mediator of cellular signalling, all of which influence vascular homeostasis. Our studies established a link between oncogenic events, angiogenesis and the elevated expression of TF in several types of cancer cells. The latter suggests that cancer coagulopathy and cellular events attributed to the coagulation system may have cancer-specific and genetic causes. Indeed, in human glioma cells, a transforming mutant of the epidermal growth factor receptor (EGFRvIII) triggers not only the expression of TF, but also of its ligand (factor VII) and protease activated receptors (PAR-1 and PAR-2). Consequently, tumour cells expressing EGFRvIII become hypersensitive to contact with blood borne proteases (VIIa, thrombin), which upregulate their production of angiogenic factors (VEGF and IL-8), and contribute to formation of the growth promoting microenvironment (niche). Moreover, TF overexpression accompanies features of cellular aggressiveness such as markers of CSCs (CD133), epithelial-to-mesenchymal transition (EMT) and expression of the angiogenic and prometastatic phenotype. Conversely, TF blocking antibodies inhibit tumour growth, angiogenesis, and especially tumour initiation upon injection of threshold numbers of tumourigenic cells. Likewise, TF depletion in the host compartment (e.g. in low-TF mice) perturbs tumour initiation. These observations suggest that both cancer cells and their adjacent host stroma contribute TF activity to the tumour microenvironment. We postulate that the TF pathway may play an important role in formation of the vascular niche for tumour initiating CSCs, through its procoagulant and signalling effects. Therapeutic blockade of these mechanisms could hamper tumour initiation processes, which are dependent on CSCs and participate in tumour onset, recurrence, drug resistance and metastasis.


Assuntos
Neoplasias/metabolismo , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/patologia , Tromboplastina/metabolismo , Animais , Diferenciação Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/genética , Processos Neoplásicos , Células-Tronco Neoplásicas/metabolismo , Receptores Ativados por Proteinase/metabolismo , Tromboplastina/genética
17.
Best Pract Res Clin Haematol ; 22(1): 71-83, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19285274

RESUMO

The linkage between activation of the coagulation system and cancer is well established, as is deregulation of tissue factor (TF) by cancer cells, their vascular stroma and cancer-associated inflammatory cells. TF is no longer perceived as an 'alternative' coagulation factor, but rather as a central trigger of the coagulation cascade and an important cell-associated signalling receptor activated by factor VIIa, and interacting with several other regulatory entities, most notably protease-activated receptors (PAR-1 and PAR-2). Preclinical studies revealed the role of oncogenic transformation and tumour micro-environment as TF regulators in cancer, along with the impact of this receptor on gene expression, tumour growth, metastasis, angiogenesis and, possibly, formation of the cancer stem cell niche. Increasing interest surrounds the shedding of TF-containing microvesicles from cancer cells, their entry into the circulation and their role in the intercellular transfer of TF activity, cancer coagulopathy and other processes. Recent data also suggest differential roles of cell autonomous versus global effects of TF in various settings. Questions are raised regarding the consequences of TF expression by tumour cells themselves and by their associated host stroma. Progress in these areas may soon begin to impact on clinical practice and, as such, raises several important questions. Can TF be exploited as a therapeutic target in cancer? Where and when may this be safe and beneficial? Is expression of TF in various disease settings useful as a biomarker of cancer progression or the associated hypercoagulability? What clinical questions related to TF are especially worthy of further exploration, at present and in the near future? Some of these developments and questions will be discussed in this chapter.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Tromboplastina/metabolismo , Progressão da Doença , Humanos , Neoplasias/fisiopatologia
18.
Semin Thromb Hemost ; 34(2): 170-81, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18645922

RESUMO

Oncogenic upregulation of tissue factor (TF) and release of TF-containing microvesicles play an important role in cancer-related coagulopathy (Trousseau's syndrome), angiogenesis, and disease progression. In addition, certain types of host cells (stromal cells, inflammatory cells, activated endothelium) may also express TF. Although the relative contribution of host-related versus tumor-related TF to tumor progression is not known, our recent studies indicate that the role of both sources of TF in tumor formation is complex and context-dependent. Disruption of TF expression/activity in cancer cells leads to tumor growth inhibition in immunodeficient mice, even in cases where TF overexpression is driven by potent oncogenes ( K-RAS or EGFR). Interestingly, TF expression in vivo appears to be influenced by many factors, including the level of oncogenic transformation, tumor microenvironment, and differentiation from cancer stem-like cells. We postulate that activation of TF signaling and coagulation may deliver growth-promoting stimuli (e.g., fibrin, thrombin, platelets) to dormant cancer stem cells (CSCs). Functionally, these influences may be tantamount to formation of a provisional (TF-dependent) cancer stem cell niche. As such, these changes may contribute to the involvement of CSCs in tumor growth, angiogenesis, and metastasis.


Assuntos
Neoplasias/fisiopatologia , Tromboplastina/biossíntese , Transtornos da Coagulação Sanguínea/etiologia , Progressão da Doença , Neoplasias/patologia , Neovascularização Patológica/fisiopatologia , Oncogenes/fisiologia , Regulação para Cima
19.
Brain Inj ; 20(5): 469-75, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16716993

RESUMO

STUDY OBJECTIVE: To examine the efficacy of the Glasgow Coma Scale-Extended (GCS-E) for the prediction of symptoms commonly associated with mild traumatic brain injury (TBI). METHOD: Three hundred and sixty-one participants with a mild TBI were evaluated using the GCS-E and the Standardized Assessment of Concussion. A sub-group of 185 participants took part in a more extensive evaluation, which also included measures of depression and vestibular symptoms. All participants had a Glasgow Coma Scale score of 15, but experienced varying lengths of post-traumatic amnesia (PTA) as measured by the GCS-E. RESULTS: Use of the GCS-E for assessment of PTA duration revealed that longer lengths of amnesia following mild TBI were associated with greater incidence of dizziness, depression and cognitive impairments during the first weeks after injury. CONCLUSION: Results suggest that the GCS-E is a useful tool for the prediction of symptoms associated with mild TBI.


Assuntos
Amnésia/diagnóstico , Escala de Coma de Glasgow , Síndrome Pós-Concussão/diagnóstico , Adulto , Feminino , Humanos , Masculino , Militares , Valor Preditivo dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA