Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cancers (Basel) ; 14(3)2022 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-35159001

RESUMEN

Although the relative risk of renal cell carcinoma associated with chronic kidney injury is particularly high among sub-Saharan African ancestry populations, it is unclear yet whether the APOL1 gene risk variants (RV) for kidney disease additionally elevate this risk. APOL1 G1 and G2 RV contribute to increased risk for kidney disease in black populations, although the disease mechanism has still not been fully deciphered. While high expression levels of all three APOL1 allelic variants, G0 (the wild type allele), G1, and G2 are injurious to normal human cells, renal carcinoma cells (RCC) naturally tolerate inherent high expression levels of APOL1. We utilized CRISPR/Cas9 gene editing to generate isogenic RCC clones expressing APOL1 G1 or G2 risk variants on a similar genetic background, thus enabling a reliable comparison between the phenotypes elicited in RCC by each of the APOL1 variants. Here, we demonstrate that knocking in the G1 or G2 APOL1 alleles, or complete elimination of APOL1 expression, has major effects on proliferation capacity, mitochondrial morphology, cell metabolism, autophagy levels, and the tumorigenic potential of RCC cells. The most striking effect of the APOL1 RV effect was demonstrated in vivo by the complete abolishment of tumor growth in immunodeficient mice. Our findings suggest that, in contrast to the WT APOL1 variant, APOL1 RV are toxic for RCC cells and may act to suppress cancer cell growth. We conclude that the inherent expression of non-risk APOL1 G0 is required for RCC tumorigenicity. RCC cancer cells can hardly tolerate increased APOL1 risk variants expression levels as opposed to APOL1 G0.

2.
Stem Cells ; 37(2): 176-189, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30379370

RESUMEN

The interactions of cancer stem cells (CSCs) within the tumor microenvironment (TME), contribute to the overall phenomenon of intratumoral heterogeneity, which also involve CSC interactions with noncancer stromal cells. Comprehensive understanding of the tumorigenesis process requires elucidating the coordinated gene expression between cancer and tumor stromal cells for each tumor. We show that human gastric cancer cells (GSC1) subvert gene expression and cytokine production by mesenchymal stem cells (GSC-MSC), thus promoting tumor progression. Using mixed composition of human tumor xenografts, organotypic culture, and in vitro assays, we demonstrate GSC1-mediated specific reprogramming of "naïve" MSC into specialized tumor associated MSC equipped with a tumor-promoting phenotype. Although paracrine effect of GSC-MSC or primed-MSC is sufficient to enable 2D growth of GSC1, cell-cell interaction with GSC-MSC is necessary for 3D growth and in vivo tumor formation. At both the transcriptional and at the protein level, RNA-Seq and proteome analyses, respectively, revealed increased R-spondin expression in primed-MSC, and paracrine and juxtacrine mediated elevation of Lgr5 expression in GSC1, suggesting GSC-MSC-mediated support of cancer stemness in GSC1. CSC properties are sustained in vivo through the interplay between GSC1 and GSC-MSC, activating the R-spondin/Lgr5 axis and WNT/ß-catenin signaling pathway. ß-Catenin+ cell clusters show ß-catenin nuclear localization, indicating the activation of the WNT/ß-catenin signaling pathway in these cells. The ß-catenin+ cluster of cells overlap the Lgr5+ cells, however, not all Lgr5+ cells express ß-catenin. A predominant means to sustain the CSC contribution to tumor progression appears to be subversion of MSC in the TME by cancer cells. Stem Cells 2018 Stem Cells 2019;37:176-189.


Asunto(s)
Reprogramación Celular/genética , Células Madre Mesenquimatosas/metabolismo , Neoplasias Gástricas/genética , Humanos , Neoplasias Gástricas/metabolismo , Microambiente Tumoral
3.
Cell Death Dis ; 7(12): e2491, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27906177

RESUMEN

Reestablishing tissue organization of breast cancer cells into acini was previously shown to override their malignant phenotype. In our study, we demonstrate that alpha(v)beta(3) integrin (Int-αvß3), previously shown to play a role in cancer progression, promoted differentiation and growth arrest of organoids derived from luminal A breast cancer cells grown in their relevant three-dimensional microenvironment. These organoids differentiated into normal-like acini resembling a benign stage of breast tissue. Likewise, we demonstrate that Int-αvß3 is selectively expressed in the epithelium of the benign stage of breast tissues, and is lost during the early stages of luminal A breast cancer progression. Notably, the organoids' reversion into normal-like acini was mediated by cancer luminal progenitor-like cells expressing both EpCAMhighCD49flowCD24+ and Int-αvß3. Furthermore, downregulation of Notch4 expression and downstream signaling was shown to mediate Int-αvß3-induced reversion. Intriguingly, when luminal A breast cancer cells expressing Int-αvß3 were injected into a humanized mouse model, differentiated tumors developed when compared with that generated by control cells. Hence, our data suggest that promoting differentiation of luminal A breast cancer cells by signaling emanating from Int-αvß3 can potentially promote 'normalization' of their malignant phenotype and may prevent the malignant cells from progressing.


Asunto(s)
Neoplasias de la Mama/patología , Integrina alfaVbeta3/metabolismo , Células Acinares/metabolismo , Células Acinares/patología , Membrana Basal/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Células Madre Embrionarias/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Hiperplasia , Células MCF-7 , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Organoides/metabolismo , Organoides/patología , Fenotipo , Proteínas Proto-Oncogénicas/metabolismo , Receptor Notch4 , Receptores Notch/metabolismo , Transducción de Señal , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Teratoma/patología
4.
Stem Cells ; 34(4): 1011-26, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26676563

RESUMEN

Non-neoplastic stromal cells harvested from patient tumors were identified as tumor-derived mesenchymal stem cells (MSCs) by their multipotential capacity to differentiate into adipocytes, osteoblasts, and chondrocytes and by the expression of MSC specific cell surface markers. These procedures yielded also epithelial cancer cells and their counterpart MSC from gastric carcinoma (GSC1) and lung carcinoma (LC2). While the LC2 cancer cell growth is independent of their LC-MSC, the GSC1 cancer cell growth is critically dependent on the presence of their counterpart GSC-MSC or their conditioned medium (CM). The fact that none of the various other tumor-derived MSCs was able to restore the specific effect of GSC-MSC on GSC1 cancer cell growth suggests specificity of tumor-derived MSC, which are specifically recruited and "educated"/reprogrammed by the cancer cells to support tumor growth. Using cytokine array analysis, we were able to demonstrate that GSC1 cell growth is mediated through hepatocyte growth factor (HGF)/c-MET signaling pathway which is activated exclusively by HGF secreted from GSC-MSC. An innovative approach demonstrates GSC1-mediated specific tropism of "naïve" MSC from the adjacent tissue in a tumor specific manner to support tumor progression. The results suggest that specific tumor tropic "naïve" MSC are reprogrammed in a tumor-specific manner to support gastric tumor progression. Understanding the mechanisms involved in the interactions of the tumor cancer cells and tumor-derived MSC will constitute the basis for developing multimodal anticancer therapeutic strategies that will also take into account the specific tumor tropism properties of MSC and their reprogramming.


Asunto(s)
Carcinoma/patología , Factor de Crecimiento de Hepatocito/biosíntesis , Neoplasias Pulmonares/patología , Células Madre Mesenquimatosas/patología , Proteínas Proto-Oncogénicas c-met/biosíntesis , Neoplasias Gástricas/patología , Adipocitos/metabolismo , Carcinogénesis/genética , Carcinoma/genética , Carcinoma/metabolismo , Ciclo Celular/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Condrocitos/metabolismo , Medios de Cultivo Condicionados/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Osteoblastos/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Transducción de Señal/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
5.
Breast Cancer Res ; 17: 78, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26040280

RESUMEN

INTRODUCTION: Breast tumors are comprised of distinct cancer cell populations which differ in their tumorigenic and metastatic capacity. Characterization of cell surface markers enables investigators to distinguish between cancer stem cells and their counterparts. CD24 is a well-known cell surface marker for mammary epithelial cells isolation, recently it was suggested as a potential prognostic marker in a wide variety of malignancies. Here, we demonstrate that CD24(+) cells create intra-tumor heterogeneity, and display highly metastatic properties. METHODS: The mammary carcinoma Mvt1 cells were sorted into CD24(-) and CD24(+) cells. Both subsets were morphologically and phenotypically characterized, and tumorigenic capacity was assessed via orthotopic inoculation of each subset into the mammary fat pad of wild-type and MKR mice. The metastatic capacity of each subset was determined with the tail vein metastasis assay. The role of CD24 in tumorigenesis was further examined with shRNA technology. GFP-labeled cells were monitored in vivo for differentiation. The genetic profile of each subset was analyzed using RNA sequencing. RESULTS: CD24(+) cells displayed a more spindle-like cytoplasm. The cells formed mammospheres in high efficiency and CD24(+) tumors displayed rapid growth in both WT and MKR mice, and were more metastatic than CD24- cells. Interestingly, CD24-KD in CD24+ cells had no effect both in vitro and in vivo on the various parameters studied. Moreover, CD24(+) cells gave rise in vivo to the CD24(-) that comprised the bulk of the tumor. RNA-seq analysis revealed enrichment of genes and pathways of the extracellular matrix in the CD24(+) cells. CONCLUSION: CD24(+) cells account for heterogeneity in mammary tumors. CD24 expression at early stages of the cancer process is an indication of a highly invasive tumor. However, CD24 is not a suitable therapeutic target; instead we suggest here new potential targets accounting for early differentiated cancer cells tumorigenic capacity.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Antígeno CD24/metabolismo , Animales , Biomarcadores , Neoplasias de la Mama/genética , Antígeno CD24/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Inmunofenotipificación , Ratones , Metástasis de la Neoplasia , Células Madre Neoplásicas/metabolismo , Fenotipo , Carga Tumoral
6.
Endocr Relat Cancer ; 22(2): 145-57, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25694511

RESUMEN

Accumulating evidence from clinical trials indicates that specific targeting of the IGF1 receptor (IGF1R) is not efficient as an anti-breast cancer treatment. One possible reason is that the mitogenic signals from the insulin receptor (IR) can be processed independently or as compensation to inhibition of the IGF1R. In this study, we highlight the role of the IR in mediating breast tumor progression in both WT mice and a hyperinsulinemic MKR mouse model by induction of Ir (Insr) or Igf1r knockdown (KD) in the mammary carcinoma Mvt-1 cell line. By using the specific IR antagonist-S961, we demonstrated that Igf1r-KD induces elevated responses by the IR to IGF1. On the other hand, Ir-KD cells generated significantly smaller tumors in the mammary fat pads of both WT and MKR mice, as opposed to control cells, whereas the Igf1r-KD cells did not. The tumorigenic effects of insulin on the Mvt-1 cells were also demonstrated using microarray analysis, which indicates alteration of genes and signaling pathways involved in proliferation, the cell cycle, and apoptosis following insulin stimulation. In addition, the correlation between IR and the potential prognostic marker for aggressive breast cancer, CD24, was examined in the Ir-KD cells. Fluorescence-activated cell sorting (FACS) analysis revealed more than 60% reduction in CD24 expression in the Ir-KD cells when compared with the control cells. Our results also indicate that CD24-expressing cells can restore, at least in part, the tumorigenic capacity of Ir-KD cells. Taken together, our results highlight the mitogenic role of the IR in mammary tumor progression with a direct link to CD24 expression.


Asunto(s)
Neoplasias Mamarias Animales/metabolismo , Receptor de Insulina/metabolismo , Animales , Antígeno CD24/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Insulina/farmacología , Neoplasias Mamarias Animales/patología , Ratones , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Carga Tumoral
7.
Exp Hematol ; 42(6): 457-63, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24495871

RESUMEN

FMS-like tyrosine kinase 3 receptor-internal tandem duplication (FLT3-ITD) commonly occurs in acute myeloid leukemia and is considered rare in acute lymphocytic leukemia. Acute leukemia has poor prognosis, mainly due to relapse. Standard FLT3-ITD diagnostic techniques are based on genomic polymerase chain reaction and have recently incorporated GeneScan (Applied Biosystems, Foster City, CA) to identify variations of the FLT3 gene. As this is an average-based assay utilized in a heterogeneous leukemic cell population, we hypothesized that cells of acute leukemia, considered FLT3-ITD-negative by standard methods, could possess a fraction of FLT3-ITD-positive cells. The present study employed single cell mutation analysis to evaluate the FLT3-ITD status in newly diagnosed acute myeloid leukemia (n = 5) and acute lymphocytic leukemia (n = 3) patients. A total of 541 single leukemic cells and 36 mononuclear cells from healthy volunteers were analyzed. Seven patients, considered FLT3-ITD-negative according to bulk DNA analysis, appeared to possess a small fraction of FLT3-ITD-positive cells based on single cell analysis. Moreover, this approach revealed the heterogeneity of the tumor as evident by different FLT3-ITD mutations present in the same patient. The presence of a minor clone carrying FLT3-ITD in almost all patients tested provides evidence that this lesion is a common late event in leukemogenesis. Additionally, 3 relapsed patients demonstrated loss of heterozygosity of the normal allele, affecting 25%-100% of the cells found to be FLT3-ITD-positive. Though further clinical testing is warranted, these findings may have implications on the prognostic significance of FLT3-ITD and the use of targeted therapy.


Asunto(s)
Carcinogénesis , Duplicación de Gen , Leucemia/genética , Leucemia/fisiopatología , Linfocitos T/patología , Tirosina Quinasa 3 Similar a fms/genética , Adulto , Anciano , Alelos , Humanos , Leucocitos/patología , Persona de Mediana Edad , Recurrencia , Análisis de la Célula Individual , Factores de Tiempo
8.
PLoS One ; 8(12): e83651, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24358304

RESUMEN

Intratumoral heterogeneity challenges existing paradigms for anti-cancer therapy. We have previously demonstrated that the human embryonic stem cells (hESC)-derived cellular microenvironment in immunocompromised mice, enables functional distinction of heterogeneous tumor cells, including cells which do not grow into a tumor in a conventional direct tumor xenograft platform. We have identified and characterized six cancer cell subpopulations each clonally expanded from a single cell, derived from human ovarian clear cell carcinoma of a single tumor, to demonstrate striking intratumoral phenotypic heterogeneity that is dynamically dependent on the tumor growth microenvironment. These cancer cell subpopulations, characterized as cancer stem cell subpopulations, faithfully recapitulate the full spectrum of histological phenotypic heterogeneity known for human ovarian clear cell carcinoma. Each of the six subpopulations displays a different level of morphologic and tumorigenic differentiation wherein growth in the hESC-derived microenvironment favors growth of CD44+/aldehyde dehydrogenase positive pockets of self-renewing cells that sustain tumor growth through a process of tumorigenic differentiation into CD44-/aldehyde dehydrogenase negative derivatives. Strikingly, these derivative cells display microenvironment-dependent plasticity with the capacity to restore self-renewal markers and CD44 expression. In the current study, we delineate the distinct gene expression and epigenetic profiles of two such subpopulations, representing extremes of phenotypic heterogeneity in terms of niche-dependent self-renewal and tumorigenic differentiation. By combining Gene Set Enrichment, Gene Ontology and Pathway-focused array analyses with methylation status, we propose a suite of robust differences in tumor self-renewal and differentiation pathways that underlie the striking intratumoral phenotypic heterogeneity which characterize this and other solid tumor malignancies.


Asunto(s)
Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Nicho de Células Madre/genética , Animales , Células Cultivadas , Femenino , Humanos , Ratones , Ratones SCID , Análisis por Micromatrices , Persona de Mediana Edad , Transcriptoma
9.
Cancer Res ; 73(12): 3555-65, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23576551

RESUMEN

Human tumor vessels express tumor vascular markers (TVM), proteins that are not expressed in normal blood vessels. Antibodies targeting TVMs could act as potent therapeutics. Unfortunately, preclinical in vivo studies testing anti-human TVM therapies have been difficult to do due to a lack of in vivo models with confirmed expression of human TVMs. We therefore evaluated TVM expression in a human embryonic stem cell-derived teratoma (hESCT) tumor model previously shown to have human vessels. We now report that in the presence of tumor cells, hESCT tumor vessels express human TVMs. The addition of mouse embryonic fibroblasts and human tumor endothelial cells significantly increases the number of human tumor vessels. TVM induction is mostly tumor-type-specific with ovarian cancer cells inducing primarily ovarian TVMs, whereas breast cancer cells induce breast cancer specific TVMs. We show the use of this model to test an anti-human specific TVM immunotherapeutics; anti-human Thy1 TVM immunotherapy results in central tumor necrosis and a three-fold reduction in human tumor vascular density. Finally, we tested the ability of the hESCT model, with human tumor vascular niche, to enhance the engraftment rate of primary human ovarian cancer stem-like cells (CSC). ALDH(+) CSC from patients (n = 6) engrafted in hESCT within 4 to 12 weeks whereas none engrafted in the flank. ALDH(-) ovarian cancer cells showed no engraftment in the hESCT or flank (n = 3). Thus, this model represents a useful tool to test anti-human TVM therapy and evaluate in vivo human CSC tumor biology.


Asunto(s)
Células Madre Neoplásicas/metabolismo , Neovascularización Patológica/metabolismo , Neoplasias Ováricas/metabolismo , Teratoma/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/inmunología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Células Madre Embrionarias/metabolismo , Femenino , Humanos , Inmunohistoquímica , Inmunotoxinas/administración & dosificación , Inmunotoxinas/inmunología , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Terapia Molecular Dirigida , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Teratoma/tratamiento farmacológico , Teratoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Blood ; 120(3): 603-12, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22645183

RESUMEN

Human cancers display substantial intratumoral genetic heterogeneity, which facilitates tumor survival under changing microenvironmental conditions. Tumor substructure and its effect on disease progression and relapse are incompletely understood. In the present study, a high-throughput method that uses neutral somatic mutations accumulated in individual cells to reconstruct cell lineage trees was applied to hundreds of cells of human acute leukemia harvested from multiple patients at diagnosis and at relapse. The reconstructed cell lineage trees of patients with acute myeloid leukemia showed that leukemia cells at relapse were shallow (divide rarely) compared with cells at diagnosis and were closely related to their stem cell subpopulation, implying that in these instances relapse might have originated from rarely dividing stem cells. In contrast, among patients with acute lymphoid leukemia, no differences in cell depth were observed between diagnosis and relapse. In one case of chronic myeloid leukemia, at blast crisis, most of the cells at relapse were mismatch-repair deficient. In almost all leukemia cases, > 1 lineage was observed at relapse, indicating that diverse mechanisms can promote relapse in the same patient. In conclusion, diverse relapse mechanisms can be observed by systematic reconstruction of cell lineage trees of patients with leukemia.


Asunto(s)
Heterogeneidad Genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Inestabilidad de Microsatélites , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Antineoplásicos/uso terapéutico , Biopsia , Crisis Blástica/tratamiento farmacológico , Crisis Blástica/genética , Crisis Blástica/patología , División Celular/efectos de los fármacos , División Celular/genética , Linaje de la Célula/genética , Resistencia a Antineoplásicos/genética , Citometría de Flujo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Recurrencia , Microambiente Tumoral/genética
11.
Stem Cells ; 30(3): 415-24, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22267284

RESUMEN

Resistance to anticancer therapy has been attributed to interindividual differences in gene expression pathways among tumors, and to the existence within tumors of cancer stem cells with self-renewal capacity. In previous studies, we have demonstrated that the human embryonic stem cell (hESC)-derived cellular microenvironment in immunocompromised mice enables functional distinction of heterogeneous tumor cells, including cells that do not grow into a tumor in conventional direct tumor xenograft platform. In the current study, we use clonally expanded subpopulations derived from ovarian clear cell carcinoma of a single tumor, to demonstrate striking intratumoral phenotypic heterogeneity that is dynamically dependent on the tumor growth microenvironment. Each of six clonally expanded subpopulations displays a different level of morphologic and tumorigenic differentiation, wherein growth in the hESC-derived microenvironment favors growth of CD44+ aldehyde dehydrogenase positive pockets of self-renewing cells that sustain tumor growth through a process of tumorigenic differentiation into CD44- aldehyde dehydrogenase negative derivatives. Strikingly, these derivative cells display microenvironment-dependent plasticity with the capacity to restore self-renewal and CD44 expression. Such intratumoral heterogeneity and plasticity at the level of the key properties of self-renewal and tumorigenic differentiation suggests that a paradigm shift is needed in the approach to anticancer therapy, with the aim of turning malignant growth into a chronic manageable disorder, based on continual monitoring of these tumor growth properties. The hESC-based in vivo model renders intratumoral heterogeneity in the self-renewal and tumorigenic differentiation amenable to biological analysis as well as anticancer therapy testing.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proliferación Celular , Receptores de Hialuranos/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/patología , Animales , Técnicas de Cultivo de Célula , Transformación Celular Neoplásica , Femenino , Citometría de Flujo , Humanos , Ratones , Ratones SCID , Persona de Mediana Edad , Trasplante de Neoplasias , Neoplasias Ováricas/metabolismo , Nicho de Células Madre , Células Tumorales Cultivadas
12.
Circulation ; 125(1): 87-99, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22095829

RESUMEN

BACKGROUND: Pericytes represent a unique subtype of microvessel-residing perivascular cells with diverse angiogenic functions and multilineage developmental features of mesenchymal stem cells. Although various protocols for derivation of endothelial and/or smooth muscle cells from human pluripotent stem cells (hPSC, either embryonic or induced) have been described, the emergence of pericytes in the course of hPSC maturation has not yet been elucidated. METHODS AND RESULTS: We found that during hPSC development, spontaneously differentiating embryoid bodies give rise to CD105(+)CD90(+)CD73(+)CD31(-) multipotent clonogenic mesodermal precursors, which can be isolated and efficiently expanded. Isolated and propagated cells expressed characteristic pericytic markers, including CD146, NG2, and platelet-derived growth factor receptor ß, but not the smooth muscle cell marker α-smooth muscle actin. Coimplantation of hPSC-derived endothelial cells with pericytes resulted in functional and rapid anastomosis to the murine vasculature. Administration of pericytes into immunodeficient mice with limb ischemia promoted significant vascular and muscle regeneration. At day 21 after transplantation, recruited hPSC pericytes were found incorporated into recovered muscle and vasculature. CONCLUSIONS: Derivation of vasculogenic and multipotent pericytes from hPSC can be used for the development of vasculogenic models using multiple vasculogenic cell types for basic research and drug screening and can contribute to angiogenic regenerative medicine.


Asunto(s)
Extremidades/irrigación sanguínea , Isquemia/cirugía , Células Madre Multipotentes/trasplante , Pericitos/trasplante , Células Madre Pluripotentes/trasplante , Recuperación de la Función/fisiología , Animales , Células Endoteliales/trasplante , Extremidades/cirugía , Humanos , Isquemia/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID
13.
J Biol Chem ; 285(17): 13045-56, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20139070

RESUMEN

Sirt1, a NAD-dependent protein deacetylase, is reported to regulate intracellular metabolism and attenuate reactive oxidative species (ROS)-induced apoptosis leading to longevity and acute stress resistance. We created transgenic (TG) mice with kidney-specific overexpression of Sirt1 using the promoter sodium-phosphate cotransporter IIa (Npt2) driven specifically in proximal tubules and investigated the kidney-specific role of Sirt1 in the protection against acute kidney injury (AKI). We also elucidated the role of number or function of peroxisome and mitochondria in mediating the mechanisms for renal protective effects of Sirt1 in AKI. Cisplatin-induced AKI decreased the number and function of peroxisomes as well as mitochondria and led to increased local levels of ROS production and renal tubular apoptotic cells. TG mice treated with cisplatin mitigated AKI, local ROS, and renal tubular apoptotic tubular cells. Consistent with these results, TG mice treated with cisplatin also exhibited recovery of peroxisome number and function, as well as rescued mitochondrial function; however, mitochondrial number was not recovered. Immunoelectron microscopic findings consistently demonstrated that the decrease in peroxisome number by cisplatin in wild type mice was restored in transgenic mice. In HK-2 cells, a cultured proximal tubule cell line, overexpression of Sirt1 rescued the cisplatin-induced cell apoptosis through the restoration of peroxisome number, although the mitochondria number was not restored. These results indicate that Sirt1 overexpression in proximal tubules rescues cisplatin-induced AKI by maintaining peroxisomes number and function, concomitant up-regulation of catalase, and elimination of renal ROS levels. Renal Sirt1 can be a potential therapeutic target for the treatment of AKI.


Asunto(s)
Enfermedades Renales/metabolismo , Túbulos Renales Proximales/lesiones , Túbulos Renales Proximales/metabolismo , Peroxisomas/metabolismo , Sirtuina 1/biosíntesis , Enfermedad Aguda , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Catalasa/biosíntesis , Catalasa/genética , Línea Celular , Cisplatino/efectos adversos , Cisplatino/farmacología , Enfermedades Renales/inducido químicamente , Enfermedades Renales/genética , Enfermedades Renales/terapia , Túbulos Renales Proximales/patología , Longevidad/efectos de los fármacos , Longevidad/genética , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/genética , Mitocondrias/metabolismo , Especificidad de Órganos , Peroxisomas/genética , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 1/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/biosíntesis , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
14.
Clin Cancer Res ; 15(1): 70-80, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19118034

RESUMEN

PURPOSE: In previous studies, we have used human embryonic stem cells (hESC) to generate a tissue microenvironment in immunocompromised mice as an experimental approach for studying human tumorigenesis. We now examine the attributes of such a cellular microenvironment in supporting the growth of human cancer cells freshly harvested from malignant ovarian ascites and to determine whether there are differences among subsets of ascites-derived cancer cells in terms of tumorigenic capacity in the conventional murine xenograft model and in the hESC-derived microenvironment. EXPERIMENTAL DESIGN: Freshly harvested malignant ovarian ascites-derived cancer cells and six derivative ovarian cancer cell subpopulations (CCSP) were characterized for ovarian cancer-associated biomarker expression both in vitro and in vivo and for their capacity to generate tumors in the two models. RESULTS: Ovarian cancer-associated biomarkers were detected in the ascites-derived cancer cells and in the six newly established CCSPs. Nevertheless, certain CCSPs that did not develop into tumors in a conventional murine xenograft model did generate tumors in the hESC-derived cellular microenvironment, indicating variable niche dependency for the tumorigenic capacity of the different CCSPs. The hESC-derived microenvironment provided an improved niche for supporting growth of certain tumor cell subpopulations. CONCLUSIONS: The results highlight the experimental utility of the hESC-derived cellular microenvironment to enable functional distinction of CCSPs, including the identification of cells that do not grow into a tumor in the conventional direct tumor xenograft platform, thereby rendering such cells accessible to characterization and testing of anticancer therapies.


Asunto(s)
Adenocarcinoma de Células Claras/patología , Ascitis/patología , Modelos Animales de Enfermedad , Neoplasias Ováricas/patología , Animales , Biomarcadores de Tumor/análisis , Femenino , Humanos , Ratones , Ratones SCID , Persona de Mediana Edad , Trasplante de Neoplasias , Trasplante Heterólogo
15.
FASEB J ; 21(10): 2551-63, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17435178

RESUMEN

Human embryonic stem cells (hESC) are pluripotent lines that can differentiate in vitro into cell derivatives of all three germ layers, including cardiomyocytes. Successful application of these unique cells in the areas of cardiovascular research and regenerative medicine has been hampered by difficulties in identifying and selecting specific cardiac progenitor cells from the mixed population of differentiating cells. We report the generation of stable transgenic hESC lines, using lentiviral vectors, and single-cell clones that express a reporter gene (eGFP) under the transcriptional control of a cardiac-specific promoter (the human myosin light chain-2V promoter). Our results demonstrate the appearance of eGFP-expressing cells during the differentiation of the hESC as embryoid bodies (EBs) that can be identified and sorted using FACS (purity>95%, viability>85%). The eGFP-expressing cells were stained positively for cardiac-specific proteins (>93%), expressed cardiac-specific genes, displayed cardiac-specific action-potentials, and could form stable myocardial cell grafts following in vivo cell transplantation. The generation of these transgenic hESC lines may be used to identify and study early cardiac precursors for developmental studies, to robustly quantify the extent of cardiomyocyte differentiation, to label the cells for in vivo grafting, and to allow derivation of purified cell populations of cardiomyocytes for future myocardial cell therapy strategies.


Asunto(s)
Células Madre Embrionarias/citología , Corazón/fisiología , Células Musculares/fisiología , Diferenciación Celular , Línea Celular , Células Clonales , Cartilla de ADN , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Corazón/embriología , Humanos , Células Musculares/citología , Miocardio/citología , Transfección
16.
Methods Mol Biol ; 331: 329-46, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16881525

RESUMEN

There is no available experimental system wherein human cancer cells can be grown in the context of a mixed population of normal differentiated human cells for testing biological aspects of cancer cell growth (tumor cell invasion, angiogenesis) or response to anti-cancer therapies. Human embryonic stem cells when implanted into immunocompromised mice develop teratomas containing complex structures, comprising differentiated cell types representing the major germline-derived lineages. We sought to determine whether human cancer cells would grow within such teratomas and display properties associated with malignancy such as invasiveness and recruitment of blood vessels. Ovarian cancer cells (HEY), stably expressing an H2A-GFP fusion protein, which allows tracking of tumor cells, were injected into mature teratomas and developed into tumors. The growth, proliferation capacity, invasion, and induction of blood vessel formation were examined. We propose using the novel experimental platform we have described, consisting of human tumor cells growing within a human cellular microenvironment derived from human embryonic stem cells, to develop a preclinical model for investigating and manipulating the stromal response in tumor cell growth, as an additional tool in cancer research.


Asunto(s)
Neoplasias Ováricas/patología , Células Madre Pluripotentes/citología , Teratoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Diferenciación Celular , División Celular , Línea Celular Tumoral , ADN de Neoplasias/aislamiento & purificación , Femenino , Perfilación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunohistoquímica , Ratones , Ratones SCID , Invasividad Neoplásica , Trasplante de Neoplasias , Neovascularización Patológica/patología , Neoplasias Ováricas/terapia , Plásmidos , ARN Neoplásico/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Teratoma/terapia , Transfección/métodos
17.
Cancer Res ; 66(7): 3792-801, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16585206

RESUMEN

The awareness of the important role that the surrounding tissue microenvironment and stromal response play in the process of tumorigenesis has grown as a result of in vivo models of tumor xenograft growth in immunocompromised mice. In the current study, we used human embryonic stem cells in order to study the interactions of tumor cells with the surrounding microenvironment of differentiated human cell tissues and structures. Several cancer cell types stably expressing an H2A-green fluorescence protein fusion protein, which allowed tracking of tumor cells, were injected into mature teratomas and developed into tumors. The salient findings were: (a) the observation of growth of tumor cells with high proliferative capacity within the differentiated microenvironment of the teratoma, (b) the identification of invasion by tumor cells into surrounding differentiated teratoma structures, and (c) the identification of blood vessels of human teratoma origin, growing adjacent to and within the cancer cell-derived tumor. Mouse embryonic stem cell-derived teratomas also supported cancer cell growth, but provided a less suitable model for human tumorigenesis studies. Anticancer immunotherapy treatment directed against A431 epidermoid carcinoma cell-related epitopes induced the complete regression of A431-derived tumor xenografts following direct i.m. injection in immunocompromised mice, as opposed to corresponding tumors growing within a human embryonic stem cell-derived microenvironment, wherein remnant foci of viable tumor cells were detected and resulted in tumor recurrence. We propose using this novel experimental model as a preclinical platform for investigating and manipulating the stromal response in tumor cell growth as an additional tool in cancer research.


Asunto(s)
Comunicación Celular/fisiología , Neoplasias/patología , Células Madre/citología , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Embrión de Mamíferos/citología , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunoterapia/métodos , Ratones , Ratones SCID , Neoplasias/irrigación sanguínea , Neoplasias/genética , Neoplasias/terapia , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Teratoma/irrigación sanguínea , Teratoma/genética , Teratoma/patología , Teratoma/terapia , Transfección , Trasplante Heterólogo
18.
J Biol Chem ; 279(49): 50976-85, 2004 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-15371422

RESUMEN

Inactivation of p53 and activation of telomerase occur in the majority of human cancers, raising the possibility of a link between these two pathways. Overexpression of wild-type p53 down-regulates the enzymatic activity of telomerase in various cancer cell lines through transcriptional repression of its catalytic subunit, human telomerase reverse transcriptase (hTERT). In this study, we re-evaluated the role of p53 in telomerase regulation using isogenic cell lines expressing physiological levels of p53. We demonstrate that endogenous wild-type p53 was able to down-regulate telomerase activity, hTERT mRNA levels, and promoter activity; however, the ability to repress hTERT expression was found to be cell type-specific. The integrity of the DNA-binding core domain, the N-terminal transactivation domain, and the C-terminal oligomerization domains of p53 was essential for hTERT promoter repression, whereas the proline-rich domain and the extreme C terminus were not required. Southwestern and chromatin immunoprecipitation experiments demonstrated lack of p53 binding to the hTERT promoter, raising the possibility of an indirect repressive mechanism. The down-regulation of hTERT promoter activity was abolished by a dominant-negative E2F1 mutant. Mutational analysis identified a specific E2F site responsible for p53-mediated repression. Knockdown of the key p53 transcriptional target, p21, was sufficient to eliminate the p53-dependent repression of hTERT. Inactivation of the Rb family using either viral oncoproteins or RNA interference attenuated the repression. Inhibition of histone deacetylases also interfered with the repression of hTERT by p53. Therefore, our results suggest that repression of hTERT by endogenous p53 is mediated by p21 and E2F.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Regulación hacia Abajo , Telomerasa/biosíntesis , Proteína p53 Supresora de Tumor/fisiología , Secuencia de Bases , Southern Blotting , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , ADN/química , Análisis Mutacional de ADN , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Genes Reporteros , Histona Desacetilasas/metabolismo , Humanos , Luciferasas/metabolismo , Datos de Secuencia Molecular , Mutación , Neoplasias/metabolismo , Plásmidos/metabolismo , Mutación Puntual , Prolina/química , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Mensajero/metabolismo , Proteína de Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo , Transcripción Genética , Activación Transcripcional , Transfección , Proteína p53 Supresora de Tumor/metabolismo
19.
Proc Natl Acad Sci U S A ; 100(23): 13507-12, 2003 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-14573705

RESUMEN

There is currently no available experimental system wherein human cancer cells can be grown in the context of a mixed population of normal differentiated human cells for testing biological aspects of cancer cell growth (e.g., tumor cell invasion and angiogenesis) or response to anti-cancer therapies. When implanted into immunocompromised mice, human embryonic stem cells develop teratomas containing complex structures comprising differentiated cell types representing the major germ line-derived lineages. We sought to determine whether human cancer cells would grow within such teratomas and display properties associated with malignancy, such as invasiveness and recruitment of blood vessels. HEY ovarian cancer cells stably expressing an H2A-GFP fusion protein (HEY-GFP) injected into mature teratomas developed into tumors, which allowed tracking of tumor cell invasion and recruitment of human teratoma-derived blood vessels. This provides a straightforward and powerful approach to studying the biological properties of cancer cells within the microenvironment of normal differentiated human cells.


Asunto(s)
Embrión de Mamíferos/citología , Trasplante de Neoplasias/métodos , Teratoma/metabolismo , Animales , Diferenciación Celular , División Celular , Línea Celular , Línea Celular Tumoral , ADN Complementario/metabolismo , Proteínas Fluorescentes Verdes , Histonas/metabolismo , Humanos , Inmunohistoquímica , Proteínas Luminiscentes/metabolismo , Ratones , Ratones SCID , Invasividad Neoplásica , Neovascularización Patológica , Plásmidos/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Transfección
20.
Discov Med ; 3(19): 51-4, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20705041

RESUMEN

Extract: There has been a growing awarenes in recent years that tumorigenesis properties are markedly affected by the surrounding tissue microenvironment at both primary and metastatic sites. Thus, it has been shown that tumor progression is associated with extensive remodeling of adjacent tissues to provide a supportive microenvironment for cancer cell proliferation, migration, invasion, and the formation of blood vessels required for supporting cancer growth. As an example, human prostate carcinoma-associated fibroblasts can promote tumorigenic transformation in initiated human prostate epithelial cells. Appreciation of the importance of the stromal response has led to the development of novel anti-cancer therapeutic agents targeted to frustrate stromal response factors, which support progressive tumor growth. Targets for investigation have included proteases, heparanase and other enzymes expressed by cancer cells or by adjacent stromal cells, which degrade extracellular matrix components and facilitate the release of cytokines and growth factors which stimulate angiogenesis, or support the growth and invasion of cancer cells. A great deal of attention has been directed at the development of anti-angiogenic molecules in particular, some of which have reached clinical trials.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA