Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 16(9): e1008758, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32881980

RESUMEN

The COVID-19 pandemic highlights the substantial public health, economic, and societal consequences of virus spillover from a wildlife reservoir. Widespread human transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) also presents a new set of challenges when considering viral spillover from people to naïve wildlife and other animal populations. The establishment of new wildlife reservoirs for SARS-CoV-2 would further complicate public health control measures and could lead to wildlife health and conservation impacts. Given the likely bat origin of SARS-CoV-2 and related beta-coronaviruses (ß-CoVs), free-ranging bats are a key group of concern for spillover from humans back to wildlife. Here, we review the diversity and natural host range of ß-CoVs in bats and examine the risk of humans inadvertently infecting free-ranging bats with SARS-CoV-2. Our review of the global distribution and host range of ß-CoV evolutionary lineages suggests that 40+ species of temperate-zone North American bats could be immunologically naïve and susceptible to infection by SARS-CoV-2. We highlight an urgent need to proactively connect the wellbeing of human and wildlife health during the current pandemic and to implement new tools to continue wildlife research while avoiding potentially severe health and conservation impacts of SARS-CoV-2 "spilling back" into free-ranging bat populations.


Asunto(s)
Animales Salvajes/virología , Betacoronavirus/patogenicidad , Infecciones por Coronavirus/virología , Neumonía Viral/virología , Animales , COVID-19 , Quirópteros/virología , Genoma Viral/genética , Especificidad del Huésped/fisiología , Humanos , Pandemias , SARS-CoV-2
2.
PLoS Genet ; 15(6): e1008216, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31246957

RESUMEN

ASAP1 is a multi-domain adaptor protein that regulates cytoskeletal dynamics, receptor recycling and intracellular vesicle trafficking. Its expression is associated with poor prognosis for a variety of cancers, and promotes cell migration, invasion and metastasis. Little is known about its physiological role. In this study, we used mice with a gene-trap inactivated ASAP1 locus to study the functional role of ASAP1 in vivo, and found defects in tissues derived from mesenchymal progenitor cells. Loss of ASAP1 led to growth retardation and delayed ossification typified by enlarged hypertrophic zones in growth plates and disorganized chondro-osseous junctions. Furthermore, loss of ASAP1 led to delayed adipocyte development and reduced fat depot formation. Consistently, deletion of ASAP1 resulted in accelerated chondrogenic differentiation of mesenchymal cells in vitro, but suppressed osteo- and adipogenic differentiation. Mechanistically, we found that FAK/Src and PI3K/AKT signaling is compromised in Asap1GT/GT MEFs, leading to impaired adipogenic differentiation. Dysregulated FAK/Src and PI3K/AKT signaling is also associated with attenuated osteogenic differentiation. Together these observations suggest that ASAP1 plays a decisive role during the differentiation of mesenchymal progenitor cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Adipogénesis/genética , Condrogénesis/genética , Osteogénesis/genética , Animales , Diferenciación Celular/genética , Quinasa 1 de Adhesión Focal/genética , Regulación del Desarrollo de la Expresión Génica/genética , Placa de Crecimiento/crecimiento & desarrollo , Placa de Crecimiento/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Proteína Oncogénica v-akt/genética , Fosfatidilinositol 3-Quinasas/genética , Familia-src Quinasas/genética
3.
Emerg Infect Dis ; 27(4): 1015-1022, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33770472

RESUMEN

The ongoing global pandemic caused by coronavirus disease has once again demonstrated the role of the family Coronaviridae in causing human disease outbreaks. Because severe acute respiratory syndrome coronavirus 2 was first detected in December 2019, information on its tropism, host range, and clinical manifestations in animals is limited. Given the limited information, data from other coronaviruses might be useful for informing scientific inquiry, risk assessment, and decision-making. We reviewed endemic and emerging infections of alphacoronaviruses and betacoronaviruses in wildlife, livestock, and companion animals and provide information on the receptor use, known hosts, and clinical signs associated with each host for 15 coronaviruses detected in humans and animals. This information can be used to guide implementation of a One Health approach that involves human health, animal health, environmental, and other relevant partners in developing strategies for preparedness, response, and control to current and future coronavirus disease threats.


Asunto(s)
Coronaviridae/aislamiento & purificación , Infecciones por Coronavirus/veterinaria , Reservorios de Enfermedades/veterinaria , Zoonosis/virología , Alphacoronavirus/aislamiento & purificación , Animales , Animales Salvajes , Betacoronavirus/aislamiento & purificación , COVID-19/virología , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/virología , Brotes de Enfermedades , Reservorios de Enfermedades/virología , Especificidad del Huésped , Humanos , Coronavirus del Síndrome Respiratorio de Oriente Medio/aislamiento & purificación , Pandemias , SARS-CoV-2 , Zoonosis/epidemiología
5.
Int J Cancer ; 147(4): 1190-1198, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31675122

RESUMEN

Metastasis is a multistep process, during which circulating tumor cells traffic through diverse anatomical locations. Stable inducible marking of tumor cells in a manner that is tightly spatially and temporally controlled would allow tracking the contribution of cells passing through specific locations to metastatic dissemination. For example, tumor cells enter the lymphatic system and can form metastases in regional lymph nodes, but the relative contribution of tumor cells that traffic through the lymphatic system to the formation of distant metastases remains controversial. Here, we developed a novel genetic switch based on mild transient warming (TW) that allows cells to be marked in a defined spatiotemporal manner in vivo. Prior to warming, cells express only EGFP. Upon TW, the EGFP gene is excised and expression of mCherry is permanently turned on. We employed this system in an experimental pancreatic cancer model and used localized TW to induce the genetic switch in tumor cells trafficking through tumor-draining lymph nodes. Thereby we found that tumor cells disseminating via the lymphatics make a major contribution to the seeding of lung metastases. The inducible genetic marking system we have developed is a powerful tool for the tracking of metastasizing cells in vivo.


Asunto(s)
Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Células Neoplásicas Circulantes/metabolismo , Animales , Línea Celular Tumoral , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ganglios Linfáticos/patología , Metástasis Linfática , Sistema Linfático/patología , Neoplasias/metabolismo , Neoplasias/patología , Células Neoplásicas Circulantes/patología , Ratas , Análisis Espacio-Temporal , Proteína Fluorescente Roja
6.
Br J Cancer ; 123(6): 942-954, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32601464

RESUMEN

BACKGROUND: The activation of the EGFR/Ras-signalling pathway in tumour cells induces a distinct chemokine repertoire, which in turn modulates the tumour microenvironment. METHODS: The effects of EGFR/Ras on the expression and translation of CCL20 were analysed in a large set of epithelial cancer cell lines and tumour tissues by RT-qPCR and ELISA in vitro. CCL20 production was verified by immunohistochemistry in different tumour tissues and correlated with clinical data. The effects of CCL20 on endothelial cell migration and tumour-associated vascularisation were comprehensively analysed with chemotaxis assays in vitro and in CCR6-deficient mice in vivo. RESULTS: Tumours facilitate progression by the EGFR/Ras-induced production of CCL20. Expression of the chemokine CCL20 in tumours correlates with advanced tumour stage, increased lymph node metastasis and decreased survival in patients. Microvascular endothelial cells abundantly express the specific CCL20 receptor CCR6. CCR6 signalling in endothelial cells induces angiogenesis. CCR6-deficient mice show significantly decreased tumour growth and tumour-associated vascularisation. The observed phenotype is dependent on CCR6 deficiency in stromal cells but not within the immune system. CONCLUSION: We propose that the chemokine axis CCL20-CCR6 represents a novel and promising target to interfere with the tumour microenvironment, and opens an innovative multimodal strategy for cancer therapy.


Asunto(s)
Quimiocina CCL20/biosíntesis , Receptores ErbB/fisiología , Neoplasias/inmunología , Microambiente Tumoral , Proteínas ras/fisiología , Animales , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Estadificación de Neoplasias , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/etiología , Receptores CCR6/fisiología , Transducción de Señal/fisiología
7.
J Pathol ; 248(4): 421-437, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30982971

RESUMEN

Tspan8 exhibits a functional role in many cancer types including pancreatic, colorectal, oesophagus carcinoma, and melanoma. We present a first study on the expression and function of Tspan8 in breast cancer. Tspan8 protein was present in the majority of human primary breast cancer lesions and metastases in the brain, bone, lung, and liver. In a syngeneic rat breast cancer model, Tspan8+ tumours formed multiple liver and spleen metastases, while Tspan8- tumours exhibited a significantly diminished ability to metastasise, indicating a role of Tspan8 in metastases. Addressing the underlying molecular mechanisms, we discovered that Tspan8 can mediate up-regulation of E-cadherin and down-regulation of Twist, p120-catenin, and ß-catenin target genes accompanied by the change of cell phenotype, resembling the mesenchymal-epithelial transition. Furthermore, Tspan8+ cells exhibited enhanced cell-cell adhesion, diminished motility, and decreased sensitivity to irradiation. As a regulator of the content and function of extracellular vesicles (EVs), Tspan8 mediated a several-fold increase in EV number in cell culture and the circulation of tumour-bearing animals. We observed increased protein levels of E-cadherin and p120-catenin in these EVs; furthermore, Tspan8 and p120-catenin were co-immunoprecipitated, indicating that they may interact with each other. Altogether, our findings show the presence of Tspan8 in breast cancer primary lesion and metastases and indicate its role as a regulator of cell behaviour and EV release in breast cancer. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Lobular/metabolismo , Tetraspaninas/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Carcinoma Lobular/patología , Línea Celular Tumoral , Vesículas Extracelulares , Femenino , Humanos , Metástasis de la Neoplasia , Ratas , Transducción de Señal
8.
Exp Lung Res ; 46(10): 393-408, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33043719

RESUMEN

PURPOSE: 3 D imaging of the lung is not a trivial undertaking as during preparation the lung may collapse. Also serial sections and scans followed by 3 D reconstruction may lead to artifacts. The present study aims to figure out the best way to perform 3 D imaging in lung research. MATERIALS AND METHODS: We applied an optical tissue clearing (OTC) method, which uses ethyl cinnamate (ECi) as a fast, nontoxic and cheap clearing solvent, for 3 D imaging of retrograde perfused lungs by laser confocal fluorescence microscopy and light sheet fluorescence microscopy. We also introduced expansion microscopy (ExM), a cutting-edge technique, in 3 D imaging of lungs. We examined and compared the usefulness of these techniques for 3 D lung imaging. The ExM protocol was further extended to paraffin-embedded lung metastases blocks. RESULTS: The MHI148-PEI labeled lung vasculature was visualized by retrograde perfusion combined with trachea ligation and ECi based OTC. As compared with trans-cardiac perfusion, the retrograde perfusion results in a better maintenance of lung morphology. 3 D structure of alveoli, vascular branches and cilia in lung were revealed by immunofluorescence staining after ExM. 3 D distribution of microvasculature and neutrophil cells in 10 years old paraffin-embedded lung metastases were analyzed by ExM. CONCLUSIONS: The retrograde perfusion combined with trachea ligation technique could be applied in the lung research in mice. 3 D structure of lung vasculature can be visualized by MHI148-PEI perfusion and ECi based OTC in an efficient way. ExM and immunofluorescence staining protocol is highly recommended to perform 3 D imaging of fresh fixed lung as well as paraffin-embedded lung blocks.


Asunto(s)
Imagenología Tridimensional , Pulmón , Animales , Cinamatos , Pulmón/diagnóstico por imagen , Ratones , Microscopía Fluorescente , Perfusión
9.
Biochem Soc Trans ; 45(1): 173-181, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28202671

RESUMEN

Given its importance in development and homeostasis, bone morphogenetic protein (BMP) signaling is tightly regulated at the extra- and intracellular level. The extracellular matrix (ECM) was initially thought to act as a passive mechanical barrier that sequesters BMPs. However, a new understanding about how the ECM plays an instructive role in regulating BMP signaling is emerging. In this mini-review, we discuss various ways in which the biochemical and physical properties of the ECM regulate BMP signaling.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Matriz Extracelular/metabolismo , Espacio Extracelular/metabolismo , Transducción de Señal , Animales , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Homeostasis , Humanos , Modelos Biológicos
10.
Virol J ; 14(1): 60, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28327168

RESUMEN

BACKGROUND: On November 20, 2016 two novel strains of H5N6 highly pathogenic avian influenza virus (HPAIVs) were isolated from three whooper swans (Cygnus cygnus) at Gangjin Bay in South Jeolla province, South Korea. Identification of HPAIVs in wild birds is significant as there is a potential risk of transmission of these viruses to poultry and humans. RESULTS: Phylogenetic analysis revealed that Gangjin H5N6 viruses classified into Asian H5 clade 2.3.4.4 lineage and were distinguishable from H5N8 and H5N1 HPAIVs previously isolated in Korea. With the exception of the polymerase acidic (PA) gene, the viruses were most closely related to A/duck/Guangdong/01.01SZSGXJK005-Y/2016 (H5N6) (98.90 ~ 99.74%). The PA genes of the two novel Gangjin H5N6 viruses were most closely related to AIV isolates previously characterized from Korea, A/hooded crane/Korea/1176/2016 (H1N1) (99.16%) and A/environment/Korea/W133/2006 (H7N7) (98.65%). The lack of more recent viruses to A/environment/Korea/W133/2006 (H7N7) indicates the need for analysis of recent wild bird AIVs isolated in Korea because they might provide further clues as to the origin of these novel reassortant H5N6 viruses. CONCLUSIONS: Although research on the origins and epidemiology of these infections is ongoing, the most likely route of infection for the whooper swans was through direct or indirect contact with reassortant viruses shed by migratory wild birds in Korea. As H5N6 HPAIVs can potentially be transmitted to poultry and humans, continuous monitoring of AIVs among wild birds will help to mitigate this risk.


Asunto(s)
Anseriformes/virología , Virus de la Influenza A/genética , Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/virología , Virus Reordenados/genética , Virus Reordenados/aislamiento & purificación , Animales , Análisis por Conglomerados , Virus de la Influenza A/clasificación , Filogenia , Virus Reordenados/clasificación , República de Corea
12.
Biochem Cell Biol ; 94(3): 289-96, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27251033

RESUMEN

CRISPR/Cas9 has emerged as a powerful methodology for the targeted editing of genomic DNA sequences. Nevertheless, the intrinsic inefficiency of transfection methods required to use this technique with cultured cells requires the selection and isolation of successfully modified cells, which invariably subjects the cells to stress. Here we report a workflow that allows the isolation of genomically modified cells, even where loss of functional alleles constitutes a selective disadvantage owing to impaired ability to survive stress. Using targeted disruption of the Id1 and Id3 genes in murine B16-F10 and Ret melanoma cell lines as an example, we show that the method allows for the footprintless isolation of CRISPR/Cas9-modified aneuploid cancer cells. We also provide evidence that serial CRISPR/Cas9 modifications can occur, for example when initial homologous recombination events introduce cryptic PAM sequences, and demonstrate that multiple alleles can be successfully targeted in aneuploid cancer cells. By sequencing individual alleles we also found evidence for CRISPR/Cas9-induced transposable element insertion, albeit at a low frequency. This workflow should have broad application in the functional analysis of prosurvival gene function in cultured cells.


Asunto(s)
Aneuploidia , Sistemas CRISPR-Cas , Proteína 1 Inhibidora de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/genética , Animales , Línea Celular Tumoral , Supervivencia Celular/genética , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Reacción en Cadena de la Polimerasa/métodos , Flujo de Trabajo
13.
Hepatology ; 61(1): 238-48, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25131778

RESUMEN

UNLABELLED: Due to its ability to inhibit prometastatic matrix metalloproteinases, tissue inhibitor of metalloproteinases (TIMP)-1 has been thought to suppress tumor metastasis. However, elevated systemic levels of TIMP-1 correlate with poor prognosis in cancer patients, suggesting a metastasis-stimulating role of TIMP-1. In colorectal cancer patients, tumor as well as plasma TIMP-1 levels were correlated with synchronous liver metastasis or distant metastasis-associated disease relapse. In mice, high systemic TIMP-1 levels increased the liver susceptibility towards metastasis by triggering the formation of a premetastatic niche. This promoted hepatic metastasis independent of origin or intrinsic metastatic potential of tumor cells. High systemic TIMP-1 led to increased hepatic SDF-1 levels, which in turn promoted recruitment of neutrophils to the liver. Both inhibition of SDF-1-mediated neutrophil recruitment and systemic depletion of neutrophils reduced TIMP-1-induced increased liver susceptibility towards metastasis. This indicates a crucial functional role of neutrophils in the TIMP-1-induced premetastatic niche. CONCLUSION: Our results identify TIMP-1 as an essential promoter of hepatic premetastatic niche formation.


Asunto(s)
Carcinoma/secundario , Quimiocina CXCL12/metabolismo , Neoplasias Hepáticas/secundario , Infiltración Neutrófila , Receptores CXCR4/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Animales , Carcinoma/sangre , Línea Celular Tumoral , Humanos , Hígado/inmunología , Hígado/metabolismo , Neoplasias Hepáticas/sangre , Ratones , Ratones Endogámicos , Células 3T3 NIH , Inhibidor Tisular de Metaloproteinasa-1/sangre
14.
Glycobiology ; 25(3): 258-68, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25354852

RESUMEN

Enhanced levels in tumors of hyaluronan, a glycosaminoglycan component of the extracellular matrix, and hyaluronidases such as hyaluronidase-1 (Hyal1) that degrade hyaluronan have both been linked to poor prognosis and metastasis, suggesting that the turnover of hyaluronan might contribute to tumor progression. Small hyaluronan oligosaccharides (sHA) can accumulate in tumor interstitial fluid (TIF), and have been implicated in a number of processes that drive tumor progression, including MMP expression and angiogenesis. The properties of Hyal1 suggest that it might contribute to the degradation of hyaluronan in tumors and the subsequent accumulation of sHA. Accumulation of Hyal1-produced sHA may therefore account for the association between Hyal1 and metastasis. Here we have investigated this hypothesis using mouse syngeneic breast tumor models. Specifically, we modulated Hyal1 expression and activity either in the tumor cells themselves, or in the stromal compartment by using Hyal1 knockout (KO) mice. These approaches did not change sHA levels in TIF, but nevertheless fostered metastasis to the lung in some of the models used in the study. Together, these data suggest that Hyal1 can promote lung metastasis in a manner that is not dependent on altered accumulation of sHA in TIF.


Asunto(s)
Neoplasias de la Mama/metabolismo , Líquido Extracelular/metabolismo , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/metabolismo , Neoplasias Pulmonares/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Hialuronoglucosaminidasa/genética , Neoplasias Pulmonares/secundario , Ratones
15.
Emerg Infect Dis ; 21(5): 886-90, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25898265

RESUMEN

Novel Eurasian lineage avian influenza A(H5N8) virus has spread rapidly and globally since January 2014. In December 2014, H5N8 and reassortant H5N2 viruses were detected in wild birds in Washington, USA, and subsequently in backyard birds. When they infect commercial poultry, these highly pathogenic viruses pose substantial trade issues.


Asunto(s)
Animales Salvajes , Aves , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Gripe Aviar/virología , Animales , Brotes de Enfermedades , Genes Virales , Historia del Siglo XXI , Virus de la Influenza A/patogenicidad , Gripe Aviar/historia , Filogenia , Análisis de Secuencia de ADN , Washingtón/epidemiología
16.
Int J Cancer ; 137(3): 686-97, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25530422

RESUMEN

Around 70% of breast cancers express the estrogen receptor α (ERα) and depend on estrogen for growth, survival and disease progression. The presence of hormone sensitivity is usually associated with a favorable prognosis. Use of adjuvant anti-endocrine therapy has significantly decreased breast cancer mortality in patients with early-stage disease, and anti-endocrine therapy also plays a central role in the treatment of advanced stages. However a subset of hormone receptor-positive breast cancers do not benefit from anti-endocrine therapy, and nearly all hormone receptor-positive metastatic breast cancers ultimately develop resistance to anti-hormonal therapies. Despite new insights into mechanisms of anti-endocrine therapy resistance, e.g., crosstalk between ERα and Her2/neu, the management of advanced hormone-receptor-positive breast cancers that are resistant to anti-endocrine agents remains a significant challenge. In the present study, we demonstrate that the proteasome inhibitor Bortezomib strongly inhibits ERα and HER2/neu expression, increases expression of cyclin-dependent kinase inhibitors, inhibits expression of multiple genes associated with poor prognosis in ERα+ breast cancer patients and induces cell death in ER+ breast cancer cells in both the presence and absence of functional p53. Although Bortezomib increased the levels of p53 and increased the expression of pro-apoptotic target genes in ERα+ breast cancer cells harboring wild-type p53, Bortezomib also exerts anti-tumoral effects on ERα+ breast cancer cells through suppression of ERα expression and inhibition of PI3K/Akt/mammalian target of rapamycin (mTOR) and ERK signaling independently of functional p53. These findings suggest that Bortezomib might have the potential to improve the management of anti-endocrine therapy resistant ERα+ breast cancers independently of their p53 status.


Asunto(s)
Antineoplásicos/uso terapéutico , Ácidos Borónicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Inhibidores de Proteasoma/uso terapéutico , Pirazinas/uso terapéutico , Receptores de Estrógenos/metabolismo , Animales , Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Bortezomib , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Receptores ErbB/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Metástasis de la Neoplasia , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Inhibidores de Proteasoma/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazinas/farmacología , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/genética , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Metastasis Rev ; 33(4): 1059-79, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25324146

RESUMEN

The properties and behavior of tumor cells are closely regulated by their microenvironment. Accordingly, stromal cells and extracellular matrix components can have a pronounced effect on cancer initiation, growth, and progression. The linear glycosaminoglycan hyaluronan (HA) is a major component of the extracellular matrix. Altered synthesis and degradation of HA in the tumor context has been implicated in many aspects of tumor biology. In particular, the accumulation of small HA oligosaccharides (sHA) in the tumor interstitial space may play a decisive role, due to the ability of sHA to activate a number of biological processes that are not modulated by high molecular weight (HMW)-HA. In this article, we review the normal physiological role and metabolism of HA and then survey the evidence implicating HA in tumor growth and progression, focusing in particular on the potential contribution of sHA to these processes.


Asunto(s)
Carcinogénesis/genética , Glicosaminoglicanos/metabolismo , Ácido Hialurónico/metabolismo , Neoplasias/genética , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/genética , Glicosaminoglicanos/antagonistas & inhibidores , Glicosaminoglicanos/química , Humanos , Ácido Hialurónico/antagonistas & inhibidores , Hialuronoglucosaminidasa/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Oligosacáridos/metabolismo , Microambiente Tumoral/efectos de los fármacos
18.
Blood ; 120(9): 1899-907, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22797697

RESUMEN

VEGFR-3 is a transmembrane receptor tyrosine kinase that is activated by its ligands VEGF-C and VEGF-D. Although VEGFR-3 has been linked primarily to the regulation of lymphangiogenesis, in the present study, we demonstrate a role for VEGFR-3 in megakaryopoiesis. Using a human erythroleukemia cell line and primary murine BM cells, we show that VEGFR-3 is expressed on megakaryocytic progenitor cells through to the promegakaryoblast stage. Functionally, specific activation of VEGFR-3 impaired the transition to polyploidy of CD41+ cells in primary BM cultures. Blockade of VEGFR-3 promoted endoreplication consistently. In vivo, long-term activation or blockade of VEGFR-3 did not affect steady-state murine megakaryopoiesis or platelet counts significantly. However, activation of VEGFR-3 in sublethally irradiated mice resulted in significantly elevated numbers of CD41+ cells in the BM and a significant increase in diploid CD41+ cells, whereas the number of polyploid CD41+ cells was reduced significantly. Moreover, activation of VEGFR-3 increased platelet counts in thrombopoietin-treated mice significantly and modulated 5-fluorouracil-induced thrombocytosis strongly, suggesting a regulatory role for VEGFR-3 in megakaryopoiesis.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células Progenitoras de Megacariocitos/metabolismo , Trombopoyesis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Antimetabolitos/farmacología , Western Blotting , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Células Cultivadas , Fluorouracilo/farmacología , Expresión Génica , Células HEK293 , Humanos , Células Progenitoras de Megacariocitos/efectos de los fármacos , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ésteres del Forbol/farmacología , Recuento de Plaquetas , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Ploidias , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombopoyetina/farmacología , Factor C de Crecimiento Endotelial Vascular/farmacología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
19.
Semin Cancer Biol ; 22(3): 216-25, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22381352

RESUMEN

Communication between cancer cells and stromal cells, often mediated by extracellular molecules in the tumor microenvironment, plays a central role in tumorigenesis and metastasis. The establishment of a pro-inflammatory milieu is increasingly recognized as an important consequence of these interactions. The family of S100 Ca2+-binding proteins has been implicated in many aspects of the interaction between cancer cells and stromal cells, and contributes to the formation of an inflammatory tumor microenvironment. Focusing on S100A4, S100A8 and S100A9, in this review we discuss the role these proteins play in primary tumors and in the development of metastases, in particular during the formation of pre-metastatic niches.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Inflamación/fisiopatología , Metástasis de la Neoplasia/fisiopatología , Neoplasias/metabolismo , Proteínas S100/metabolismo , Células del Estroma/metabolismo , Microambiente Tumoral/fisiología , Expresión Génica , Humanos , Inflamación/inmunología , Proteínas S100/genética
20.
Semin Cancer Biol ; 22(3): 174-86, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22374376

RESUMEN

The ability of tumor cells to leave a primary tumor, to disseminate through the body, and to ultimately seed new secondary tumors is universally agreed to be the basis for metastasis formation. An accurate description of the cellular and molecular mechanisms that underlie this multistep process would greatly facilitate the rational development of therapies that effectively allow metastatic disease to be controlled and treated. A number of disparate and sometimes conflicting hypotheses and models have been suggested to explain various aspects of the process, and no single concept explains the mechanism of metastasis in its entirety or encompasses all observations and experimental findings. The exciting progress made in metastasis research in recent years has refined existing ideas, as well as giving rise to new ones. In this review we survey some of the main theories that currently exist in the field, and show that significant convergence is emerging, allowing a synthesis of several models to give a more comprehensive overview of the process of metastasis. As a result we postulate a stromal progression model of metastasis. In this model, progressive modification of the tumor microenvironment is equally as important as genetic and epigenetic changes in tumor cells during primary tumor progression. Mutual regulatory interactions between stroma and tumor cells modify the stemness of the cells that drive tumor growth, in a manner that involves epithelial-mesenchymal and mesenchymal-epithelial-like transitions. Similar interactions need to be recapitulated at secondary sites for metastases to grow. Early disseminating tumor cells can progress at the secondary site in parallel to the primary tumor, both in terms of genetic changes, as well as progressive development of a metastatic stroma. Although this model brings together many ideas in the field, there remain nevertheless a number of major open questions, underscoring the need for further research to fully understand metastasis, and thereby identify new and effective ways of treating metastatic disease.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Matriz Extracelular/genética , Células Madre Mesenquimatosas/metabolismo , Metástasis de la Neoplasia/patología , Neoplasias/patología , Células Madre Neoplásicas/metabolismo , Microambiente Tumoral/fisiología , Hibridación Genómica Comparativa , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Expresión Génica , Humanos , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA