Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Front Immunol ; 15: 1328602, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38361951

RESUMEN

Introduction: Quantitative, multiplexed imaging is revealing complex spatial relationships between phenotypically diverse tumor infiltrating leukocyte populations and their prognostic implications. The underlying mechanisms and tissue structures that determine leukocyte distribution within and around tumor nests, however, remain poorly understood. While presumed players in metastatic dissemination, new preclinical data demonstrates that blood and lymphatic vessels (lymphovasculature) also dictate leukocyte trafficking within tumor microenvironments and thereby impact anti-tumor immunity. Here we interrogate these relationships in primary human cutaneous melanoma. Methods: We established a quantitative, multiplexed imaging platform to simultaneously detect immune infiltrates and tumor-associated vessels in formalin-fixed paraffin embedded patient samples. We performed a discovery, retrospective analysis of 28 treatment-naïve, primary cutaneous melanomas. Results: Here we find that the lymphvasculature and immune infiltrate is heterogenous across patients in treatment naïve, primary melanoma. We categorized five lymphovascular subtypes that differ by functionality and morphology and mapped their localization in and around primary tumors. Interestingly, the localization of specific vessel subtypes, but not overall vessel density, significantly associated with the presence of lymphoid aggregates, regional progression, and intratumoral T cell infiltrates. Discussion: We describe a quantitative platform to enable simultaneous lymphovascular and immune infiltrate analysis and map their spatial relationships in primary melanoma. Our data indicate that tumor-associated vessels exist in different states and that their localization may determine potential for metastasis or immune infiltration. This platform will support future efforts to map tumor-associated lymphovascular evolution across stage, assess its prognostic value, and stratify patients for adjuvant therapy.


Asunto(s)
Vasos Linfáticos , Melanoma , Neoplasias Cutáneas , Humanos , Estudios Retrospectivos , Inmunohistoquímica , Vasos Linfáticos/patología , Microambiente Tumoral
2.
J Invest Dermatol ; 143(3): 444-455.e8, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35988589

RESUMEN

There is currently no targeted therapy to treat NF1-mutant melanomas. In this study, we compared the genomic and transcriptomic signatures of NF1-mutant and NF1 wild-type melanoma to reveal potential treatment targets for this subset of patients. Genomic alterations were verified using qPCR, and differentially expressed genes were independently validated using The Cancer Genome Atlas data and immunohistochemistry. Digital spatial profiling with multiplex immunohistochemistry and immunofluorescence were used to validate the signatures. The efficacy of combinational regimens driven by these signatures was tested through in vitro assays using low-passage cell lines. Pathogenic NF1 mutations were identified in 27% of cases. NF1-mutant melanoma expressed higher proliferative markers MK167 and CDC20 than NF1 wild-type (P = 0.008), which was independently validated both in The Cancer Genome Atlas dataset (P = 0.01, P = 0.03) and with immunohistochemistry (P = 0.013, P = 0.036), respectively. Digital spatial profiling analysis showed upregulation of LY6E within the tumor cells (false discovery rate < 0.01, log2 fold change > 1), confirmed with multiplex immunofluorescence showing colocalization of LY6E in melanoma cells. The combination of MAPK/extracellular signal‒regulated kinase kinase and CDC20 coinhibition induced both cytotoxic and cytostatic effects, decreasing CDC20 expression in multiple NF1-mutant cell lines. In conclusion, NF1-mutant melanoma is associated with a distinct genomic and transcriptomic profile. Our data support investigating CDC20 inhibition with MAPK pathway inhibitors as a targeted regimen in this melanoma subtype.


Asunto(s)
Melanoma , Transcriptoma , Humanos , Neurofibromina 1/genética , Melanoma/genética , Genómica , Perfilación de la Expresión Génica , Inhibidores de Proteínas Quinasas/farmacología , Mutación
3.
Cancers (Basel) ; 16(1)2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38201446

RESUMEN

BACKGROUND: We previously reported a higher incidence of a pathogenic germline variant in the kinase insert domain receptor (KDR) in melanoma patients compared to the general population. Here, we dissect the impact of this genotype on melanoma tumor growth kinetics, tumor phenotype, and response to treatment with immune checkpoint inhibitors (ICIs) or targeted therapy. METHODS: The KDR genotype was determined and the associations between the KDR Q472H variant (KDR-Var), angiogenesis, tumor immunophenotype, and response to MAPK inhibition or ICI treatment were examined. Melanoma B16 cell lines were transfected with KDR-Var or KDR wild type (KDR-WT), and the differences in tumor kinetics were evaluated. We also examined the impact of KDR-Var on the response of melanoma cells to a combination of VEGFR inhibition with MAPKi. RESULTS: We identified the KDR-Var genotype in 81/489 (37%) patients, and it was associated with a more angiogenic (p = 0.003) and immune-suppressive tumor phenotype. KDR-Var was also associated with decreased PFS to MAPKi (p = 0.022) and a trend with worse PFS to anti-PD1 therapy (p = 0.06). KDR-Var B16 murine models had increased average tumor volume (p = 0.0027) and decreased CD45 tumor-infiltrating lymphocytes (p = 0.0282). The anti-VEGFR treatment Lenvatinib reduced the tumor size of KDR-Var murine tumors (p = 0.0159), and KDR-Var cells showed synergistic cytotoxicity to the combination of dabrafenib and lenvatinib. CONCLUSIONS: Our data demonstrate a role of germline KDR-Var in modulating melanoma behavior, including response to treatment. Our data also suggest that anti-angiogenic therapy might be beneficial in patients harboring this genotype, which needs to be tested in clinical trials.

4.
Clin Cancer Res ; 27(6): 1778-1791, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33402361

RESUMEN

PURPOSE: Women treated with radiotherapy before 30 years of age have increased risk of developing breast cancer at an early age. Here, we sought to investigate mechanisms by which radiation promotes aggressive cancer. EXPERIMENTAL DESIGN: The tumor microenvironment (TME) of breast cancers arising in women treated with radiotherapy for Hodgkin lymphoma was compared with that of sporadic breast cancers. To investigate radiation effects on carcinogenesis, we analyzed tumors arising from Trp53-null mammary transplants after irradiation of the target epithelium or host using immunocompetent and incompetent mice, some of which were treated with aspirin. RESULTS: Compared with age-matched specimens of sporadic breast cancer, radiation-preceded breast cancers (RP-BC) were characterized by TME rich in TGFß, cyclooxygenase 2, and myeloid cells, indicative of greater immunosuppression, even when matched for triple-negative status. The mechanism by which radiation impacts TME construction was investigated in carcinomas arising in mice bearing Trp53-null mammary transplants. Immunosuppressive TMEs (iTME) were recapitulated in mice irradiated before transplantation, which implicated systemic immune effects. In nu/nu mice lacking adaptive immunity irradiated before Trp53-null mammary transplantation, cancers also established an iTME, which pointed to a critical role for myeloid cells. Consistent with this, irradiated mammary glands contained more macrophages and human cells cocultured with polarized macrophages underwent dysplastic morphogenesis mediated by IFNγ. Treating mice with low-dose aspirin for 6 months postirradiation prevented establishment of an iTME and resulted in less aggressive tumors. CONCLUSIONS: These data show that radiation acts via nonmutational mechanisms to promote markedly immunosuppressive features of aggressive, RP-BCs.


Asunto(s)
Neoplasias de la Mama/radioterapia , Inflamación/complicaciones , Neoplasias Inflamatorias de la Mama/patología , Macrófagos/inmunología , Radioterapia/efectos adversos , Microambiente Tumoral , Animales , Apoptosis , Neoplasias de la Mama/patología , Proliferación Celular , Femenino , Humanos , Inflamación/patología , Neoplasias Inflamatorias de la Mama/etiología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Clin Cancer Res ; 27(1): 131-140, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33208341

RESUMEN

PURPOSE: Several biomarkers of response to immune checkpoint inhibitors (ICI) show potential but are not yet scalable to the clinic. We developed a pipeline that integrates deep learning on histology specimens with clinical data to predict ICI response in advanced melanoma. EXPERIMENTAL DESIGN: We used a training cohort from New York University (New York, NY) and a validation cohort from Vanderbilt University (Nashville, TN). We built a multivariable classifier that integrates neural network predictions with clinical data. A ROC curve was generated and the optimal threshold was used to stratify patients as high versus low risk for progression. Kaplan-Meier curves compared progression-free survival (PFS) between the groups. The classifier was validated on two slide scanners (Aperio AT2 and Leica SCN400). RESULTS: The multivariable classifier predicted response with AUC 0.800 on images from the Aperio AT2 and AUC 0.805 on images from the Leica SCN400. The classifier accurately stratified patients into high versus low risk for disease progression. Vanderbilt patients classified as high risk for progression had significantly worse PFS than those classified as low risk (P = 0.02 for the Aperio AT2; P = 0.03 for the Leica SCN400). CONCLUSIONS: Histology slides and patients' clinicodemographic characteristics are readily available through standard of care and have the potential to predict ICI treatment outcomes. With prospective validation, we believe our approach has potential for integration into clinical practice.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Aprendizaje Automático , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Piel/patología , Adulto , Anciano , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Femenino , Estudios de Seguimiento , Humanos , Procesamiento de Imagen Asistido por Computador , Inhibidores de Puntos de Control Inmunológico/farmacología , Masculino , Melanoma/diagnóstico , Melanoma/inmunología , Melanoma/mortalidad , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Supervivencia sin Progresión , Estudios Prospectivos , Curva ROC , Medición de Riesgo/métodos , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad
6.
Mod Pathol ; 34(3): 562-571, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33005020

RESUMEN

Tumor-infiltrating lymphocytes (TIL) have potential prognostic value in melanoma and have been considered for inclusion in the American Joint Committee on Cancer (AJCC) staging criteria. However, interobserver discordance continues to prevent the adoption of TIL into clinical practice. Computational image analysis offers a solution to this obstacle, representing a methodological approach for reproducibly counting TIL. We sought to evaluate the ability of a TIL-quantifying machine learning algorithm to predict survival in primary melanoma. Digitized hematoxylin and eosin (H&E) slides from prospectively enrolled patients in the NYU melanoma database were scored for % TIL using machine learning and manually graded by pathologists using Clark's model. We evaluated the association of % TIL with recurrence-free survival (RFS) and overall survival (OS) using Cox proportional hazards modeling and concordance indices. Discordance between algorithmic and manual TIL quantification was assessed with McNemar's test and visually by an attending dermatopathologist. In total, 453 primary melanoma patients were scored using machine learning. Automated % TIL scoring significantly differentiated survival using an estimated cutoff of 16.6% TIL (log-rank P < 0.001 for RFS; P = 0.002 for OS). % TIL was associated with significantly longer RFS (adjusted HR = 0.92 [0.84-1.00] per 10% increase in % TIL) and OS (adjusted HR = 0.90 [0.83-0.99] per 10% increase in % TIL). In comparison, a subset of the cohort (n = 240) was graded for TIL by melanoma pathologists. However, TIL did not associate with RFS between groups (P > 0.05) when categorized as brisk, nonbrisk, or absent. A standardized and automated % TIL scoring algorithm can improve the prognostic impact of TIL. Incorporation of quantitative TIL scoring into the AJCC staging criteria should be considered.


Asunto(s)
Diagnóstico por Computador , Interpretación de Imagen Asistida por Computador , Linfocitos Infiltrantes de Tumor/inmunología , Aprendizaje Automático , Melanoma/inmunología , Microscopía , Neoplasias Cutáneas/inmunología , Adulto , Anciano , Automatización de Laboratorios , Biopsia , Bases de Datos Factuales , Progresión de la Enfermedad , Femenino , Humanos , Recuento de Linfocitos , Masculino , Melanoma/mortalidad , Melanoma/patología , Melanoma/terapia , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Estadificación de Neoplasias , Valor Predictivo de las Pruebas , Supervivencia sin Progresión , Reproducibilidad de los Resultados , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Microambiente Tumoral/inmunología
7.
Cancer Immunol Res ; 8(2): 217-229, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31831632

RESUMEN

Because the incidence of breast cancer increases decades after ionizing radiation exposure, aging has been implicated in the evolution of the tumor microenvironment and tumor progression. Here, we investigated radiation-induced carcinogenesis using a model in which the mammary glands of 10-month-old BALB/c mice were transplanted with Trp53-null mammary tissue 3 days after exposure to low doses of sparsely ionizing γ-radiation or densely ionizing particle radiation. Mammary transplants in aged, irradiated hosts gave rise to significantly more tumors that grew more rapidly than those in sham-irradiated mice, with the most pronounced effects seen in mice irradiated with densely ionizing particle radiation. Tumor transcriptomes identified a characteristic immune signature of these aggressive cancers. Consistent with this, fast-growing tumors exhibited an immunosuppressive tumor microenvironment with few infiltrating lymphocytes, abundant immunosuppressive myeloid cells, and high COX-2 and TGFß. Only irradiated hosts gave rise to tumors lacking cytotoxic CD8+ lymphocytes (defined here as immune desert), which also occurred in younger irradiated hosts. These data suggest that host irradiation may promote immunosuppression. To test this, young chimera mice were fed chow containing a honeybee-derived compound with anti-inflammatory and immunomodulatory properties, caffeic acid phenethyl ester (CAPE). CAPE prevented the detrimental effects of host irradiation on tumor growth rate, immune signature, and immunosuppression. These data indicated that low-dose radiation, particularly densely ionizing exposure of aged mice, promoted more aggressive cancers by suppressing antitumor immunity. Dietary intervention with a nontoxic immunomodulatory agent could prevent systemic effects of radiation that fuel carcinogenesis, supporting the potential of this strategy for cancer prevention.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Dieta , Inflamación/dietoterapia , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Experimentales/prevención & control , Neoplasias Inducidas por Radiación/prevención & control , Factores de Edad , Animales , Linfocitos T CD8-positivos/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Inflamación/etiología , Inflamación/patología , Linfocitos Infiltrantes de Tumor/efectos de la radiación , Neoplasias Mamarias Experimentales/etiología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neoplasias Inducidas por Radiación/etiología , Neoplasias Inducidas por Radiación/inmunología , Transcriptoma , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/inmunología , Proteína p53 Supresora de Tumor/metabolismo
8.
J Invest Dermatol ; 139(2): 430-438, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30148988

RESUMEN

The extent of PTEN loss that confers clinical and biological impact in melanoma is unclear. We evaluated the clinical and biologic relevance of PTEN dosage in melanoma and tested the postulate that partial PTEN loss is due to epigenetic mechanisms. PTEN expression was assessed by immunohistochemistry in a stage III melanoma cohort (n = 190) with prospective follow up. Overall, 21 of 190 (11%) tumors had strong PTEN expression, 51 of 190 (27%) had intermediate PTEN, 44 of 190 (23%) had weak PTEN, and 74 of 190 (39%) had absent PTEN. Both weak and absent PTEN expression predicted shorter survival in multivariate analyses (hazard ratio = 2.13, P < 0.01). We show a continuous negative correlation between PTEN and activated Akt in melanoma cells with titrated PTEN expression and in two additional independent tumor datasets. PTEN genomic alterations (deletion, mutation), promoter methylation, and protein destabilization did not fully explain PTEN loss in melanoma, whereas PTEN levels increased with treatment of melanoma cells with the histone deacetylase inhibitor LBH589. Our data indicate that partial PTEN loss is due to modifiable epigenetic mechanisms and drives Akt activation and worse prognosis, suggesting a potential approach to improve the clinical outcome for a subset of patients with advanced melanoma.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Fosfohidrolasa PTEN/genética , Neoplasias Cutáneas/genética , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Femenino , Estudios de Seguimiento , Dosificación de Gen , Humanos , Masculino , Melanoma/mortalidad , Melanoma/patología , Persona de Mediana Edad , Mutación , Estadificación de Neoplasias , Pronóstico , Regiones Promotoras Genéticas , Estudios Prospectivos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piel/patología , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Análisis de Supervivencia , Adulto Joven
9.
Oncotarget ; 8(39): 64932-64953, 2017 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-29029402

RESUMEN

Glioblastoma (GBM) stem cells (GSCs) reside in both hypoxic and vascular microenvironments within tumors. The molecular mechanisms that allow GSCs to occupy such contrasting niches are not understood. We used patient-derived GBM cultures to identify GSC subtypes with differential activation of Notch signaling, which co-exist in tumors but occupy distinct niches and match their metabolism accordingly. Multipotent GSCs with Notch pathway activation reside in perivascular niches, and are unable to entrain anaerobic glycolysis during hypoxia. In contrast, most CD133-expressing GSCs do not depend on canonical Notch signaling, populate tumors regardless of local vascularity and selectively utilize anaerobic glycolysis to expand in hypoxia. Ectopic activation of Notch signaling in CD133-expressing GSCs is sufficient to suppress anaerobic glycolysis and resistance to hypoxia. These findings demonstrate a novel role for Notch signaling in regulating GSC metabolism and suggest intratumoral GSC heterogeneity ensures metabolic adaptations to support tumor growth in diverse tumor microenvironments.

10.
Sci Signal ; 9(457): ra118, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27923913

RESUMEN

Maintenance of mammary functional capacity during cycles of proliferation and regression depends on appropriate cell fate decisions of mammary progenitor cells to populate an epithelium consisting of secretory luminal cells and contractile myoepithelial cells. It is well established that transforming growth factor-ß (TGFß) restricts mammary epithelial cell proliferation and that sensitivity to TGFß is decreased in breast cancer. We show that TGFß also exerts control of mammary progenitor self-renewal and lineage commitment decisions by stringent regulation of breast cancer associated 1 (BRCA1), which controls stem cell self-renewal and lineage commitment. Either genetic depletion of Tgfb1 or transient blockade of TGFß increased self-renewal of mammary progenitor cells in mice, cultured primary mammary epithelial cells, and also skewed lineage commitment toward the myoepithelial fate. TGFß stabilized the abundance of BRCA1 by reducing the abundance of microRNA-182 (miR-182). Ectopic expression of BRCA1 or antagonism of miR-182 in cultured TGFß-deficient mammary epithelial cells restored luminal lineage commitment. These findings reveal that TGFß modulation of BRCA1 directs mammary epithelial cell fate and, because stem or progenitor cells are thought to be the cell of origin for aggressive breast cancer subtypes, suggest that TGFß dysregulation during tumorigenesis may promote distinct breast cancer subtypes.


Asunto(s)
Diferenciación Celular , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/metabolismo , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Animales , Proteína BRCA1 , Femenino , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Células Madre Neoplásicas/patología , Factor de Crecimiento Transformador beta1/genética , Proteínas Supresoras de Tumor/genética
11.
Breast Cancer Res ; 16(6): 463, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25385439

RESUMEN

INTRODUCTION: Estrogen inhibition is effective in preventing breast cancer in only up to 50% of women with precancerous lesions and many experience side effects that are poorly tolerated. As insulin-like growth factor I (IGF-I) underlies both estrogen and progesterone actions and has other direct effects on mammary development and carcinogenesis, we hypothesized that IGF-I inhibition might provide a novel approach for breast cancer chemoprevention. METHODS: In total, 13 women with core breast biopsies diagnostic of atypical hyperplasia (AH) were treated for 10 days with pasireotide, a somatostatin analog which uniquely inhibits IGF-I action in the mammary gland. They then had excision biopsies. 12 patients also had proliferative lesions and one a ductal carcinoma in situ (DCIS). Primary outcomes were changes in cell proliferation and apoptosis after treatment. Expression of estrogen receptor (ER), progesterone receptor (PR), and phosphorylated Insulin-like growth factor I receptor (IGF-1R), protein kinase B (AKT) and extracellular signal-regulated kinases 1/2 (ERK1/2) were also assessed. Core and excision biopsies from 14 untreated patients served as non-blinded controls. Hyperglycemia and other side effects were carefully monitored. RESULTS: Pasireotide decreased proliferation and increased apoptosis in all AH (from 3.6 ± 2.6% to 1.3 ± 1.2% and from 0.3 ± 0.2% to 1.5 ± 1.6%, respectively) and proliferative lesions (from 3.8 ± 2.5% to 1.8 ± 1.8% and from 0.3 ± 0.2% to 1.3 ± 0.6%, respectively). The DCIS responded similarly. ER and PR were not affected by pasireotide, while IGF-1R, ERK1/2 and AKT phosphorylation decreased significantly. In contrast, tissue from untreated controls showed no change in cell proliferation or phosphorylation of IGF-1R, AKT or ERK 1/2. Mild to moderate hyperglycemia associated with reduced insulin levels was found. Glucose fell into the normal range after discontinuing treatment. Pasireotide was well tolerated and did not cause symptoms of estrogen deprivation. CONCLUSIONS: IGF-I inhibition by pasireotide, acting through the IGF-1R, was associated with decreased proliferation and increased apoptosis in pre-malignant breast lesions and one DCIS. Assuming hyperglycemia can be controlled, these data suggest that inhibiting the IGF-I pathway may prove an effective alternative for breast cancer chemoprevention. TRIAL REGISTRATION: NCT01372644 Trial date: July 1, 2007.


Asunto(s)
Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma Intraductal no Infiltrante/tratamiento farmacológico , Proliferación Celular , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Lesiones Precancerosas/tratamiento farmacológico , Somatostatina/análogos & derivados , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patología , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Femenino , Humanos , Hiperplasia/tratamiento farmacológico , Hiperplasia/metabolismo , Hiperplasia/patología , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1 , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Receptores de Somatomedina/metabolismo , Somatostatina/uso terapéutico
12.
Cancer Res ; 74(23): 7137-48, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25304265

RESUMEN

Densely ionizing radiation, which is present in the space radiation environment and used in radiation oncology, has potentially greater carcinogenic effect compared with sparsely ionizing radiation that is prevalent on earth. Here, we used a radiation chimera in which mice were exposed to densely ionizing 350 MeV/amu Si-particles, γ-radiation, or sham-irradiated and transplanted 3 days later with syngeneic Trp53-null mammary fragments. Trp53-null tumors arising in mice irradiated with Si-particles had a shorter median time to appearance and grew faster once detected compared with those in sham-irradiated or γ-irradiated mice. Tumors were further classified by markers keratin 8/18 (K18, KRT18), keratin 14 (K14, KRT14) and estrogen receptor (ER, ESR1), and expression profiling. Most tumors arising in sham-irradiated hosts were comprised of both K18- and K14-positive cells (K14/18) while those tumors arising in irradiated hosts were mostly K18. Keratin staining was significantly associated with ER status: K14/18 tumors were predominantly ER-positive, whereas K18 tumors were predominantly ER-negative. Genes differentially expressed in K18 tumors compared with K14/18 tumor were associated with ERBB2 and KRAS, metastasis, and loss of E-cadherin. Consistent with this, K18 tumors tended to grow faster and be more metastatic than K14/18 tumors, however, K18 tumors in particle-irradiated mice grew significantly larger and were more metastatic compared with sham-irradiated mice. An expression profile that distinguished K18 tumors arising in particle-irradiated mice compared with sham-irradiated mice was enriched in mammary stem cell, stroma, and Notch signaling genes. These data suggest that carcinogenic effects of densely ionizing radiation are mediated by the microenvironment, which elicits more aggressive tumors compared with similar tumors arising in sham-irradiated hosts.


Asunto(s)
Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Microambiente Tumoral/genética , Proteína p53 Supresora de Tumor/genética , Animales , Biomarcadores de Tumor/genética , Cadherinas/genética , Femenino , Perfilación de la Expresión Génica/métodos , Queratinas/genética , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas p21(ras)/genética , Radiación Ionizante , Receptor ErbB-2/genética , Receptores de Estrógenos/genética , Receptores Notch/genética , Células Madre/patología
13.
Stem Cells ; 32(3): 649-61, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24038768

RESUMEN

Children exposed to ionizing radiation have a substantially greater breast cancer risk than adults; the mechanism for this strong age dependence is not known. Here we show that pubertal murine mammary glands exposed to sparsely or densely ionizing radiation exhibit enrichment of mammary stem cell and Notch pathways, increased mammary repopulating activity indicative of more stem cells, and propensity to develop estrogen receptor (ER) negative tumors thought to arise from stem cells. We developed a mammary lineage agent-based model (ABM) to evaluate cell inactivation, self-renewal, or dedifferentiation via epithelial-mesenchymal transition (EMT) as mechanisms by which radiation could increase stem cells. ABM rejected cell inactivation and predicted increased self-renewal would only affect juveniles while dedifferentiation could act in both juveniles and adults. To further test self-renewal versus dedifferentiation, we used the MCF10A human mammary epithelial cell line, which recapitulates ductal morphogenesis in humanized fat pads, undergoes EMT in response to radiation and transforming growth factor ß (TGFß) and contains rare stem-like cells that are Let-7c negative or express both basal and luminal cytokeratins. ABM simulation of population dynamics of double cytokeratin cells supported increased self-renewal in irradiated MCF10A treated with TGFß. Radiation-induced Notch concomitant with TGFß was necessary for increased self-renewal of Let-7c negative MCF10A cells but not for EMT, indicating that these are independent processes. Consistent with these data, irradiating adult mice did not increase mammary repopulating activity or ER-negative tumors. These studies suggest that irradiation during puberty transiently increases stem cell self-renewal, which increases susceptibility to developing ER-negative breast cancer.


Asunto(s)
Envejecimiento/patología , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/efectos de la radiación , Neoplasias Mamarias Animales/patología , Radiación Ionizante , Receptores de Estrógenos/metabolismo , Células Madre/patología , Animales , Biomarcadores/metabolismo , Línea Celular , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Simulación por Computador , Relación Dosis-Respuesta en la Radiación , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Células Epiteliales/efectos de la radiación , Femenino , Humanos , Neoplasias Mamarias Animales/metabolismo , Ratones , Morfogénesis/efectos de los fármacos , Morfogénesis/efectos de la radiación , Receptores Notch/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células Madre/efectos de la radiación , Factor de Crecimiento Transformador beta/farmacología
14.
Cancer Cell ; 19(5): 640-51, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21575864

RESUMEN

Tissue microenvironment is an important determinant of carcinogenesis. We demonstrate that ionizing radiation, a known carcinogen, affects cancer frequency and characteristics by acting on the microenvironment. Using a mammary chimera model in which an irradiated host is transplanted with oncogenic Trp53 null epithelium, we show accelerated development of aggressive tumors whose molecular signatures were distinct from tumors arising in nonirradiated hosts. Molecular and genetic approaches show that TGFß mediated tumor acceleration. Tumor molecular signatures implicated TGFß, and genetically reducing TGFß abrogated the effect on latency. Surprisingly, tumors from irradiated hosts were predominantly estrogen receptor negative. This effect was TGFß independent and linked to mammary stem cell activity. Thus, the irradiated microenvironment affects latency and clinically relevant features of cancer through distinct and unexpected mechanisms.


Asunto(s)
Neoplasias de la Mama/etiología , Transformación Celular Neoplásica/efectos de la radiación , Células Epiteliales/efectos de la radiación , Glándulas Mamarias Animales/efectos de la radiación , Neoplasias Inducidas por Radiación/etiología , Microambiente Tumoral/efectos de la radiación , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Relación Dosis-Respuesta en la Radiación , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/trasplante , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neoplasias Inducidas por Radiación/genética , Neoplasias Inducidas por Radiación/metabolismo , Neoplasias Inducidas por Radiación/patología , Quimera por Radiación , Tiempo de Reacción , Receptores de Estrógenos/deficiencia , Factores de Tiempo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Carga Tumoral , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Irradiación Corporal Total
15.
Methods Mol Biol ; 621: 1-28, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20405356

RESUMEN

The mammary gland consists of an epithelial ductal tree embedded in a fat pad. Adult mammary epithelium has been demonstrated to have outstanding regenerative potential, consistent with the presence of resident, adult stem cells. However, there are currently no bona fide markers to identify these cells within their tissue context. Here, we introduce long-term label retention as a method to investigate the location of quiescent cells (a property attributed to adult stem cells) in situ. Long-term label retaining cells divide actively during tissue development and remain quiescent at homeostasis. These two properties have been attributed to adult stem cells. Therefore, label-retaining cells can be used to identify populations that contain stem cells. We describe the materials and methods necessary to identify and image mammary label-retaining cells, to carry out morphometric analysis on these cells and to map their distribution of the mammary epithelium. The morphometric and spatial analyses described here are generally applicable to any mammary cell populations, and will therefore be useful to characterize mammary stem cells once bona fide mammary stem cell markers become available.


Asunto(s)
Glándulas Mamarias Animales/citología , Coloración y Etiquetado , Animales , Biomarcadores/metabolismo , Bromodesoxiuridina/metabolismo , Femenino , Formaldehído/metabolismo , Glándulas Mamarias Animales/metabolismo , Ratones , Microscopía Fluorescente , Imagen Molecular , Factores de Tiempo
16.
Methods Mol Biol ; 621: 49-55, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20405358

RESUMEN

The regenerative potential of mammary epithelium facilitates assessment of the "stemness" of any epithelial subpopulation in transplantation assays. Thus, mammary tissue can be dissociated into single cells, stained for cell surface markers of interest and classified using fluorescence-activated cell sorting. The selected cells can then be transplanted into epithelium-devoided fat pads from recipient hosts. Recent publications have described markers that enrich for mammary repopulating potential. Here, we describe the materials and methods necessary to sort cells according to these markers. This approach can be used interchangeably with other cell surface markers with slight variation to the protocol.


Asunto(s)
Citometría de Flujo/métodos , Glándulas Mamarias Animales/citología , Células Madre/metabolismo , Animales , Biomarcadores/metabolismo , Ratones , Coloración y Etiquetado
17.
Methods Mol Biol ; 621: 29-47, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20405357

RESUMEN

Mammary reconstitution assays can be used to measure the stem cell frequency within an epithelial population by transplanting increasingly diluted single-cell preparations of the population of interest. There are fundamental steps in the single-cell isolation protocol which are directly related to the number of single epithelial cells obtained. Once single-cell suspensions have been obtained, serial dilutions are prepared and transplanted into the cleared fat pads of the host mice. After 8-10 weeks, the transplanted fat pads are reevaluated for the presence of epithelial outgrowths. Based on the frequency of no outgrowth for each one of the transplanted dilutions, it is possible to estimate the frequency of mammary repopulating cells present in a given cell population. Here, we give details on how to carry out all these steps.


Asunto(s)
Trasplante de Células/métodos , Glándulas Mamarias Animales/citología , Células Madre/citología , Tejido Adiposo/citología , Tejido Adiposo/trasplante , Animales , Proliferación Celular , Separación Celular , Centrifugación , Células Epiteliales/trasplante , Ratones , Coloración y Etiquetado
18.
Integr Biol (Camb) ; 1(1): 80-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20023794

RESUMEN

We have built a novel computational microscopy platform that integrates image acquisition, storage, processing and analysis to study cell populations in situ. This platform allows high-content studies where multiple features are measured and linked at multiple scales. We used this approach to study the cellular composition and architecture of the mouse mammary gland by quantitatively tracking the distribution and type, position, proliferative state, and hormone receptor status of epithelial cells that incorporated bromodeoxyuridine while undergoing DNA synthesis during puberty and retained this label in the adult gland as a function of tissue structure. Immunofluorescence was used to identify label-retaining cells, as well as epithelial cells expressing the proteins progesterone receptor and P63. Only 3.6% of luminal cells were label-retaining cells, the majority of which did not express the progesterone receptor. Multi-scale in situ analysis revealed that luminal label-retaining cells have a distinct nuclear morphology, are enriched 3.4-fold in large ducts, and are distributed asymmetrically across the tissue. We postulated that LRC enriched in the ventral mammary gland represent progenitor cells. Epithelial cells isolated from the ventral versus the dorsal portion of the gland were enriched for the putative stem cell markers CD24 and CD49f as measured by fluorescence activated cell sorting. Thus, quantitative analysis of the cellular composition of the mammary epithelium across spatial scales identified a previously unrecognized architecture in which the ventral-most, large ducts contain a reservoir of undifferentiated, putative stem cells.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/patología , Perfilación de la Expresión Génica/métodos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Microscopía/métodos , Receptores de Somatotropina/metabolismo , Animales , Antígeno CD24/metabolismo , Femenino , Integrina alfa6/metabolismo , Ratones
19.
J Exp Biol ; 209(Pt 11): 2215-23, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16709922

RESUMEN

A better knowledge of the regulatory mechanisms involved in stem cell proliferation and/or differentiation could reveal new methods for the treatment of some diseases. Most previous studies in the field of stem cell biology have been carried out on cultured isolated cells. In the case of adult tissue stem cells, mesenchymal bone marrow derived cells have been most widely studied, while the undifferentiated stem cells present in the epithelial tissues are less known. In order to advance further our understanding of epithelial tissue stem cells, new in vivo models are required. The present study focuses on the dynamics of a new and simple model of intestinal epithelial regeneration found in the midgut of the migratory locust, Locusta migratoria (Linnaeus 1758). The locust midgut consists of three cell types: columnar cells, endocrine cells and undifferentiated regenerative clustered cells. The undifferentiated epithelial midgut cells give rise to two other cell types and are located in a nest of regenerative cells known as regenerative niche. We have performed single and continuous bromodeoxyuridine (BrdU) administration experiments to study regeneration niches and their cellular dynamics. Immunocytochemistry and immunofluorescence techniques were used to detect the incorporation of BrdU into regenerative niches and the presence of FMRFamide-like immunoreactivity, as a marker for endocrine cell differentiation. Some isolated label retaining cells (LRC) were observed at the niche base 10-15 days after the final BrdU administration. We propose that these cells are the stem cells of this epithelial tissue. We also calculated the length of the cell cycle phases for a subpopulation of transit amplifying cells within the regenerative niche: G1, 2.5+/-0.5 h; S, 5.5+/-0.5 h; G2, 0.75+/-0.25 h; M, 2.5+/-0.5 h. These amplifying cells will give rise to the columnar epithelial non-endocrine lineage. The differentiation of an endocrine cell from a niche stem cell occurs less frequently and thus leads to a lower proportion of endocrine cells as compared with epithelial columnar digestive cells (up to three endocrine cells per niche). Endocrine cell commitment seems to occur very early in the differentiation process within the niche, as double-labelled BrdU and FMRF endocrine cells have never been found. The only exception is the endocrine cells located in the ampullar region of the midgut, some of which show double immunostaining after long-term chronic BrdU injection. In summary, we have characterized a new and simple animal model of epithelial stem cell regeneration that may be useful for understanding the complex biological process that drives tissue renewal from undifferentiated and uncommitted progenitor cells.


Asunto(s)
Diferenciación Celular , Células Epiteliales/citología , Saltamontes/citología , Saltamontes/fisiología , Modelos Biológicos , Regeneración/fisiología , Células Madre/citología , Animales , Antimetabolitos/farmacología , Bromodesoxiuridina/farmacología , Diferenciación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Femenino , Tracto Gastrointestinal/citología , Saltamontes/anatomía & histología , Masculino , Regeneración/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...