Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Thromb Res ; 164 Suppl 1: S136-S142, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29703472

RESUMEN

Molecular profiling of human cancers revealed a startling diversity in disease-causing mechanisms superseding histological and anatomical commonalities. The emerging molecular subtypes and disease entities are often driven by distinct oncogenic pathways and their effectors, including those acting extracellularly on the vascular and coagulation systems. Indeed, several oncogenic mutations such as those affecting protein-coding genes (RAS, EGFR, PTEN, TP53) and non-coding RNA (microRNA) regulate multiple effectors of the coagulation system (coagulome), including tissue factor, protease activated receptors, clotting factors, mediators of platelet function and fibrinolysis. This is exemplified by differential coagulome profiles in the molecular subtypes of glioblastoma, medulloblastoma and other human tumours. There is mounting clinical evidence that the mutational status of cancer driver genes such as KRAS or IDH1 may influence the risk of venous thromboembolism in patients with colorectal, lung or brain cancers. Notably, single cell sequencing in glioblastoma revealed a remarkable intra-tumoural heterogeneity of cancer cell populations with regard to their individual coagulomes, suggesting a combinatorial and dynamic nature of the global pro-thrombotic phenotype. We suggest that the cellular complexity of specific cancers may define their mechanisms of interactions with the coagulation system, and the risks of thrombosis. Thus, more biologically- based, disease-specific and personalized approaches may be needed to diagnose and manage cancer-related thrombosis.


Asunto(s)
Neoplasias Encefálicas/genética , Oncogenes/genética , Neoplasias Encefálicas/patología , Humanos , Fenotipo
2.
Biochem Biophys Res Commun ; 494(3-4): 700-705, 2017 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-29111325

RESUMEN

Tissue factor (TF) is a transmembrane receptor for coagulation factor VII/VIIa and is frequently overexpressed by cancer cells. The TF/VIIa complex acts as the main initiator of the clotting cascade in blood and a trigger of intracellular signaling that changes gene expression and the cellular phenotype. However, pathways mediating these changes are still poorly characterized and especially the impact of TF signals on regulatory microRNA (miR) networks in cancer remains unknown. We show that the monoclonal antibody that selectively neutralises the signaling (but not coagulant) function of human TF (CNTO 2559) inhibits progression of MDA-MB-231 breast cancer xenografts in mice and prolongs animal survival. CNTO 2559 blocks FVIIa-induced expression of interleukin 8 (IL-8) by cancer cells without impacting factor Xa (FXa) generation. Notably, acute exposure of MDA-MB-231 tumour xenografts to CNTO 2559 systemic injections triggers wide spread changes in the tumour miR profile including alterations in 75 miRs (55 downregulated) and impacting several miR-regulated and cancer-related pathways. These results suggest that TF signaling in the tumour microenvironment may provoke vast changes in the miR profile of cancer cells, affect disease biology, and reflect tumour interaction with the coagulation system, thereby presenting itself as a possible biomarker.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Tromboplastina/antagonistas & inhibidores , Tromboplastina/metabolismo , Microambiente Tumoral , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Redes Reguladoras de Genes , Humanos , Ratones , Transducción de Señal
3.
Semin Cell Dev Biol ; 67: 11-22, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28077296

RESUMEN

Pathogenesis of human cancers bridges intracellular oncogenic driver events and their impact on intercellular communication. Among multiple mediators of this 'pathological connectivity' the role of extracellular vesicles (EVs) and their subsets (exosomes, ectosomes, oncosomes) is of particular interest for several reasons. The release of EVs from cancer cells represents a unique mechanism of regulated expulsion of bioactive molecules, a process that also mediates cell-to-cell transfer of lipids, proteins, and nucleic acids. Biological effects of these processes have been implicated in several aspects of cancer-related pathology, including tumour growth, invasion, angiogenesis, metastasis, immunity and thrombosis. Notably, the emerging evidence suggests that oncogenic mutations may impact several aspects of EV-mediated cell-cell communication including: (i) EV release rate and protein content; (ii) molecular composition of cancer EVs; (iii) the inclusion of oncogenic and mutant macromolecules in the EV cargo; (iv) EV-mediated release of genomic DNA; (v) deregulation of mechanisms responsible for EV biogenesis (vesiculome) and (vi) mechanisms of EV uptake by cancer cells. Intriguingly, EV-mediated intercellular transfer of mutant and oncogenic molecules between subpopulations of cancer cells, their indolent counterparts and stroma may exert profound biological effects that often resemble (but are not tantamount to) oncogenic transformation, including changes in cell growth, clonogenicity and angiogenic phenotype, or cause cell stress and death. However, several biological barriers likely curtail a permanent horizontal transformation of normal cells through EV-mediated mechanisms. The ongoing analysis and targeting of EV-mediated intercellular communication pathways can be viewed as a new therapeutic paradigm in cancer, while the analysis of oncogenic cargo contained in EVs released from cancer cells into biofluids is being developed for clinical use as a biomarker and companion diagnostics. Indeed, studies are underway to further explore the multiple links between molecular causality in cancer and various aspects of cellular vesiculation.


Asunto(s)
Transformación Celular Neoplásica/genética , Vesículas Extracelulares/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/genética , Neoplasias/metabolismo , Neovascularización Patológica/genética , Antineoplásicos/uso terapéutico , Transporte Biológico/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Progresión de la Enfermedad , Vesículas Extracelulares/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metástasis Linfática , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Patológica/prevención & control , Oncogenes
4.
Methods Mol Biol ; 1458: 39-58, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27581013

RESUMEN

Angiogenesis represents one aspect in the complex process that leads to the generation of the vascular tumor stroma. The related functional constituents include responses of endothelial, mural, bone marrow-derived, and resident inflammatory cells as well as activation of coagulation and fibrinolytic systems in blood. Multiple molecular and cellular effectors participate in these events, often in a tumor-specific manner and with changes enforced through the microenvironment, genetic evolution, and responses to anticancer therapies. To capture various elements of these interactions several surrogate assays have been devised, which can be mechanistically useful and are amenable to quantification, but are individually insufficient to describe the underlying complexity and are best used in a targeted and combinatorial manner. Below, we present a survey of angiogenesis assays and experimental approaches to analyze vascular events in cancer. We also provided specific examples of validated protocols, which are less described, but enable the straightforward analysis of vascular structures and coagulant properties of cancer cells in vivo and in vitro.


Asunto(s)
Coagulación Sanguínea , Neoplasias/sangre , Neoplasias/patología , Neovascularización Patológica , Microambiente Tumoral , Animales , Biomarcadores , Células Endoteliales/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Neoplasias/metabolismo , Tromboplastina/metabolismo
5.
Oncotarget ; 7(32): 51991-52002, 2016 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-27437771

RESUMEN

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.


Asunto(s)
Comunicación Celular/fisiología , Transformación Celular Neoplásica/genética , Vesículas Extracelulares/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Vesículas Extracelulares/genética , Vesículas Extracelulares/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Xenoinjertos , Humanos , Ratones , Ratones SCID , Proteínas Proto-Oncogénicas p21(ras)/genética
6.
Thromb Res ; 140 Suppl 1: S37-43, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27067976

RESUMEN

Activation of stromal response pathways in cancer is increasingly viewed as both a local and systemic extension of molecular alterations driving malignant transformation. Rather than reflecting passive and unspecific responses to anatomical abnormalities, the coagulation system is a target of oncogenic deregulation, impacting the role of clotting and fibrinolytic proteins, and integrating hemostasis, inflammation, angiogenesis and cellular growth effects in cancer. These processes signify, but do not depend on, the clinically manifest coagulopathy and thrombosis. In this regard, the role of driver mutations affecting oncoprotein coding genes such as RAS, EGFR or MET and tumour suppressors (PTEN, TP53) are well described as regulators of tissue factor (TF), protease activated receptors (PAR-1/2) and ectopic coagulation factors (FVII). Indeed, in both adult and pediatric brain tumours the expression patterns of coagulation and angiogenesis regulators (coagulome and angiome, respectively) reflect the molecular subtypes of the underlying diseases (glioblastoma or medulloblastoma) as defined by their oncogenic classifiers and clinical course. This emerging understanding is still poorly established in relation to the transforming effects of non-coding genes, including those responsible for the expression of microRNA (miR). Indeed, several miRs have been recently found to regulate TF and other effectors. We recently documented that in the context of the aggressive embryonal tumour with multilayered rosettes (ETMR) the oncogenic driver miR (miR-520g) suppresses the expression of TF and correlates with hypocoagulant tumour characteristics. Unlike in adult cancers, the growth of pediatric embryonal brain tumour cells as spheres (to maintain stem cell properties) results in upregulation of miR-520g and downregulation of TF expression and activity. We postulate that oncogenic protein and miR coding genes form alternative pathways of coagulation system regulation in different tumour settings, a property necessitating more personalised and biologically-based approaches to anticoagulation.


Asunto(s)
Anticoagulantes/uso terapéutico , Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Coagulación Sanguínea/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Meduloblastoma/tratamiento farmacológico , Animales , Anticoagulantes/farmacología , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/complicaciones , Trastornos de la Coagulación Sanguínea/genética , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/genética , Glioblastoma/sangre , Glioblastoma/complicaciones , Glioblastoma/genética , Humanos , Meduloblastoma/sangre , Meduloblastoma/complicaciones , Meduloblastoma/genética , MicroARNs/genética , MicroARNs/metabolismo , Oncogenes/efectos de los fármacos , Medicina de Precisión/métodos , Tromboplastina/genética , Tromboplastina/metabolismo
7.
Cell Mol Neurobiol ; 36(3): 383-407, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26993504

RESUMEN

Brain tumors can be viewed as multicellular 'ecosystems' with increasingly recognized cellular complexity and systemic impact. While the emerging diversity of malignant disease entities affecting brain tissues is often described in reference to their signature alterations within the cellular genome and epigenome, arguably these cell-intrinsic changes can be regarded as hardwired adaptations to a variety of cell-extrinsic microenvironmental circumstances. Conversely, oncogenic events influence the microenvironment through their impact on the cellular secretome, including emission of membranous structures known as extracellular vesicles (EVs). EVs serve as unique carriers of bioactive lipids, secretable and non-secretable proteins, mRNA, non-coding RNA, and DNA and constitute pathway(s) of extracellular exit of molecules into the intercellular space, biofluids, and blood. EVs are also highly heterogeneous as reflected in their nomenclature (exosomes, microvesicles, microparticles) attempting to capture their diverse origin, as well as structural, molecular, and functional properties. While EVs may act as a mechanism of molecular expulsion, their non-random uptake by heterologous cellular recipients defines their unique roles in the intercellular communication, horizontal molecular transfer, and biological activity. In the central nervous system, EVs have been implicated as mediators of homeostasis and repair, while in cancer they may act as regulators of cell growth, clonogenicity, angiogenesis, thrombosis, and reciprocal tumor-stromal interactions. EVs produced by specific brain tumor cell types may contain the corresponding oncogenic drivers, such as epidermal growth factor receptor variant III (EGFRvIII) in glioblastoma (and hence are often referred to as 'oncosomes'). Through this mechanism, mutant oncoproteins and nucleic acids may be transferred horizontally between cellular populations altering their individual and collective phenotypes. Oncogenic pathways also impact the emission rates, types, cargo, and biogenesis of EVs, as reflected by preliminary analyses pointing to differences in profiles of EV-regulating genes (vesiculome) between molecular subtypes of glioblastoma, and in other brain tumors. Molecular regulators of vesiculation can also act as oncogenes. These intimate connections suggest the context-specific roles of different EV subsets in the progression of specific brain tumors. Advanced efforts are underway to capture these events through the use of EVs circulating in biofluids as biomarker reservoirs and to guide diagnostic and therapeutic decisions.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Progresión de la Enfermedad , Vesículas Extracelulares/metabolismo , Animales , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Comunicación Celular , Transformación Celular Neoplásica/patología , Humanos , Mutación/genética
8.
Am J Pathol ; 186(2): 446-59, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26687818

RESUMEN

Pediatric embryonal brain tumors with multilayered rosettes demonstrate a unique oncogenic amplification of the chromosome 19 miRNA cluster, C19MC. Because oncogenic lesions often cause deregulation of vascular effectors, including procoagulant tissue factor (TF), this study explores whether there is a link between C19MC oncogenic miRNAs (oncomirs) and the coagulant properties of cancer cells, a question previously not studied. In a pediatric embryonal brain tumor tissue microarray, we observed an association between C19MC amplification and reduced fibrin content and TF expression, indicative of reduced procoagulant activity. In medulloblastoma cell lines (DAOY and UW228) engineered to express miR-520g, a biologically active constituent of the C19MC cluster, we observed reduced TF expression, procoagulant and TF signaling activities (responses to factor VIIa stimulation), and diminished TF emission as cargo of extracellular vesicles. Antimir and luciferase reporter assays revealed a specific and direct effect of miR-520g on the TF 3' untranslated region. Although the endogenous MIR520G locus is methylated in differentiated cells, exposure of DAOY cells to 5-aza-2'-deoxycytidine or their growth as stem cell-like spheres up-regulated endogenous miR-520g with a coincident reduction in TF expression. We propose that the properties of tumors harboring oncomirs may include unique alterations of the vascular microenvironment, including deregulation of TF, with a possible impact on the biology, therapy, and hemostatic adverse effects of both disease progression and treatment.


Asunto(s)
Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/genética , Microambiente Tumoral/genética , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Diferenciación Celular/genética , Humanos , Meduloblastoma/genética , Transducción de Señal/genética , Células Madre/citología , Tromboplastina/metabolismo
9.
Angiogenesis ; 19(1): 25-38, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26374632

RESUMEN

Oncogenic transformation is believed to impact the vascular phenotype and microenvironment in cancer, at least in part, through mechanisms involving extracellular vesicles (EVs). We explored these questions in the context of acute promyelocytic leukemia cells (NB4) expressing oncogenic fusion protein, PML-RARa and exquisitely sensitive to its clinically used antagonist, the all-trans retinoic acid (ATRA). We report that NB4 cells produce considerable numbers of EVs, which are readily taken up by cultured endothelial cells triggering their increased survival. NB4 EVs contain PML-RARa transcript, but no detectable protein, which is also absent in endothelial cells upon the vesicle uptake, thereby precluding an active intercellular trafficking of this oncogene in this setting. ATRA treatment changes the emission profile of NB4-related EVs resulting in preponderance of smaller vesicles, an effect that occurs in parallel with the onset of cellular differentiation. ATRA also increases IL-8 mRNA and protein content in NB4 cells and their EVs, while decreasing the levels of VEGF and tissue factor (TF). Endothelial cell uptake of NB4-derived EVs renders these cells more TF-positive and procoagulant, and this effect is diminished by pre-treatment of EV donor cells with ATRA. Profiling angiogenesis-related transcripts in intact and ATRA-treated APL cells and their EVs reveals multiple differences attributable to cellular responses and EV molecular packaging. These observations point to the potential significance of changes in the angiogenic signature and activity associated with EVs released from tumor cells subjected to targeted therapy.


Asunto(s)
Vesículas Extracelulares/patología , Leucemia Promielocítica Aguda/patología , Neovascularización Patológica/patología , Proteínas de Fusión Oncogénica/metabolismo , Coagulación Sanguínea/efectos de los fármacos , Línea Celular Tumoral , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestructura , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Neovascularización Patológica/genética , Proteínas de Fusión Oncogénica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tromboplastina/metabolismo , Tretinoina/farmacología
10.
Rambam Maimonides Med J ; 5(4): e0030, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25386346

RESUMEN

The coagulation system constitutes an important facet of the unique vascular microenvironment in which primary and metastatic brain tumors evolve and progress. While brain tumor cells express tissue factor (TF) and other effectors of the coagulation system (coagulome), their propensity to induce local and peripheral thrombosis is highly diverse, most dramatic in the case of glioblastoma multiforme (GBM), and less obvious in pediatric tumors. While the immediate medical needs often frame the discussion on current clinical challenges, the coagulation pathway may contribute to brain tumor progression through subtle, context-dependent, and non-coagulant effects, such as induction of inflammation, angiogenesis, or by responding to iatrogenic insults (e.g. surgery). In this regard, the emerging molecular diversity of brain tumor suptypes (e.g. in glioma and medulloblastoma) highlights the link between oncogenic pathways and the tumor repertoire of coagulation system regulators (coagulome). This relationship may influence the mechanisms of spontaneous and therapeutically provoked tumor cell interactions with the coagulation system as a whole. Indeed, oncogenes (EGFR, MET) and tumor suppressors (PTEN, TP53) may alter the expression, activity, and vesicular release of tissue factor (TF), and cause other changes. Conversely, the coagulant microenvironment may also influence the molecular evolution of brain tumor cells through selective and instructive cues. We suggest that effective targeting of the coagulation system in brain tumors should be explored through molecular stratification, stage-specific analysis, and more personalized approaches including thromboprophylaxis and adjuvant treatment aimed at improvement of patient survival.

11.
Mech Ageing Dev ; 140: 13-22, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25068886

RESUMEN

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.


Asunto(s)
Envejecimiento/fisiología , Inhibidores de la Angiogénesis/uso terapéutico , Aterosclerosis/complicaciones , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Indoles/uso terapéutico , Pirroles/uso terapéutico , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Carcinoma Pulmonar de Lewis/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Sunitinib , Resultado del Tratamiento
12.
Thromb Res ; 133 Suppl 2: S1-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24862126

RESUMEN

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.


Asunto(s)
Coagulación Sanguínea/genética , Neoplasias Encefálicas , Glioma , Oncogenes/fisiología , Animales , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Cerebelosas/sangre , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Glioma/sangre , Glioma/genética , Glioma/secundario , Humanos , Meduloblastoma/sangre , Meduloblastoma/genética , Meduloblastoma/secundario , Transcriptoma , Microambiente Tumoral/genética
13.
Semin Thromb Hemost ; 40(3): 284-95, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24599437

RESUMEN

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.


Asunto(s)
Neoplasias/complicaciones , Neoplasias/genética , Trombosis/complicaciones , Trombosis/genética , Animales , Humanos , Neoplasias/sangre
14.
Semin Thromb Hemost ; 39(8): 881-95, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24108471

RESUMEN

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.


Asunto(s)
Coagulación Sanguínea , Neoplasias Encefálicas/irrigación sanguínea , Glioblastoma/irrigación sanguínea , Neovascularización Patológica/patología , Adulto , Niño , Humanos , Modelos Biológicos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Transducción de Señal , Tromboplastina/metabolismo
15.
Front Physiol ; 3: 294, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22934045

RESUMEN

The brain is a frequent site of neoplastic growth, including both primary and metastatic tumors. The clinical intractability of many brain tumors and their distinct biology are implicitly linked to the unique microenvironment of the central nervous system (CNS) and cellular interactions within. Among the most intriguing forms of cellular interactions is that mediated by membrane-derived extracellular vesicles (EVs). Their biogenesis (vesiculation) and uptake by recipient cells serves as a unique mechanism of intercellular trafficking of complex biological messages including the exchange of molecules that cannot be released through classical secretory pathways, or that are prone to extracellular degradation. Tumor cells produce EVs containing molecular effectors of several cancer-related processes such as growth, invasion, drug resistance, angiogenesis, and coagulopathy. Notably, tumor-derived EVs (oncosomes) also contain oncogenic proteins, transcripts, DNA, and microRNA (miR). Uptake of this material may change properties of the recipient cells and impact the tumor microenvironment. Examples of transformation-related molecules found in the cargo of tumor-derived EVs include the oncogenic epidermal growth factor receptor (EGFRvIII), tumor suppressors (PTEN), and oncomirs (miR-520g). It is postulated that EVs circulating in blood or cerebrospinal fluid (CSF) of brain tumor patients may be used to decipher molecular features (mutations) of the underlying malignancy, reflect responses to therapy, or molecular subtypes of primary brain tumors [e.g., glioma or medulloblastoma (MB)]. It is possible that metastases to the brain may also emit EVs with clinically relevant oncogenic signatures. Thus, EVs emerge as a novel and functionally important vehicle of intercellular communication that can mediate multiple biological effects. In addition, they provide a unique platform to develop molecular biomarkers in brain malignancies.

16.
Thromb Res ; 129 Suppl 1: S22-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22682129

RESUMEN

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.


Asunto(s)
Factores de Coagulación Sanguínea/genética , Factores de Coagulación Sanguínea/inmunología , Hemostasis/genética , Hemostasis/inmunología , Neoplasias/genética , Neoplasias/inmunología , Animales , Humanos
17.
Semin Immunopathol ; 33(5): 455-67, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21318413

RESUMEN

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.


Asunto(s)
Comunicación Celular , Micropartículas Derivadas de Células/metabolismo , Exosomas/metabolismo , Neoplasias/metabolismo , Animales , Transporte Biológico , Exosomas/química , Humanos , Investigación Biomédica Traslacional
18.
Endocrinology ; 151(4): 1685-94, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20160134

RESUMEN

Endocannabinoids (eCBs) are products of phospholipid (PL)-derived arachidonic acid (AA) that regulate hypothalamus-pituitary-adrenal axis activity. We hypothesized that differences in the quality and quantity of maternal dietary fat would modulate the PL AA content in the neonatal brain affecting stress responsiveness via differences in eCB production and activity in stress-activated brain areas. Pregnant rats were fed a 5% [control (C)] or 30% fat [high fat (HF)] diet rich in either n-6 (HF-n-6) or n-3 (HF-n-3) fat during the last week of gestation and lactation. Postnatal d 10 offspring were tested for metabolic hormones, AA (n-6) and eCB brain content, and hormonal effects of eCB receptor antagonism (AM251, 1 or 3 mg/kg ip) on stress responses. Like maternal diet, milk from HF-n-3 mothers had a reduced n-6/n-3 fat ratio compared with that of C and HF-n-6 mothers. Hypothalamic and hippocampal levels of PL AA were diet specific, reflecting the maternal milk and dietary n-6/n-3 ratio, with HF-n-3 offspring displaying reduced AA content relative to C and HF-n-6 offspring. Plasma corticosterone and insulin were elevated in HF-fed pups, whereas leptin was increased only in HF-n-6 pups. Basal eCB concentrations were also diet and brain region specific. In C pups, eCB receptor antagonist pretreatment increased stress-induced ACTH secretion, but not in the HF groups. Stress-induced corticosterone secretion was not sensitive to AM251 treatment in HF-n-3 pups. Thus, the nature of preweaning dietary fat differentially influences neonatal metabolic hormones, brain PL AA levels, and eCB, with functional consequences on hypothalamus-pituitary-adrenal axis modulation in developing rat pups.


Asunto(s)
Moduladores de Receptores de Cannabinoides/metabolismo , Grasas de la Dieta/metabolismo , Endocannabinoides , Efectos Tardíos de la Exposición Prenatal/metabolismo , Fenómenos Fisiologicos de la Nutrición Prenatal/fisiología , Estrés Fisiológico/fisiología , Tejido Adiposo/metabolismo , Hormona Adrenocorticotrópica/sangre , Análisis de Varianza , Animales , Animales Lactantes , Peso Corporal/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Química Encefálica , Moduladores de Receptores de Cannabinoides/análisis , Corticosterona/sangre , Grasas de la Dieta/administración & dosificación , Ácidos Grasos/análisis , Ácidos Grasos/metabolismo , Femenino , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Insulina/sangre , Lactancia/metabolismo , Leptina/metabolismo , Piperidinas/farmacología , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Embarazo , Pirazoles/farmacología , ARN Mensajero/metabolismo , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Análisis de Regresión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Fisiológico/efectos de los fármacos
19.
Ann N Y Acad Sci ; 1144: 189-202, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19076377

RESUMEN

Both undernutrition and overnutrition of the mother during pregnancy and lactation produce a syndrome of altered energy balance in the offspring and has long-lasting consequences on CNS systems regulating food intake, metabolism, and food reward. Homeostatic circulating factors like insulin, glucocorticoids, and leptin that are generally increased by exposure to high fat/high caloric diets constitute important signals in these processes. They trigger functional activation of specific intracellular cascades mediating cellular sensitivity, survival, and synaptic plasticity. Using a model whereby the late fetal and neonatal rat is exposed to increased high fat (HF) via HF feeding of the mother, we investigated the proximal (neonatal) and distal (adult) consequences on metabolism and hippocampal function in the offspring. Adult offspring of HF-fed mothers displayed several of the physiological and behavioral changes susceptible to leading to metabolic complications. These include elevated circulating concentrations of leptin and corticosterone, increased body weight gain and food intake, modest preference for fat-containing food types, as well as the onset of hypothalamic leptin resistance. In the hippocampus, HF-fed offspring or neonates treated with leptin show similar increases in neurogenesis and survival of newborn neurons. We identified some of the direct effects of leptin to increase synaptic proteins, N-methyl-d-aspartate (NMDA), and glucocorticoid receptors, and to reduce long-term potentiation (LTP) prior to weaning. While these studies have documented effects in animal models, concepts can easily be translated to human nutrition in order to help design better perinatal diets and nutritional preventive measures for mothers in a coordinated effort to curb the obesity trend.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Hipocampo/metabolismo , Leptina/metabolismo , Animales , Femenino , Hipocampo/efectos de los fármacos , Leptina/farmacología , Exposición Materna , Modelos Biológicos , N-Metilaspartato/metabolismo , Embarazo , Ratas , Receptores de Glucocorticoides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...