Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Int J Cancer ; 147(6): 1519-1527, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32010970

RESUMEN

Hepatocellular carcinoma (HCC) is the most common type of primary tumor in the liver and is a leading cause of cancer-related death worldwide. Activated hepatic stellate cells (HSCs) are key components of the HCC microenvironment and play an important role in the onset and progression of HCC through the secretion of growth factors and cytokines. Current treatment modalities that include chemotherapy, radiotherapy and ablation are able to activate HSCs and remodel the tumor microenvironment. Growing evidence has demonstrated that the complex interaction between activated HSCs and tumor cells can facilitate cancer chemoresistance and metastasis. Therefore, therapeutic targeting of activated HSCs has emerged as a promising strategy to improve treatment outcomes for HCC. This review summarizes the molecular mechanisms of HSC activation triggered by treatment modalities, the function of activated HSCs in HCC, as well as the crosstalk between tumor cells and activated HSCs. Pathways of activated HSC reduction are discussed, including inhibition, apoptosis, and reversion to the inactivated state. Finally, we outline the progress and challenges of therapeutic approaches targeting activated HSCs in the development of HCC treatment.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Hepatocelular/terapia , Células Estrelladas Hepáticas/efectos de los fármacos , Neoplasias Hepáticas/terapia , Neovascularización Patológica/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Comunicación Celular/efectos de los fármacos , Comunicación Celular/inmunología , Comunicación Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Quimioradioterapia/efectos adversos , Quimioradioterapia/métodos , Progresión de la Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/inmunología , Resistencia a Antineoplásicos/efectos de la radiación , Células Estrelladas Hepáticas/inmunología , Células Estrelladas Hepáticas/patología , Células Estrelladas Hepáticas/efectos de la radiación , Humanos , Hígado/irrigación sanguínea , Hígado/citología , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Terapia Molecular Dirigida/métodos , Neovascularización Patológica/etiología , Neovascularización Patológica/patología , Ablación por Radiofrecuencia/efectos adversos , Ablación por Radiofrecuencia/métodos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología , Escape del Tumor/efectos de la radiación , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación
2.
Hum Mutat ; 39(12): 2025-2039, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30204945

RESUMEN

The widespread use of next generation sequencing for clinical testing is detecting an escalating number of variants in noncoding regions of the genome. The clinical significance of the majority of these variants is currently unknown, which presents a significant clinical challenge. We have screened over 6,000 early-onset and/or familial breast cancer (BC) cases collected by the ENIGMA consortium for sequence variants in the 5' noncoding regions of BC susceptibility genes BRCA1 and BRCA2, and identified 141 rare variants with global minor allele frequency < 0.01, 76 of which have not been reported previously. Bioinformatic analysis identified a set of 21 variants most likely to impact transcriptional regulation, and luciferase reporter assays detected altered promoter activity for four of these variants. Electrophoretic mobility shift assays demonstrated that three of these altered the binding of proteins to the respective BRCA1 or BRCA2 promoter regions, including NFYA binding to BRCA1:c.-287C>T and PAX5 binding to BRCA2:c.-296C>T. Clinical classification of variants affecting promoter activity, using existing prediction models, found no evidence to suggest that these variants confer a high risk of disease. Further studies are required to determine if such variation may be associated with a moderate or low risk of BC.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama/genética , Mutación de Línea Germinal , Regiones Promotoras Genéticas , Regiones no Traducidas 5' , Edad de Inicio , Proteína BRCA1/química , Proteína BRCA1/metabolismo , Proteína BRCA2/química , Proteína BRCA2/metabolismo , Factor de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Femenino , Predisposición Genética a la Enfermedad , Humanos , Células MCF-7 , Factor de Transcripción PAX5/metabolismo , Unión Proteica
3.
Cell Rep ; 43(7): 114512, 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39003738

RESUMEN

Tumor self-seeding is a process whereby circulating tumor cells (CTCs) recolonize the primary tumor, which promotes tumor growth, angiogenesis, and invasion. However, the detailed nature and functions of tumor self-seeded cells (TSCs) have not been well defined due to challenges in tracking and isolating TSCs. Here, we report an accurate animal model using photoconvertible tagging to recapitulate the spontaneous process of tumor self-seeding and identify TSCs as a subpopulation of primary tumor cells with enhanced invasiveness and survival. We demonstrate transmembrane-4-L-six-family-1 (TM4SF1) as a marker of TSCs, which promotes migration, invasion, and anchorage-independent survival in cancer cells. By analyzing single-cell RNA sequencing datasets, we identify a potential TSC population with a metastatic profile in patients with cancer, which is detectable in early-stage disease and expands during cancer progression. In summary, we establish a framework to study TSCs and identify emerging cell targets with diagnostic, prognostic, or therapeutic potential in cancers.

4.
Biochim Biophys Acta Mol Basis Dis ; 1867(7): 166142, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33839281

RESUMEN

BACKGROUND & AIMS: Iron has been proposed as influencing the progression of liver disease in subjects with non-alcoholic fatty liver disease (NAFLD). We have previously shown that, in the Hfe-/- mouse model of hemochromatosis, feeding of a high-calorie diet (HCD) leads to increased liver injury. In this study we investigated whether the feeding of an iron deficient/HCD to Hfe-/- mice influenced the development of NAFLD. METHODS: Liver histology was assessed in Hfe-/- mice fed a standard iron-containing or iron-deficient diet plus or minus a HCD. Hepatic iron concentration, serum transferrin saturation and free fatty acid were measured. Expression of genes implicated in iron regulation and fatty liver disease was determined by quantitative real-time PCR (qRT-PCR). RESULTS: Standard iron/HCD-fed mice developed severe steatosis whereas NAS score was reduced in mice fed iron-deficient HCD. Mice fed iron-deficient HCD had lower liver weights, lower transferrin saturation and decreased ferroportin and hepcidin gene expression than HCD-fed mice. Serum non-esterified fatty acids were increased in iron-deficient HCD-fed mice compared with standard iron HCD. Expression analysis indicated that genes involved in fatty-acid binding and mTOR pathways were regulated by iron depletion. CONCLUSIONS: Our results indicate that decreasing iron intake attenuates the development of steatosis resulting from a high calorie diet. These results also suggest that human studies of agents that modify iron balance in patients with NAFLD should be revisited.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Hígado Graso/prevención & control , Proteína de la Hemocromatosis/fisiología , Deficiencias de Hierro , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Animales , Ácidos Grasos no Esterificados/metabolismo , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
Theranostics ; 10(15): 6715-6727, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32550899

RESUMEN

Acetaminophen (APAP) is the foremost cause of drug-induced liver injury in the Western world. Most studies of APAP hepatotoxicity have focused on the hepatocellular injury, but current hepatocyte-related biomarkers have delayed presentation time and a lack of sensitivity. APAP overdose can induce hepatic microvascular congestion, which importantly precedes the injury of hepatocytes. However, the underlying molecular mechanisms remain unclear. It is imperative to discover and validate sensitive and specific translational biomarkers of APAP-induced liver injury. Methods: In this study, we assessed APAP toxicity in sinusoidal endothelial cells and hepatocytes in mice treated with overdose APAP at different time points. The underlying mechanisms of APAP overdose induced sinusoidal endothelial cell injury were investigated by RT2 Profiler PCR arrays. The impact of APAP overdose on endothelial cell function was assessed by pseudovessel formation of endothelial cells in 2D Matrigel and in vivo hepatic vascular integrity using multiphoton microscopy. Finally, the effects of APAP overdose on oxygen levels in the liver and hepatic microcirculation were evaluated by contrast enhanced ultrasonography. Potential imaging-based vascular-related markers for early detection of APAP induced liver injury were assessed. Results: Our study confirmed that hepatic endothelial cells are an early and direct target for APAP hepatotoxicity. ICAM1-related cellular adhesion pathways played a prominent role in APAP-induced endothelial cell injury, which was further validated in primary human sinusoidal endothelial cells and human livers after APAP overdose. APAP overdose impacted pseudovessel formation of endothelial cells and in vivo hepatic vascular integrity. Use of ultrasound to detect APAP-induced liver injury demonstrated that mean transit time, an imaging-based vascular-related biomarker, was more sensitive and precise for early detection of APAP hepatotoxicity and monitoring the treatment response in comparison with a conventional blood-based biomarker. Conclusion: Imaging-based vascular-related biomarkers can identify early and mild liver injury induced by APAP overdose. With further development, such biomarkers may improve the assessment of liver injury and the efficacy of clinical decision-making, which can be extended to other microvascular dysfunction of deep organs.


Asunto(s)
Acetaminofén/toxicidad , Biomarcadores/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Endotelio Vascular/metabolismo , Hepatocitos/efectos de los fármacos , Ultrasonografía/métodos , Analgésicos no Narcóticos/toxicidad , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico por imagen , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Microcirculación , Transcriptoma
6.
Biomaterials ; 31(31): 7928-36, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20688384

RESUMEN

Prostate cancer metastasis is reliant on the reciprocal interactions between cancer cells and the bone niche/micro-environment. The production of suitable matrices to study metastasis, carcinogenesis and in particular prostate cancer/bone micro-environment interaction has been limited to specific protein matrices or matrix secreted by immortalised cell lines that may have undergone transformation processes altering signaling pathways and modifying gene or receptor expression. We hypothesize that matrices produced by primary human osteoblasts are a suitable means to develop an in vitro model system for bone metastasis research mimicking in vivo conditions. We have used a decellularized matrix secreted from primary human osteoblasts as a model for prostate cancer function in the bone micro-environment. We show that this collagen I rich matrix is of fibrillar appearance, highly mineralized, and contains proteins, such as osteocalcin, osteonectin and osteopontin, and growth factors characteristic of bone extracellular matrix (ECM). LNCaP and PC3 cells grown on this matrix, adhere strongly, proliferate, and express markers consistent with a loss of epithelial phenotype. Moreover, growth of these cells on the matrix is accompanied by the induction of genes associated with attachment, migration, increased invasive potential, Ca(2+) signaling and osteolysis. In summary, we show that growth of prostate cancer cells on matrices produced by primary human osteoblasts mimics key features of prostate cancer bone metastases and thus is a suitable model system to study the tumor/bone micro-environment interaction in this disease.


Asunto(s)
Matriz Ósea/metabolismo , Calcificación Fisiológica , Comunicación Celular , Modelos Biológicos , Osteoblastos/metabolismo , Neoplasias de la Próstata/patología , Microambiente Tumoral , Huesos/metabolismo , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Forma de la Célula , Células Epiteliales/metabolismo , Células Epiteliales/patología , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Queratina-8/genética , Queratina-8/metabolismo , Masculino , Metaloproteinasas de la Matriz/metabolismo , Osteoblastos/citología , Osteoblastos/ultraestructura , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA