Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell ; 181(6): 1346-1363.e21, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32473126

RESUMEN

Enhanced blood vessel (BV) formation is thought to drive tumor growth through elevated nutrient delivery. However, this observation has overlooked potential roles for mural cells in directly affecting tumor growth independent of BV function. Here we provide clinical data correlating high percentages of mural-ß3-integrin-negative tumor BVs with increased tumor sizes but no effect on BV numbers. Mural-ß3-integrin loss also enhances tumor growth in implanted and autochthonous mouse tumor models with no detectable effects on BV numbers or function. At a molecular level, mural-cell ß3-integrin loss enhances signaling via FAK-p-HGFR-p-Akt-p-p65, driving CXCL1, CCL2, and TIMP-1 production. In particular, mural-cell-derived CCL2 stimulates tumor cell MEK1-ERK1/2-ROCK2-dependent signaling and enhances tumor cell survival and tumor growth. Overall, our data indicate that mural cells can control tumor growth via paracrine signals regulated by ß3-integrin, providing a previously unrecognized mechanism of cancer growth control.


Asunto(s)
Integrina beta3/metabolismo , Neoplasias/metabolismo , Carga Tumoral/fisiología , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Femenino , Humanos , Masculino , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología
2.
Gastroenterology ; 162(4): 1197-1209.e13, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34973296

RESUMEN

BACKGROUND & AIMS: Barrett's esophagus (BE) is a risk factor for esophageal adenocarcinoma but our understanding of how it evolves is poorly understood. We investigated BE gland phenotype distribution, the clonal nature of phenotypic change, and how phenotypic diversity plays a role in progression. METHODS: Using immunohistochemistry and histology, we analyzed the distribution and the diversity of gland phenotype between and within biopsy specimens from patients with nondysplastic BE and those who had progressed to dysplasia or had developed postesophagectomy BE. Clonal relationships were determined by the presence of shared mutations between distinct gland types using laser capture microdissection sequencing of the mitochondrial genome. RESULTS: We identified 5 different gland phenotypes in a cohort of 51 nondysplastic patients where biopsy specimens were taken at the same anatomic site (1.0-2.0 cm superior to the gastroesophageal junction. Here, we observed the same number of glands with 1 and 2 phenotypes, but 3 phenotypes were rare. We showed a common ancestor between parietal cell-containing, mature gastric (oxyntocardiac) and goblet cell-containing, intestinal (specialized) gland phenotypes. Similarly, we have shown a clonal relationship between cardiac-type glands and specialized and mature intestinal glands. Using the Shannon diversity index as a marker of gland diversity, we observed significantly increased phenotypic diversity in patients with BE adjacent to dysplasia and predysplasia compared to nondysplastic BE and postesophagectomy BE, suggesting that diversity develops over time. CONCLUSIONS: We showed that the range of BE phenotypes represents an evolutionary process and that changes in gland diversity may play a role in progression. Furthermore, we showed a common ancestry between gastric and intestinal-type glands in BE.


Asunto(s)
Esófago de Barrett , Neoplasias Esofágicas , Esófago de Barrett/patología , Neoplasias Esofágicas/patología , Unión Esofagogástrica/patología , Humanos , Fenotipo
3.
Nature ; 514(7520): 112-6, 2014 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-25079333

RESUMEN

Chemoresistance is a serious limitation of cancer treatment. Until recently, almost all the work done to study this limitation has been restricted to tumour cells. Here we identify a novel molecular mechanism by which endothelial cells regulate chemosensitivity. We establish that specific targeting of focal adhesion kinase (FAK; also known as PTK2) in endothelial cells is sufficient to induce tumour-cell sensitization to DNA-damaging therapies and thus inhibit tumour growth in mice. The clinical relevance of this work is supported by our observations that low blood vessel FAK expression is associated with complete remission in human lymphoma. Our study shows that deletion of FAK in endothelial cells has no apparent effect on blood vessel function per se, but induces increased apoptosis and decreased proliferation within perivascular tumour-cell compartments of doxorubicin- and radiotherapy-treated mice. Mechanistically, we demonstrate that endothelial-cell FAK is required for DNA-damage-induced NF-κB activation in vivo and in vitro, and the production of cytokines from endothelial cells. Moreover, loss of endothelial-cell FAK reduces DNA-damage-induced cytokine production, thus enhancing chemosensitization of tumour cells to DNA-damaging therapies in vitro and in vivo. Overall, our data identify endothelial-cell FAK as a regulator of tumour chemosensitivity. Furthermore, we anticipate that this proof-of-principle data will be a starting point for the development of new possible strategies to regulate chemosensitization by targeting endothelial-cell FAK specifically.


Asunto(s)
Daño del ADN , Resistencia a Antineoplásicos/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Citocinas/biosíntesis , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos/genética , Células Endoteliales/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/deficiencia , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Humanos , Ratones , FN-kappa B/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Neoplasias/radioterapia , Fosforilación/efectos de los fármacos
4.
J Pathol ; 244(1): 61-70, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28940516

RESUMEN

It is widely accepted that the cell of origin of breast cancer is the adult mammary epithelial stem cell; however, demonstrating the presence and location of tissue stem cells in the human breast has proved difficult. Furthermore, we do not know the clonal architecture of the normal and premalignant mammary epithelium or its cellular hierarchy. Here, we use deficiency in the mitochondrial enzyme cytochrome c oxidase (CCO), typically caused by somatic mutations in the mitochondrial genome, as a means to perform lineage tracing in the human mammary epithelium. PCR sequencing of laser-capture microdissected cells in combination with immunohistochemistry for markers of lineage differentiation was performed to determine the clonal nature of the mammary epithelium. We have shown that in the normal human breast, clonal expansions (defined here by areas of CCO deficiency) are typically uncommon and of limited size, but can occur at any site within the adult mammary epithelium. The presence of a stem cell population was shown by demonstrating multi-lineage differentiation within CCO-deficient areas. Interestingly, we observed infrequent CCO deficiency that was restricted to luminal cells, suggesting that niche succession, and by inference stem cell location, is located within the luminal layer. CCO-deficient areas appeared large within areas of ductal carcinoma in situ, suggesting that the rate of clonal expansion was altered in the premalignant lesion. © 2017 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/patología , Carcinoma Intraductal no Infiltrante/patología , Linaje de la Célula , Células Madre/fisiología , Mama/patología , Diferenciación Celular , Células Clonales , Complejo IV de Transporte de Electrones/genética , Células Epiteliales/fisiología , Epitelio/patología , Femenino , Humanos , Mitocondrias/enzimología , Lesiones Precancerosas
5.
Proc Natl Acad Sci U S A ; 110(27): E2490-9, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23766371

RESUMEN

The genetic and morphological development of colorectal cancer is a paradigm for tumorigenesis. However, the dynamics of clonal evolution underpinning carcinogenesis remain poorly understood. Here we identify multipotential stem cells within human colorectal adenomas and use methylation patterns of nonexpressed genes to characterize clonal evolution. Numerous individual crypts from six colonic adenomas and a hyperplastic polyp were microdissected and characterized for genetic lesions. Clones deficient in cytochrome c oxidase (CCO(-)) were identified by histochemical staining followed by mtDNA sequencing. Topographical maps of clone locations were constructed using a combination of these data. Multilineage differentiation within clones was demonstrated by immunofluorescence. Methylation patterns of adenomatous crypts were determined by clonal bisulphite sequencing; methylation pattern diversity was compared with a mathematical model to infer to clonal dynamics. Individual adenomatous crypts were clonal for mtDNA mutations and contained both mucin-secreting and neuroendocrine cells, demonstrating that the crypt contained a multipotent stem cell. The intracrypt methylation pattern was consistent with the crypts containing multiple competing stem cells. Adenomas were epigenetically diverse populations, suggesting that they were relatively mitotically old populations. Intratumor clones typically showed less diversity in methylation pattern than the tumor as a whole. Mathematical modeling suggested that recent clonal sweeps encompassing the whole adenoma had not occurred. Adenomatous crypts within human tumors contain actively dividing stem cells. Adenomas appeared to be relatively mitotically old populations, pocketed with occasional newly generated subclones that were the result of recent rapid clonal expansion. Relative stasis and occasional rapid subclone growth may characterize colorectal tumorigenesis.


Asunto(s)
Adenoma/patología , Linaje de la Célula/genética , Neoplasias Colorrectales/patología , Células Madre Multipotentes/patología , Células Madre Neoplásicas/patología , Adenoma/genética , Adenoma/metabolismo , Diferenciación Celular/genética , Células Clonales/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , ADN Mitocondrial/genética , ADN de Neoplasias/genética , Epigénesis Genética , Humanos , Modelos Biológicos , Células Madre Multipotentes/metabolismo , Mutación , Células Madre Neoplásicas/metabolismo
6.
BMC Cancer ; 14: 237, 2014 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-24693876

RESUMEN

BACKGROUND: Breast cancer is a heterogeneous disease that can be classified into one of 4 main molecular sub-types: luminal A, luminal B, Her2 over-expressing and basal-like (BL). These tumour sub-types require different treatments and have different risks of disease progression. BL cancers can be considered a sub-group of Triple negative (TN) cancers since they lack estrogen (ER), progesterone (PR) and Her2 expression. No targeted treatment currently exists for TN/BL cancers. Thus it is important to identify potential therapeutic targets and describe their relationship with established prognostic factors. Focal adhesion kinase (FAK) is upregulated in several human cancers and also plays a functional role in tumour angiogenesis. However, the association between breast cancer sub-types and tumour endothelial-FAK expression is unknown. METHODS: Using immunofluorescence, we quantified FAK expression in tumour endothelial and tumour cell compartments in 149 invasive breast carcinomas and correlated expression with clinical, pathological and molecular parameters. RESULTS: Low endothelial-FAK expression was independently associated with luminal A tumours at univariate (p < 0.001) and multivariate (p = 0.001) analysis. There was a positive correlation between FAK expression in the vascular and tumour cell compartments (Spearman's correlation co-efficient = 0.394, p < 0.001). Additionally, endothelial and tumour cell FAK expression were significantly increased in TN tumours (p = 0.043 and p = 0.033 respectively), in tumours with negative ER and PR status, and in high grade tumours at univariate analysis. CONCLUSION: Our findings establish a relationship between endothelial-FAK expression levels and the molecular sub-type of invasive breast cancer, and suggest that endothelial-FAK expression is potentially more clinically relevant than tumour cell FAK expression in breast cancer.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Proteína-Tirosina Quinasas de Adhesión Focal/biosíntesis , Invasividad Neoplásica/genética , Neoplasias de la Mama Triple Negativas/genética , Biomarcadores de Tumor/genética , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Clasificación del Tumor , Pronóstico , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias de la Mama Triple Negativas/clasificación , Neoplasias de la Mama Triple Negativas/patología
7.
Gastroenterology ; 142(4): 855-864.e8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22178590

RESUMEN

BACKGROUND & AIMS: Tumors that develop in patients with Crohn's disease tend be multifocal, so field cancerization (the replacement of normal cells with nondysplastic but tumorigenic clones) might contribute to intestinal carcinogenesis. We investigated patterns of tumor development from pretumor intestinal cell clones. METHODS: We performed genetic analyses of multiple areas of intestine from 10 patients with Crohn's disease and intestinal neoplasia. Two patients had multifocal neoplasia; longitudinal sections were collected from 3 patients. Individual crypts were microdissected and genotyped; clonal dependency analysis was used to determine the order and timing of mutations that led to tumor development. RESULTS: The same mutations in KRAS, CDKN2A(p16), and TP53 that were observed in neoplasias were also present in nontumor, nondysplastic, and dysplastic epithelium. In 2 patients, carcinogenic mutations were detected in nontumor epithelium 4 years before tumors developed. The same mutation (TP53 p.R248W) was detected at multiple sites along the entire length of the colon from 1 patient; it was the apparent founder mutation for synchronous tumors and multiple dysplastic areas. Disruption of TP53, CDKN2A, and KRAS were all seen as possible initial events in tumorigenesis; the sequence of mutations (the tumor development pathway) differed among lesions. CONCLUSIONS: Pretumor clones can grow extensively in the intestinal epithelium of patients with Crohn's disease. Segmental resections for neoplasia in patients with Crohn's disease might therefore leave residual pretumor disease, and dysplasia might be an unreliable biomarker for cancer risk. Characterization of the behavior of pretumor clones might be used to predict the development of intestinal neoplasia.


Asunto(s)
Biomarcadores de Tumor/genética , Transformación Celular Neoplásica/genética , Colitis/genética , Enfermedad de Crohn/genética , Ileítis/genética , Neoplasias Intestinales/genética , Mutación Puntual , Adulto , Anciano , Biomarcadores de Tumor/análisis , Transformación Celular Neoplásica/patología , Niño , Células Clonales/patología , Colitis/complicaciones , Colitis/metabolismo , Colitis/patología , Enfermedad de Crohn/complicaciones , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Análisis Mutacional de ADN , Predisposición Genética a la Enfermedad , Humanos , Ileítis/complicaciones , Ileítis/metabolismo , Ileítis/patología , Inmunohistoquímica , Neoplasias Intestinales/química , Neoplasias Intestinales/patología , Captura por Microdisección con Láser , Persona de Mediana Edad , Fenotipo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Factores de Tiempo , Proteína p53 Supresora de Tumor/análisis , Proteína p53 Supresora de Tumor/genética , Proteínas ras/genética
8.
Am J Pathol ; 175(2): 636-48, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19608876

RESUMEN

Pancreatic cancer is characterized by an intense stromal reaction. Reproducible three-dimensional in vitro systems for exploring interactions of the stroma with pancreatic cancer cells have not previously been available, prompting us to develop such a model. Cancer cells were grown on collagen/Matrigel and embedded with or without stromal cells (hTERT-immortalized human PS-1 stellate cells or MRC-5 fibroblasts) for 7 days. Proliferation and apoptosis, as well as important cell-cell adhesion and cytoskeleton-regulating proteins, were studied. PS-1 cells were confirmed as stellate based on the expression of key cytoskeletal proteins and lipid vesicles. Capan-1, and to a lesser extent PaCa-3, cells differentiated into luminal structures, exhibiting a central apoptotic core with a proliferating peripheral rim and an apico-basal polarity. Presence of either stromal cell type translocated Ezrin from apical (when stromal cells were absent) to basal aspects of cancer cells, where it was associated with invasive activity. Interestingly, the presence of 'normal' (not tumor-derived) stromal cells induced total tumor cell number reduction (P < 0.005) associated with a significant decrease in E-cadherin expression (P < 0.005). Conversely, beta-catenin expression was up-regulated (P < 0.01) in the presence of stromal cells with predominant cytoplasmic expression. Moreover, patient samples confirmed that these data recapitulated the clinical situation. In conclusion, pancreatic organotypic culture offers a reproducible, bio-mimetic, three-dimensional in vitro model that allows examination of the interactions between stromal elements and pancreatic cancer cells.


Asunto(s)
Cadherinas/biosíntesis , Proteínas del Citoesqueleto/biosíntesis , Neoplasias Pancreáticas/patología , beta Catenina/biosíntesis , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Técnicas de Cultivo de Órganos , Neoplasias Pancreáticas/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología
9.
Hepatology ; 49(5): 1655-63, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19309719

RESUMEN

UNLABELLED: We have used immunohistochemical and histochemical techniques to identify patches of hepatocytes deficient in the enzyme cytochrome c oxidase, a component of the electron transport chain and encoded by mitochondrial DNA (mtDNA). These patches invariably abutted the portal tracts and expanded laterally as they spread toward the hepatic veins. Here we investigate, using mtDNA mutations as a marker of clonal expansion, the clonality of these patches. Negative hepatocytes were laser-capture microdissected and mutations identified by polymerase chain reaction sequencing of the entire mtDNA genome. Patches of cytochrome c oxidase-deficient hepatocytes were clonal, suggesting an origin from a long-lived cell, presumably a stem cell. Immunohistochemical analysis of function and proliferation suggested that these mutations in cytochrome c oxidase-deficient hepatocytes were nonpathogenic. CONCLUSION: These data show, for the first time, that clonal proliferative units exist in the human liver, an origin from a periportal niche is most likely, and that the trajectory of the units is compatible with a migration of cells from the periportal regions to the hepatic veins.


Asunto(s)
Linaje de la Célula , Complejo IV de Transporte de Electrones/metabolismo , Hepatocitos/enzimología , Hígado/citología , Nicho de Células Madre/citología , Análisis Mutacional de ADN , ADN Mitocondrial/genética , Complejo IV de Transporte de Electrones/genética , Humanos , Inmunohistoquímica
10.
Stem Cells ; 27(6): 1410-20, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19489031

RESUMEN

Methods for lineage tracing of stem cell progeny in human tissues are currently not available. We describe a technique for detecting the expansion of a single cell's progeny that contain clonal mitochondrial DNA (mtDNA) mutations affecting the expression of mtDNA-encoded cytochrome c oxidase (COX). Because such mutations take up to 40 years to become phenotypically apparent, we believe these clonal patches originate in stem cells. Dual-color enzyme histochemistry was used to identify COX-deficient cells, and mutations were confirmed by microdissection of single cells with polymerase chain reaction sequencing of the entire mtDNA genome. These techniques have been applied to human intestine, liver, pancreas, and skin. Our results suggest that the stem cell niche is located at the base of colonic crypts and above the Paneth cell region in the small intestine, in accord with dynamic cell kinetic studies in animals. In the pancreas, exocrine tissue progenitors appeared to be located in or close to interlobular ducts, and, in the liver, we propose that stem cells are located in the periportal region. In the skin, the origin of a basal cell carcinoma appeared to be from the outer root sheath of the hair follicle. We propose that this is a general method for detecting clonal cell populations from which the location of the niche can be inferred, also affording the generation of cell fate maps, all in human tissues. In addition, the technique allows analysis of the origin of human tumors from specific tissue sites.


Asunto(s)
Linaje de la Célula , ADN Mitocondrial/genética , Células Epiteliales/citología , Células Clonales , Complejo IV de Transporte de Electrones/genética , Humanos , Inmunohistoquímica , Mutación , Nicho de Células Madre/citología
11.
J Pathol ; 217(4): 489-96, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19156773

RESUMEN

Little is known about the clonal structure or stem cell architecture of the human small intestinal crypt/villus unit, or how mutations spread and become fixed. Using mitochondrial DNA (mtDNA) mutations as a marker of clonal expansion of stem cell progeny, we aimed to provide answers to these questions. Enzyme histochemistry (for cytochrome c oxidase and succinate dehydrogenase) was performed on frozen sections of normal human duodenum. Laser-capture microdissected cells were taken from crypts/villi. The entire mitochondrial genome was amplified using a nested PCR protocol; sequencing identified mutations and immunohistochemistry demonstrated specific cell lineages. Cytochrome c oxidase-deficient small bowel crypts were observed within all sections: negative crypts contained the same clonal mutation and all differentiated epithelial lineages were present, indicating a common stem cell origin. Mixed crypts were also detected, confirming the existence of multiple stem cells. We observed crypts where Paneth cells were positive but the rest of the crypt was deficient. We have demonstrated patches of deficient crypts that shared a common mutation, suggesting that they have divided by fission. We have shown that all cells within a small intestinal crypt are derived from one common stem cell. Partially-mutated crypts revealed some novel features of Paneth cell biology, suggesting that either they are long-lived or a committed Paneth cell-specific long-lived progenitor was present. We have demonstrated that mutations are fixed in the small bowel by fission and this has important implications for adenoma development.


Asunto(s)
ADN Mitocondrial/genética , Duodeno , Mucosa Intestinal/citología , Mutación/genética , Células Madre/citología , Anciano , Biomarcadores/análisis , Linaje de la Célula , Células Clonales/citología , Células Clonales/enzimología , Análisis Mutacional de ADN , Complejo IV de Transporte de Electrones/análisis , Células Epiteliales/citología , Células Epiteliales/enzimología , Femenino , Histocitoquímica , Humanos , Inmunohistoquímica , Mucosa Intestinal/enzimología , Masculino , Persona de Mediana Edad , Células de Paneth/citología , Células de Paneth/enzimología , Células Madre/enzimología
12.
Gastroenterology ; 134(2): 500-10, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18242216

RESUMEN

BACKGROUND & AIMS: How mutations are established and spread through the human stomach is unclear because the clonal structure of gastric mucosal units is unknown. Here we investigate, using mitochondrial DNA (mtDNA) mutations as a marker of clonal expansion, the clonality of the gastric unit and show how mutations expand in normal mucosa and gastric mucosa showing intestinal metaplasia. This has important implications in gastric carcinogenesis. METHODS: Mutated units were identified by a histochemical method to detect activity of cytochrome c oxidase. Negative units were laser-capture microdissected, and mutations were identified by polymerase chain reaction sequencing. Differentiated epithelial cells were identified by immunohistochemistry for lineage markers. RESULTS: We show that mtDNA mutations establish themselves in stem cells within normal human gastric body units, and are passed on to all their differentiated progeny, thereby providing evidence for clonal conversion to a new stem cell-derived unit-monoclonal conversion, encompassing all gastric epithelial lineages. The presence of partially mutated units indicates that more than one stem cell is present in each unit. Mutated units can divide by fission to form patches, with each unit sharing an indentical, mutant mtDNA genotype. Furthermore, we show that intestinal metaplastic crypts are clonal, possess multiple stem cells, and that fission is a mechanism by which intestinal metaplasia spreads. CONCLUSIONS: These data show that human gastric body units are clonal, contain multiple multipotential stem cells, and provide definitive evidence for how mutations spread within the human stomach, and show how field cancerization develops.


Asunto(s)
Mucosa Gástrica/patología , Células Madre Multipotentes/patología , Neoplasias Gástricas/patología , Neoplasias Gástricas/fisiopatología , Estómago/patología , Transformación Celular Neoplásica/patología , ADN Mitocondrial/genética , Complejo IV de Transporte de Electrones/metabolismo , Epitelio/enzimología , Epitelio/patología , Epitelio/fisiopatología , Mucosa Gástrica/enzimología , Mucosa Gástrica/fisiopatología , Genotipo , Humanos , Metaplasia/patología , Células Madre Multipotentes/enzimología , Mutación , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/patología , Lesiones Precancerosas/fisiopatología , Estómago/enzimología , Estómago/fisiopatología
13.
Cancer Res ; 66(3): 1265-9, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16452177

RESUMEN

Evidence has emerged that bone marrow cells have a greater degree of plasticity than previously thought. However, there has been a call to establish proof that these bone marrow-derived cells function appropriately in their new environment. We have already shown that the bone marrow contributes to myofibroblasts in multiple organs and that this is exacerbated by injury and occurs in a mouse tumor model. Here, we provide evidence that these cells are functioning appropriately by showing that bone marrow-derived myofibroblasts are expressing mRNA for the alpha(1) chain of type I (pro)collagen using a new customized technique. This provides evidence that the bone marrow-tumor stroma axis is functionally relevant and may therefore subsequently be exploited to develop new strategies for anticancer therapy.


Asunto(s)
Células de la Médula Ósea/fisiología , ARN Mensajero/biosíntesis , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Linaje de la Célula , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/genética , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/fisiología , Expresión Génica , Masculino , Ratones , ARN Mensajero/genética , Células del Estroma/citología , Células del Estroma/metabolismo , Células del Estroma/fisiología
14.
J Clin Endocrinol Metab ; 91(11): 4593-8, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16954163

RESUMEN

CONTEXT: Activation of the hypoxia-inducible transcription factors HIF-1 and HIF-2 and a HIF-independent defect in developmental apoptosis have been implicated in the pathogenesis of pheochromocytoma (PCC) associated with VHL, SDHB, and SDHD mutations. OBJECTIVE: Our objective was to compare protein (HIF-1alpha, EPAS1, SDHB, JunB, CCND1, CD34, CLU) and gene (VEGF, BNIP3) expression patterns in VHL and SDHB/D associated tumors. RESULTS: Overexpression of HIF-2 was relatively more common in VHL than SDHB/D PCC (12 of 13 vs. 14 of 20, P = 0.02), whereas nuclear HIF-1 staining was relatively more frequent in SDHB/D PCC (19 of 20 vs. 13 of 16, P = 0.04). In addition, CCND1 and VEGF expression (HIF-2 target genes) was significantly higher in VHL than in SDHB/D PCC. These findings suggest that VHL inactivation leads to preferential HIF-2 activation and CCND1 expression as described previously in VHL-defective renal cell carcinoma cell lines but not in other cell types. These similarities between the downstream consequences of VHL inactivation and HIF dysregulation in renal cell carcinoma and PCC may explain how inactivation of the ubiquitously expressed VHL protein results in susceptibility to specific tumor types. Both VHL and SDHB/D PCC demonstrated reduced CLU and SDHB expression. SDHB PCC are associated with a high risk of malignancy, and expression of (proapototic) BNIP3 was significantly lower in SDHB than VHL PCC. CONCLUSION: Although inactivation of VHL and SDHB/D may disrupt similar HIF-dependent and HIF-independent signaling pathways, their effects on target gene expression are not identical, and this may explain the observed clinical differences in PCC and associated tumors seen with germline VHL and SDHB/D mutations.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Paraganglioma/metabolismo , Feocromocitoma/metabolismo , Succinato Deshidrogenasa/genética , Factores de Transcripción/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Médula Suprarrenal/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Regulación Neoplásica de la Expresión Génica , Mutación de Línea Germinal , Humanos , Inmunohistoquímica , Paraganglioma/genética , Feocromocitoma/genética
15.
Cancer Res ; 63(13): 3819-25, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12839979

RESUMEN

The adenoma:carcinoma sequence is well established. Understanding the molecular pathology of the adenoma is therefore important. There is great controversy within the field. The Vogelstein group champions the "top-down" theory (colorectal adenomas arise and grow across the mucosal surface and down into the crypts), whereas other studies, including our own, propose "bottom-up" spread. Serial sections of 40 small (<3 mm) sporadic colorectal adenomas were stained with H&E, MIB-1, and for beta-catenin. 10 early adenomas were Feulgen-stained and microdissected. We also examined the flat mucosa of three patients who had undergone colectomies for familial adenomatous polyposis (FAP) and specimens from a XO/XY individual with FAP, the latter using in situ hybridization for the Y chromosome. In the earliest sporadic adenomas, there were crypts entirely filled with adenomatous epithelium, which showed proliferative activity and nuclear localization of beta-catenin. There was a sharp cutoff between crypt epithelial cells showing nuclear beta-catenin and surface cells with membrane staining. In slightly larger lesions, adenomatous spread from above was seen. Microdissected adenomas showed multiple fission events, with proliferation distributed equally throughout. In FAP tissue, numerous isolated monocryptal adenomas, which were clonal in origin, were seen. Examination of adenomas in the XO/XY individual showed no instances of XY or XO adenomatous epithelium growing down into crypts of the other genotype. Both sporadic and FAP adenomas start as a unicryptal adenomas and grow initially by crypt fission--a bottom-up pattern. Later, in sporadic adenomas, there is evidence of growth down into adjacent crypts (top-down).


Asunto(s)
Adenoma/patología , Poliposis Adenomatosa del Colon/patología , Neoplasias del Colon/patología , Mucosa Intestinal/citología , Adenoma/cirugía , Colonoscopía , Disección/métodos , Humanos , Mucosa Intestinal/patología
16.
Sci Rep ; 5: 8654, 2015 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-25728748

RESUMEN

LGR5 is known to be a stem cell marker in the murine small intestine and colon, however the localization of LGR5 in human adenoma samples has not been examined in detail, and previous studies have been limited by the lack of specific antibodies. Here we used in situ hybridization to specifically examine LGR5 mRNA expression in a panel of human adenoma and carcinoma samples (n = 66). We found that a small number of cells express LGR5 at the base of normal colonic crypts. We then showed that conventional adenomas widely express high levels of LGR5, and there is no evidence of stereotypic cellular hierarchy. In contrast, serrated lesions display basal localization of LGR5, and the cellular hierarchy resembles that of a normal crypt. Moreover, ectopic crypts found in traditional serrated adenomas show basal LGR5 mRNA, indicating that they replicate the stem cell organization of normal crypts with the development of a cellular hierarchy. These data imply differences in the stem cell dynamics between the serrated and conventional pathways of colorectal carcinogenesis. Furthermore we noted high LGR5 expression in invading cells, with later development of a stem cell niche in adenocarcinomas of all stages.


Asunto(s)
Adenoma/metabolismo , Adenoma/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Receptores Acoplados a Proteínas G/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Colon/patología , Humanos , Hiperplasia , Invasividad Neoplásica , Estadificación de Neoplasias , Pólipos/patología , Nicho de Células Madre
17.
Cancer Cell ; 27(1): 123-37, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25584895

RESUMEN

Increasing chemotherapy delivery to tumors, while enhancing drug uptake and reducing side effects, is a primary goal of cancer research. In mouse and human cancer models in vivo, we show that coadministration of low-dose Cilengitide and Verapamil increases tumor angiogenesis, leakiness, blood flow, and Gemcitabine delivery. This approach reduces tumor growth, metastasis, and minimizes side effects while extending survival. At a molecular level, this strategy alters Gemcitabine transporter and metabolizing enzyme expression levels, enhancing the potency of Gemcitabine within tumor cells in vivo and in vitro. Thus, the dual action of low-dose Cilengitide, in vessels and tumor cells, improves chemotherapy efficacy. Overall, our data demonstrate that vascular promotion therapy is a means to improve cancer treatment.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/patología , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/tratamiento farmacológico , Venenos de Serpiente/administración & dosificación , Verapamilo/administración & dosificación , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/uso terapéutico , Sinergismo Farmacológico , Humanos , Pulmón/irrigación sanguínea , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neovascularización Patológica/tratamiento farmacológico , Páncreas/irrigación sanguínea , Páncreas/patología , Neoplasias Pancreáticas/patología , Venenos de Serpiente/uso terapéutico , Verapamilo/uso terapéutico , Gemcitabina
18.
Nat Commun ; 5: 5054, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25270220

RESUMEN

Metastasis is the main cause of cancer-related death and thus understanding the molecular and cellular mechanisms underlying this process is critical. Here, our data demonstrate, contrary to established dogma, that loss of haematopoietic-derived focal adhesion kinase (FAK) is sufficient to enhance tumour metastasis. Using both experimental and spontaneous metastasis models, we show that genetic ablation of haematopoietic FAK does not affect primary tumour growth but enhances the incidence of metastasis significantly. At a molecular level, haematopoietic FAK deletion results in an increase in PU-1 levels and decrease in GATA-1 levels causing a shift of hematopoietic homeostasis towards a myeloid commitment. The subsequent increase in circulating granulocyte number, with an increase in serum CXCL12 and granulocyte CXCR4 levels, was required for augmented metastasis in mice lacking haematopoietic FAK. Overall our findings provide a mechanism by which haematopoietic FAK controls cancer metastasis.


Asunto(s)
Quinasa 1 de Adhesión Focal/deficiencia , Sistema Hematopoyético/enzimología , Neoplasias/enzimología , Neoplasias/patología , Animales , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quinasa 1 de Adhesión Focal/genética , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Homeostasis , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/fisiopatología , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
19.
Cell Rep ; 8(4): 940-7, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25127143

RESUMEN

Human intestinal stem cell and crypt dynamics remain poorly characterized because transgenic lineage-tracing methods are impractical in humans. Here, we have circumvented this problem by quantitatively using somatic mtDNA mutations to trace clonal lineages. By analyzing clonal imprints on the walls of colonic crypts, we show that human intestinal stem cells conform to one-dimensional neutral drift dynamics with a "functional" stem cell number of five to six in both normal patients and individuals with familial adenomatous polyposis (germline APC(-/+)). Furthermore, we show that, in adenomatous crypts (APC(-/-)), there is a proportionate increase in both functional stem cell number and the loss/replacement rate. Finally, by analyzing fields of mtDNA mutant crypts, we show that a normal colon crypt divides around once every 30-40 years, and the division rate is increased in adenomas by at least an order of magnitude. These data provide in vivo quantification of human intestinal stem cell and crypt dynamics.


Asunto(s)
Poliposis Adenomatosa del Colon/patología , Colon/patología , Focos de Criptas Aberrantes/patología , Poliposis Adenomatosa del Colon/genética , Células Madre Adultas/fisiología , Secuencia de Bases , Estudios de Casos y Controles , Diferenciación Celular , Proliferación Celular , Análisis Mutacional de ADN , Humanos , Mucosa Intestinal/patología , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA