Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Anticancer Res ; 44(8): 3269-3276, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39060075

RESUMEN

BACKGROUND/AIM: How tumors regulate the genes of the coagulome is crucial for cancer-associated thrombosis and the occurrence of venous thromboembolic complications in patients with cancer. We have previously reported potent yet complex effects of glucocorticoids (GC) on the expression of three genes that play a key role in the regulation of thrombin/plasmin activation (F3, PLAU, and SERPINE1). This study aimed to extend the investigation of GC effects to the whole tumor coagulome and assess the resulting impact on the ability of cancer cells to activate thrombin and plasmin. MATERIALS AND METHODS: Cancer RNA expression data were retrieved from various sources. Additionally, oral squamous cell carcinoma (OSCC) cells exposed to GC in vitro were analyzed using QPCR, enzymatic assays measuring thrombin and urokinase-type Plasminogen Activator (uPA) activity, and D-dimer concentrations. RESULTS: Our findings highlight the potent and specific stimulatory effect of GC on SERPINE1 expression across different types of cancer. Consistently, GC were found to inhibit uPA proteolytic activity and reduce the concentrations of D-dimers in OSCC in vitro. CONCLUSION: Fibrinolysis inhibition is a key consequence of cancer cell exposure to GC, possibly leading to the stabilization of the fibrin clot in cancer.


Asunto(s)
Fibrinólisis , Glucocorticoides , Inhibidor 1 de Activador Plasminogénico , Humanos , Inhibidor 1 de Activador Plasminogénico/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Fibrinólisis/efectos de los fármacos , Glucocorticoides/farmacología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/genética , Trombina/metabolismo , Trombina/farmacología , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Activación Transcripcional/efectos de los fármacos , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias/genética , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Coagulación Sanguínea/efectos de los fármacos
2.
Discov Oncol ; 15(1): 256, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38955935

RESUMEN

Cancer-associated thrombosis is a significant complication in cancer patients, leading to increased morbidity and mortality. The expression of coagulation/fibrinolysis genes, termed the "coagulome", plays a critical role in this process. Using the single-sample gene set enrichment analysis (ssGSEA), we identified seven cancer types with significantly activated coagulation pathways, focusing on lower-grade glioma (LGG) and stomach adenocarcinoma due to their predictive value for overall survival. Through 1000 iterations of the Least Absolute Shrinkage and Selection Operator (LASSO), we selected prognostic genes and constructed effective Cox regression models, particularly for LGG. Incorporating clinical characteristics, we constructed a nomogram for LGG, achieving an impressive area under the curve (AUCs) of 0.79, 0.82, and 0.81 at 1, 3, and 5 years in the test dataset, indicating strong potential for clinical application. Functional enrichment analysis between high-risk and low-risk LGG groups revealed significant enrichment of genes involved in the inflammatory response, interferon-gamma response, and epithelial-mesenchymal transition pathways. Combined with CIBERSORT and single-cell RNA sequencing analysis of LGG, our results demonstrated that the interplay between coagulation and the tumor microenvironment, particularly involving gliomas and myeloid cells, significantly influences tumor progression and patient outcomes.

3.
Adv Drug Deliv Rev ; 200: 115027, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37517779

RESUMEN

The rising incidence and persistent thrombosis in multiple cancers including those that are immunosuppressive highlight the need for understanding the tumor coagulome system and its role beyond hemostatic complications. Immunotherapy has shown significant benefits in solid organ tumors but has been disappointing in the treatment of hypercoagulable cancers, such as glioblastoma and pancreatic ductal adenocarcinomas. Thus, targeting thrombosis to prevent immunosuppression seems a clinically viable approach in cancer treatment. Hypercoagulable tumors often develop fibrin clots within the tumor microenvironment (TME) that dictates the biophysical characteristics of the tumor tissue. The application of systems biology and single-cell approaches highlight the potential role of coagulome or thrombocytosis in shaping the tumor immune microenvironment (TIME). In-depth knowledge of the tumor coagulome would provide unprecedented opportunities to better predict the hemostatic complications, explore how thrombotic stroma modulates tumor immunity, reexamine the significance of clinical biomarkers, and enable steering the stromal versus systemic immune response for boosting the effectiveness of immune checkpoint inhibitors in cancer treatment. We focus on the role of coagulation factors in priming a suppressive TIME and the huge potential of existing anticoagulant drugs in the clinical settings of cancer immunotherapy.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Microambiente Tumoral , Inmunoterapia/efectos adversos , Neoplasias Pancreáticas/patología , Terapia de Inmunosupresión/efectos adversos , Carcinoma Ductal Pancreático/patología
4.
Cancers (Basel) ; 15(5)2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36900323

RESUMEN

BACKGROUND: The coagulome, defined as the repertoire of genes that locally regulate coagulation and fibrinolysis, is a key determinant of vascular thromboembolic complications of cancer. In addition to vascular complications, the coagulome may also regulate the tumor microenvironment (TME). Glucocorticoids are key hormones that mediate cellular responses to various stresses and exert anti-inflammatory effects. We addressed the effects of glucocorticoids on the coagulome of human tumors by investigating interactions with Oral Squamous Cell Carcinoma, Lung Adenocarcinoma, and Pancreatic Adenocarcinoma tumor types. METHODS: We analyzed the regulation of three essential coagulome components, i.e., the tissue factor (TF), urokinase-type plasminogen activator (uPA), and plasminogen activator inhibitor-1 (PAI-1) in cancer cell lines exposed to specific agonists of the glucocorticoid receptor (GR) (dexamethasone and hydrocortisone). We used QPCR, immunoblots, small-interfering RNA, Chromatin immunoprecipitation sequencing (ChIPseq) and genomic data from whole tumor and single-cell analyses. RESULTS: Glucocorticoids modulate the coagulome of cancer cells through a combination of indirect and direct transcriptional effects. Dexamethasone directly increased PAI-1 expression in a GR-dependent manner. We confirmed the relevance of these findings in human tumors, where high GR activity/high SERPINE1 expression corresponded to a TME enriched in active fibroblasts and with a high TGF-ß response. CONCLUSION: The transcriptional regulation of the coagulome by glucocorticoids that we report may have vascular consequences and account for some of the effects of glucocorticoids on the TME.

5.
Front Med (Lausanne) ; 10: 1252417, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38188342

RESUMEN

Single cell analysis of cancer cell transcriptome may shed a completely new light on cancer-associated thrombosis (CAT). CAT causes morbid, and sometimes lethal complications in certain human cancers known to be associated with high risk of venous thromboembolism (VTE), pulmonary embolism (PE) or arterial thromboembolism (ATE), all of which worsen patients' prognosis. How active cancers drive these processes has long evaded scrutiny. While "unspecific" microenvironmental effects and consequences of patient care (e.g., chemotherapy) have been implicated in pathogenesis of CAT, it has also been suggested that oncogenic pathways driven by either genetic (mutations), or epigenetic (methylation) events may influence the coagulant phenotype of cancer cells and stroma, and thereby modulate the VTE/PE risk. Consequently, the spectrum of driver events and their downstream effector mechanisms may, to some extent, explain the heterogeneity of CAT manifestations between cancer types, molecular subtypes, and individual cases, with thrombosis-promoting, or -protective mutations. Understanding this molecular causation is important if rationally designed countermeasures were to be deployed to mitigate the clinical impact of CAT in individual cancer patients. In this regard, multi-omic analysis of human cancers, especially at a single cell level, has brought a new meaning to concepts of cellular heterogeneity, plasticity, and multicellular complexity of the tumour microenvironment, with profound and still relatively unexplored implications for the pathogenesis of CAT. Indeed, cancers may contain molecularly distinct cellular subpopulations, or dynamic epigenetic states associated with different profiles of coagulant activity. In this article we discuss some of the relevant lessons from the single cell "omics" and how they could unlock new potential mechanisms through which cancer driving oncogenic lesions may modulate CAT, with possible consequences for patient stratification, care, and outcomes.

6.
Best Pract Res Clin Haematol ; 35(1): 101349, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-36030072

RESUMEN

Cancer associated thrombosis (CAT) including venous and arterial thromboembolism (VTE and ATE respectively), as well as subclinical hypercoagulable states pose a risk of serious morbidity and mortality and poor outcomes in cancer patients. It is increasingly clear that rather than being unspecific aftermaths of tumour growth, CAT is causally linked to the molecular phenotype of cancer cells and its genetic and epigenetic oncogenic drivers. Emerging data suggest that mutational events and factors modifying chromatin architecture in cancer cells influence the repertoire of genes (coagulome) the products of which may interact with the hemostatic system either directly or through modification of inflammatory system or release of cancer-related prothrombotic extracellular vesicles (EVs). Single cell transcriptomic analysis of brain tumours reveals the coexistence of multiple coagulant mechanisms associated with different cancer cell subpopulations and sites. These observations may suggest that a multipronged, biologically based approach may be needed to effectively predict and manage CAT.


Asunto(s)
Coagulación Sanguínea , Neoplasias , Oncogenes , Trombosis , Epigenómica , Humanos , Neoplasias/complicaciones , Trombosis/etiología
7.
Oncoimmunology ; 11(1): 2045696, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35251772

RESUMEN

Besides a number of physical consequences (reduced blood supply, stabilization of circulating tumor microemboli, shielding from the attack of immune cells), the coagulation cascade may specifically regulate antitumor immunity. We recently applied systems biology and genomics to explore the regulation of the tumor immune microenvironment by coagulation.


Asunto(s)
Neoplasias , Microambiente Tumoral , Coagulación Sanguínea , Humanos
8.
Cancers (Basel) ; 14(2)2022 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-35053621

RESUMEN

BACKGROUND: Hemostatic complications, ranging from thromboembolism to bleeding, are a significant source of morbidity and mortality in cancer patients. The tumor coagulome represents the multiple genes and proteins that locally contribute to the equilibrium between coagulation and fibrinolysis. We aimed to study the coagulome of Oral Squamous Cell Carcinoma (OSCC) and examine its link to the tumor microenvironment (TME). METHODS: We used data from bulk tumor DNA/RNA-seq (The Cancer Genome Atlas), single-cell RNA-seq data and OSCC cells in culture. RESULTS: Among all tumor types, OSCC was identified as the tumor with the highest mRNA expression levels of F3 (Tissue Factor, TF) and PLAU (urokinase type-plasminogen activator, uPA). Great inter- and intra-tumor heterogeneity were observed. Single-cell analyses showed the coexistence of subpopulations of pro-coagulant and pro-fibrinolytic cancer cells within individual tumors. Interestingly, OSCC with high F3 expressed higher levels of the key immune checkpoint molecules CD274/PD-L1, PDCD1LG2/PD-L2 and CD80, especially in tumor dendritic cells. In vitro studies confirmed the particularity of the OSCC coagulome and suggested that thrombin exerts indirect effects on OSCC cells. CONCLUSIONS: OSCC presents a specific coagulome. Further studies examining a possible negative modulation of the tumor's adaptive immune response by the coagulation process are warranted.

9.
Trends Cancer ; 8(5): 369-383, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35027336

RESUMEN

Human tumors often trigger a hypercoagulable state that promotes hemostatic complications, including venous thromboembolism. The recent application of systems biology to the study of the coagulome highlighted its link to shaping the tumor microenvironment (TME), both within and outside of the vascular space. Addressing this link provides the opportunity to revisit the significance of biomarkers of hemostasis and assess the communication between vasculature and tumor parenchyma, an important topic considering the advent of immune checkpoint inhibitors and vascular normalization strategies. Understanding how the coagulome and TME influence each other offers exciting new prospects for predicting hemostatic complications and boosting the effectiveness of cancer treatment.


Asunto(s)
Hemostáticos , Neoplasias , Biomarcadores , Humanos , Microambiente Tumoral
10.
Cancer Immunol Immunother ; 70(4): 923-933, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33057845

RESUMEN

OBJECTIVE: Solid tumors often establish a procoagulable state that can lead to venous thromboembolism (VTE). Although some of the key genes involved in this process are known, no previous study has compared the "coagulome", i.e., the expression of coagulation/fibrinolysis genes, across different primary tumor types. It is also unclear whether the coagulome is associated with specific characteristics of the tumor microenvironment (TME). We aimed to address this question. METHODS: We analyzed the expression of the genes F3, PLAU, PLAT, PLAUR, SERPINB2, and SERPINE1 in 32 cancer types using data from The Cancer Genome Atlas (TCGA) and other freely available resources. RESULTS: We identified specific expression patterns of procoagulant and fibrinolytic genes. The expression of the Tissue Factor (F3) was found to be tumor type dependent, with the highest expression in glioblastoma (GBM), a highly procoagulable tumor type. Conversely, high expression of the fibrinolysis gene cluster PLAU, PLAUR, SERPINE1 was consistently linked to the characteristics of the TME (monocytic infiltration) and high expression of important checkpoints of the immune response, such as PD-L2 and CD276/B7-H3. CONCLUSION: These tumor-specific patterns of expression might partially explain the differences in VTE risk among tumor types. We propose that biomarkers of coagulation fibrinolysis might provide valuable information about the TME in cancer patients.


Asunto(s)
Biomarcadores de Tumor/genética , Coagulación Sanguínea/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Neovascularización Patológica/genética , Transcriptoma , Microambiente Tumoral/genética , Perfilación de la Expresión Génica , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/inmunología , Neovascularización Patológica/inmunología , Microambiente Tumoral/inmunología
11.
Thromb Res ; 191 Suppl 1: S99-S105, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32736787

RESUMEN

Cancer-associated thrombosis (CAT) is a morbid, potentially life threatening and biologically impactful paraneoplastic state. At least in part, CAT is likely driven by cancer-specific mechanisms the nature of which is still poorly understood, hampering diagnostic, prophylactic and therapeutic efforts. It is increasingly appreciated that cancer-specific drivers of CAT include a constellation of oncogenic mutations and their superimposed epigenetic states that shape the transcriptome, phenotype and secretome of cancer cell populations, including the repertoire of genes impacting the vascular and coagulation systems. High-grade brain tumours, such as glioblastoma multiforme (GBM) represent a paradigm of locally initiated haemostatic abnormalities that propagate systemically, likely through circulating mediators, such as extracellular vesicles and soluble factors. Reciprocally, CAT impacts the biology of cancer cells and may drive tumour evolution. The constituent, oncogene-transformed cancer cell populations form complex ecosystems, the intricate architecture of which has been recently revealed by single cell sequencing technologies. Amidst this phenotypic heterogeneity, several alternative pathways of CAT may exist both between and within individual tumours and their subtypes, including GBM. Indeed, different contributions of cells expressing key coagulant mediators, such as tissue factor, or podoplanin, have been identified in GBM subtypes driven by oncogenic mutations in EGFR, IDH1 and other transforming genes. Thus, a better understanding of cellular sources of CAT, including dominant cancer cell phenotypes and their dynamic shifts, may help design more personalised approaches to thrombosis in cancer patients to improve outcomes.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Trombosis/patología , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/genética , Ecosistema , Epigénesis Genética , Glioblastoma/complicaciones , Glioblastoma/genética , Humanos , Oncogenes , Trombosis/etiología
12.
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
13.
Clin Exp Metastasis ; 35(4): 237-246, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29492795

RESUMEN

Patients with cancer are at high risk of both thromboembolic and haemorrhagic events during the course of their disease. The pathogenesis of haemostatic dysfunction in cancer is complex and involves the interplay of multiple factors. There is growing evidence that interactions between malignancies and the coagulation system are not random but can represent coordinated and clinically-significant adaptations that enhance tumour cell survival, proliferation and metastatic potential. A detailed understanding of the interactions between the haemostatic systems and the pathophysiology of metastasis may not only provide insight into strategies that could potentially reduce the incidence of thrombohaemorrhagic events and complications, but could also help design strategies that are capable of modifying tumour biology, progression and metastatic potential in ways that could enhance anticancer therapies and thereby improve overall survival.


Asunto(s)
Coagulación Sanguínea , Neoplasias/sangre , Neoplasias/patología , Animales , Hemostasis , Humanos , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Células Neoplásicas Circulantes , Neovascularización Patológica/patología
14.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA