Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Dev Dyn ; 244(10): 1215-48, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26197970

RESUMEN

BACKGROUND: During gastrulation, endoderm and mesoderm are specified from a bipotential precursor (endomesoderm) that is argued to be homologous across bilaterians. Spiralians also generate mesoderm from ectodermal precursors (ectomesoderm), which arises near the blastopore. While a conserved gene regulatory network controls specification of endomesoderm in deuterostomes and ecdysozoans, little is known about genes controlling specification or behavior of either source of spiralian mesoderm or the digestive tract. RESULTS: Using the mollusc Crepidula, we examined conserved regulatory factors and compared their expression to fate maps to score expression in the germ layers, blastopore lip, and digestive tract. Many genes were expressed in both ecto- and endomesoderm, but only five were expressed in ectomesoderm exclusively. The latter may contribute to epithelial-to-mesenchymal transition seen in ectomesoderm. CONCLUSIONS: We present the first comparison of genes expressed during spiralian gastrulation in the context of high-resolution fate maps. We found variation of genes expressed in the blastopore lip, mouth, and cells that will form the anus. Shared expression of many genes in both mesodermal sources suggests that components of the conserved endomesoderm program were either co-opted for ectomesoderm formation or that ecto- and endomesoderm are derived from a common mesodermal precursor that became subdivided into distinct domains during evolution.


Asunto(s)
Gastrulación , Genes Reguladores , Caracoles/embriología , Animales , Expresión Génica , Estratos Germinativos/metabolismo , Organogénesis , Caracoles/genética , Caracoles/metabolismo
2.
Stem Cells ; 32(7): 1831-42, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24519994

RESUMEN

Adipose-derived stromal/stem cells (ASCs) ameliorate hyperglycemia in rodent models of islet transplantation and autoimmune diabetes, yet the precise human ASC (hASC)-derived factors responsible for these effects remain largely unexplored. Here, we show that systemic administration of hASCs improved glucose tolerance, preserved ß cell mass, and increased ß cell proliferation in streptozotocin-treated nonobese diabetic/severe combined immunodeficient mice. Coculture experiments combining mouse or human islets with hASCs demonstrated that islet viability and function were improved by hASCs following prolonged culture or treatment with proinflammatory cytokines. Analysis of hASC-derived factors revealed vascular endothelial growth factor and tissue inhibitor of metalloproteinase 1 (TIMP-1) to be highly abundant factors secreted by hASCs. Notably, TIMP-1 secretion increased in the presence of islet stress from cytokine treatment, while TIMP-1 blockade was able to abrogate in vitro prosurvival effects of hASCs. Following systemic administration by tail vein injection, hASCs were detected in the pancreas and human TIMP-1 was increased in the serum of injected mice, while recombinant TIMP-1 increased viability in INS-1 cells treated with interleukin-1beta, interferon-gamma, and tumor necrosis factor alpha. In aggregate, our data support a model whereby factors secreted by hASCs, such as TIMP-1, are able to mitigate against ß cell death in rodent and in vitro models of type 1 diabetes through a combination of local paracrine as well as systemic effects.


Asunto(s)
Células Madre Adultas/trasplante , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Hiperglucemia/terapia , Grasa Subcutánea/citología , Adulto , Células Madre Adultas/metabolismo , Animales , Tamaño de la Célula , Células Cultivadas , Técnicas de Cocultivo , Citocinas/fisiología , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Tipo 1/inducido químicamente , Femenino , Intolerancia a la Glucosa , Humanos , Hiperglucemia/inducido químicamente , Células Secretoras de Insulina/patología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Comunicación Paracrina , Estreptozocina , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
3.
Cancer Immunol Res ; 9(1): 89-102, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33093219

RESUMEN

Increased regulatory T cells (Treg) after radiotherapy have been reported, but the mechanisms of their induction remain incompletely understood. TGFß is known to foster Treg differentiation within tumors and is activated following radiotherapy. Thus, we hypothesized that TGFß blockade would result in decreased Tregs within the irradiated tumor microenvironment. We found increased Tregs in the tumors of mice treated with focal radiotherapy and TGFß blockade. This increase was mediated by upregulation of another TGFß family member, activin A. In vitro, activin A secretion was increased following irradiation of mouse and human breast cancer cells, and its expression was further enhanced upon TGFß blockade. In vivo, dual blockade of activin A and TGFß was required to decrease intratumoral Tregs in the context of radiotherapy. This resulted in an increase in CD8+ T-cell priming and was associated with a reduced tumor recurrence rate. Combination of immune checkpoint inhibitors with the dual blockade of activin A and TGFß led to the development of tumor-specific memory responses in irradiated breast cancer. Supporting the translational value of activin A targeting to reduce Treg-mediated immunosuppression, retrospective analysis of a public dataset of patients with breast cancer revealed a positive correlation between activin A gene expression and Treg abundance. Overall, these results shed light on an immune escape mechanism driven by activin A and suggest that dual targeting of activin A and TGFß may be required to optimally unleash radiation-induced antitumor immunity against breast cancer.


Asunto(s)
Activinas/metabolismo , Neoplasias de la Mama/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Terapia de Inmunosupresión , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Recurrencia Local de Neoplasia , Microambiente Tumoral
4.
Cancer Immunol Res ; 6(8): 910-920, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29907693

RESUMEN

Radiotherapy (RT) used at immunogenic doses leads to accumulation of cytosolic double-stranded DNA (dsDNA) in cancer cells, which activates type I IFN (IFN-I) via the cGAS/STING pathway. Cancer cell-derived IFN-I is required to recruit BATF3-dependent dendritic cells (DC) to poorly immunogenic tumors and trigger antitumor T-cell responses in combination with immune checkpoint blockade. We have previously demonstrated that the exonuclease TREX1 regulates radiation immunogenicity by degrading cytosolic dsDNA. Tumor-derived DNA can also activate cGAS/STING-mediated production of IFN-I by DCs infiltrating immunogenic tumors. However, how DNA from cancer cells is transferred to the cytoplasm of DCs remains unclear. Here, we showed that tumor-derived exosomes (TEX) produced by irradiated mouse breast cancer cells (RT-TEX) transfer dsDNA to DCs and stimulate DC upregulation of costimulatory molecules and STING-dependent activation of IFN-I. In vivo, RT-TEX elicited tumor-specific CD8+ T-cell responses and protected mice from tumor development significantly better than TEX from untreated cancer cells in a prophylactic vaccination experiment. We demonstrated that the IFN-stimulatory dsDNA cargo of RT-TEX is regulated by TREX1 expression in the parent cells. Overall, these results identify RT-TEX as a mechanism whereby IFN-stimulatory dsDNA is transferred from irradiated cancer cells to DCs. We have previously shown that the expression of TREX1 is dependent on the RT dose size. Thus, these data have important implications for the use of RT with immunotherapy. Cancer Immunol Res; 6(8); 910-20. ©2018 AACR.


Asunto(s)
ADN de Neoplasias/inmunología , Células Dendríticas/inmunología , Exodesoxirribonucleasas/inmunología , Exosomas/genética , Neoplasias Mamarias Animales/inmunología , Fosfoproteínas/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Exosomas/inmunología , Femenino , Interferón Tipo I/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/prevención & control , Neoplasias Mamarias Animales/radioterapia , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Bazo/inmunología , Células Tumorales Cultivadas
5.
Nat Commun ; 8: 15618, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28598415

RESUMEN

Radiotherapy is under investigation for its ability to enhance responses to immunotherapy. However, the mechanisms by which radiation induces anti-tumour T cells remain unclear. We show that the DNA exonuclease Trex1 is induced by radiation doses above 12-18 Gy in different cancer cells, and attenuates their immunogenicity by degrading DNA that accumulates in the cytosol upon radiation. Cytosolic DNA stimulates secretion of interferon-ß by cancer cells following activation of the DNA sensor cGAS and its downstream effector STING. Repeated irradiation at doses that do not induce Trex1 amplifies interferon-ß production, resulting in recruitment and activation of Batf3-dependent dendritic cells. This effect is essential for priming of CD8+ T cells that mediate systemic tumour rejection (abscopal effect) in the context of immune checkpoint blockade. Thus, Trex1 is an upstream regulator of radiation-driven anti-tumour immunity. Trex1 induction may guide the selection of radiation dose and fractionation in patients treated with immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Exodesoxirribonucleasas/metabolismo , Exodesoxirribonucleasas/efectos de la radiación , Neoplasias Mamarias Animales/genética , Neoplasias/inmunología , Neoplasias/radioterapia , Fosfoproteínas/metabolismo , Fosfoproteínas/efectos de la radiación , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Células Dendríticas/efectos de la radiación , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Inmunoterapia/métodos , Interferón beta/metabolismo , Interferón beta/efectos de la radiación , Neoplasias Mamarias Animales/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Nucleotidiltransferasas/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Proteínas Represoras/inmunología
6.
Cancer Res ; 75(11): 2232-42, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25858148

RESUMEN

T cells directed to endogenous tumor antigens are powerful mediators of tumor regression. Recent immunotherapy advances have identified effective interventions to unleash tumor-specific T-cell activity in patients who naturally develop them. Eliciting T-cell responses to a patient's individual tumor remains a major challenge. Radiation therapy can induce immune responses to model antigens expressed by tumors, but it remains unclear whether it can effectively prime T cells specific for endogenous antigens expressed by poorly immunogenic tumors. We hypothesized that TGFß activity is a major obstacle hindering the ability of radiation to generate an in situ tumor vaccine. Here, we show that antibody-mediated TGFß neutralization during radiation therapy effectively generates CD8(+) T-cell responses to multiple endogenous tumor antigens in poorly immunogenic mouse carcinomas. Generated T cells were effective at causing regression of irradiated tumors and nonirradiated lung metastases or synchronous tumors (abscopal effect). Gene signatures associated with IFNγ and immune-mediated rejection were detected in tumors treated with radiation therapy and TGFß blockade in combination but not as single agents. Upregulation of programmed death (PD) ligand-1 and -2 in neoplastic and myeloid cells and PD-1 on intratumoral T cells limited tumor rejection, resulting in rapid recurrence. Addition of anti-PD-1 antibodies extended survival achieved with radiation and TGFß blockade. Thus, TGFß is a fundamental regulator of radiation therapy's ability to generate an in situ tumor vaccine. The combination of local radiation therapy with TGFß neutralization offers a novel individualized strategy for vaccinating patients against their tumors.


Asunto(s)
Neoplasias de la Mama/inmunología , Inmunoterapia , Recurrencia Local de Neoplasia/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Anticuerpos Neutralizantes/uso terapéutico , Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/patología , Neoplasias de la Mama/radioterapia , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Recurrencia Local de Neoplasia/radioterapia , Linfocitos T Citotóxicos/inmunología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA