Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 11(1): 609, 2020 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-32001710

RESUMEN

Tumor-associated macrophages affect tumor progression and resistance to immune checkpoint therapy. Here, we identify the chemokine signal regulator FROUNT as a target to control tumor-associated macrophages. The low level FROUNT expression in patients with cancer correlates with better clinical outcomes. Frount-deficiency markedly reduces tumor progression and decreases macrophage tumor-promoting activity. FROUNT is highly expressed in macrophages, and its myeloid-specific deletion impairs tumor growth. Further, the anti-alcoholism drug disulfiram (DSF) acts as a potent inhibitor of FROUNT. DSF interferes with FROUNT-chemokine receptor interactions via direct binding to a specific site of the chemokine receptor-binding domain of FROUNT, leading to inhibition of macrophage responses. DSF monotherapy reduces tumor progression and decreases macrophage tumor-promoting activity, as seen in the case of Frount-deficiency. Moreover, co-treatment with DSF and an immune checkpoint antibody synergistically inhibits tumor growth. Thus, inhibition of FROUNT by DSF represents a promising strategy for macrophage-targeted cancer therapy.


Asunto(s)
Cadenas Pesadas de Clatrina/metabolismo , Disulfiram/farmacología , Neoplasias Pulmonares/patología , Macrófagos/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Quimiocinas/metabolismo , Progresión de la Enfermedad , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inmunoterapia , Cinética , Neoplasias Pulmonares/genética , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Metástasis de la Neoplasia , Proteínas de Complejo Poro Nuclear/genética , Pronóstico , Factores de Riesgo
2.
J Immunother Cancer ; 7(1): 21, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30696484

RESUMEN

BACKGROUND: Transient depletion of CD4+ T cells results in tumor suppression and survival benefit in murine models; however, the tumor progression and recurrence still occur over more long-term monitoring of mice. Thus, we explored an additional strategy to enhance endogenous immune responses by an alarmin, high mobility group nucleosome binding protein 1 (HMGN1). METHODS: The anti-tumor effects of HMGN1, anti-CD4 depleting antibody, and their combined treatment were monitored in the Colon26 or the B16F10 subcutaneous murine models. The tumor-infiltrating CD8+ T cell proliferation, differentiation, exhaustion, and its gene expression were determined by flow cytometry, transcriptome analysis, and quantitative real-time PCR. RESULTS: Our results show that a systemic administration of low doses of HMGN1 with an anti-CD4 depleting antibody (HMGN1/αCD4) promoted expansion of CD8+ T cell populations (e.g. CD137+ PD-1+ and CD44hi PD-1+), recruited CCR7+ migratory dendritic cells to the tumor, and reduced co-inhibitory molecules (e.g. PD-1, LAG-3, and TIM-3) to counteract CD8+ T cell exhaustion. CONCLUSION: The HMGN1/αCD4 treatment expanded effector CD8+ T cells and prolonged their anti-tumor activities by rescuing them from exhaustion, thus resulting in tumor regression and even rejection in long-term monitored mice.


Asunto(s)
Anticuerpos/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteína HMGN1/uso terapéutico , Neoplasias/terapia , Animales , Línea Celular Tumoral , Femenino , Proteína HMGN1/genética , Inmunoterapia , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/inmunología , Proteínas Recombinantes/uso terapéutico
3.
Sci Rep ; 8(1): 16642, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30413725

RESUMEN

Lung fibroblasts play a pivotal role in pulmonary fibrosis, a devastating lung disease, by producing extracellular matrix. MicroRNAs (miRNAs) suppress numerous genes post-transcriptionally; however, the roles of miRNAs in activated fibroblasts in fibrotic lungs remain poorly understood. To elucidate these roles, we performed global miRNA-expression profiling of fibroblasts from bleomycin- and silica-induced fibrotic lungs and investigated the functions of miRNAs in activated lung fibroblasts. Clustering analysis of global miRNA-expression data identified miRNA signatures exhibiting increased expression during fibrosis progression. Among these signatures, we found that a miR-19a-19b-20a sub-cluster suppressed TGF-ß-induced activation of fibroblasts in vitro. Moreover, to elucidate whether fibroblast-specific intervention against the sub-cluster modulates pathogenic activation of fibroblasts in fibrotic lungs, we intratracheally transferred the sub-cluster-overexpressing fibroblasts into bleomycin-treated lungs. Global transcriptome analysis of the intratracheally transferred fibroblasts revealed that the sub-cluster not only downregulated expression of TGF-ß-associated pro-fibrotic genes, including Acta2, Col1a1, Ctgf, and Serpine1, but also upregulated expression of the anti-fibrotic genes Dcn, Igfbp5, and Mmp3 in activated lung fibroblasts. Collectively, these findings indicated that upregulation of the miR-19a-19b-20a sub-cluster expression in lung fibroblasts counteracted TGF-ß-associated pathogenic activation of fibroblasts in murine pulmonary fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/metabolismo , MicroARNs/genética , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Proliferación Celular , Células Cultivadas , Femenino , Fibroblastos/citología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/citología , Ratones , Ratones Endogámicos C57BL , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/genética
4.
Toxicol Lett ; 265: 140-146, 2017 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-27916735

RESUMEN

INTRODUCTION: The mechanisms underlying the effects of cigarette smoke and smoking cessation on respiratory secretion, especially in the larynx, remain unclear. OBJECTIVES: The aims of this study were to determine the effects of cigarette smoke and smoking cessation on laryngeal mucus secretion and inflammation, and to investigate the effects of glucocorticoid administration. METHODS: We administered cigarette smoke solution (CSS) to eight-week-old male Sprague Dawley rats for four weeks, then examined laryngeal mucus secretion and inflammatory cytokine expression on days 1, 28 and 90 after smoking cessation. We also investigated the effects of the glucocorticoid triamcinolone acetonide when administered on day 1 after smoking cessation. RESULTS: Exposure to CSS resulted in an increase in laryngeal mucus secretion that was further excacerbated following smoking cessation. This change coincided with an increase in the expression of mRNA for the inflammatory cytokines tumor necrosis factor and interleukin-6, as well as mRNA for MUC5AC, which is involved in mucin production. Triamcinolone suppressed CSS-induced laryngeal mucus hypersecretion and pro-inflammatory cytokine production. CONCLUSION: Cigarette smoke-associated inflammation may contribute to the exacerbated laryngeal mucus hypersecretion that occurs following smoking cessation. The inflammatory response represents a promising target for the treatment of cigarette smoke-associated mucus hypersecretion.


Asunto(s)
Glucocorticoides/farmacología , Mucosa Laríngea/efectos de los fármacos , Moco/metabolismo , Cese del Hábito de Fumar , Triamcinolona/farmacología , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Glucocorticoides/administración & dosificación , Glucocorticoides/uso terapéutico , Mucosa Laríngea/inmunología , Mucosa Laríngea/metabolismo , Laringitis/tratamiento farmacológico , Laringitis/etiología , Laringitis/inmunología , Laringitis/metabolismo , Masculino , Ratas Sprague-Dawley , Fumar/efectos adversos , Triamcinolona/administración & dosificación , Triamcinolona/uso terapéutico
5.
Front Immunol ; 8: 1842, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29326717

RESUMEN

Chronic graft-versus-host disease (cGVHD) is a major complication in long-term survivors of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Graft-derived T cells (TG) have been implicated in the induction of cGVHD; however, the extent of their contribution to the pathogenesis of cGVHD remains unclear. Using a mouse model of cGVHD, we demonstrate that TG predominate over hematopoietic stem cell-derived T cells generated de novo (THSC) in cGVHD-affected organs such as the liver and lung even at day 63 after allo-HSCT. Persisting TG, in particular CD8+ TG, not only displayed an exhausted or senescent phenotype but also contained a substantial proportion of cells that had the potential to proliferate and produce inflammatory cytokines. Host antigens indirectly presented by donor HSC-derived hematopoietic cells were involved in the maintenance of TG in the reconstituted host. Selective depletion of TG in the chronic phase of disease resulted in the expansion of THSC and thus neither the survival nor histopathology of cGVHD was ameliorated. On the other hand, THSC depletion caused activation of TG and resulted in a lethal TG-mediated exacerbation of GVHD. The findings presented here clarify the pathological role of long-lasting TG in cGVHD.

6.
Sci Rep ; 6: 35002, 2016 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-27756896

RESUMEN

Foxp3+ regulatory T cells (Tregs) migrating from the skin to the draining lymph node (dLN) have a strong immunosuppressive effect on the cutaneous immune response. However, the subpopulations responsible for their inhibitory function remain unclear. We investigated single-cell gene expression heterogeneity in Tregs from the dLN of inflamed skin in a contact hypersensitivity model. The immunosuppressive genes Ctla4 and Tgfb1 were expressed in the majority of Tregs. Although Il10-expressing Tregs were rare, unexpectedly, the majority of Il10-expressing Tregs co-expressed Gzmb and displayed Th1-skewing. Single-cell profiling revealed that CD43+ CCR5+ Tregs represented the main subset within the Il10/Gzmb-expressing cell population in the dLN. Moreover, CD43+ CCR5+ CXCR3- Tregs expressed skin-tropic chemokine receptors, were preferentially retained in inflamed skin and downregulated the cutaneous immune response. The identification of a rare Treg subset co-expressing multiple immunosuppressive molecules and having tissue-remaining capacity offers a novel strategy for the control of skin inflammatory responses.


Asunto(s)
Dermatitis por Contacto/genética , Granzimas/genética , Interleucina-10/genética , Linfocitos T Reguladores/inmunología , Animales , Dermatitis por Contacto/inmunología , Modelos Animales de Enfermedad , Granzimas/metabolismo , Humanos , Interleucina-10/metabolismo , Leucosialina/metabolismo , Ratones , Receptores CCR5/metabolismo , Análisis de la Célula Individual , Piel
7.
Am J Pathol ; 185(11): 2923-38, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26456580

RESUMEN

Pulmonary fibrosis (PF) is an intractable disorder with a poor prognosis. Lung macrophages have been reported to regulate both progression and remission of bleomycin-induced diffuse PF. However, it remains unclear how macrophages contribute to silica-induced progressive nodular PF and the associated tissue cell responses in vivo. We found that lack of monocyte-derived macrophages results in the formation of diffuse PF after silica instillation. We found that the proportion and the number of monocyte-derived macrophages were persistently higher in silica-induced progressive PF compared with bleomycin-induced PF. Surprisingly, in Ccr2(-/-) mice, in which monocyte-derived macrophage infiltration is impaired, silica administration induced diffuse PF with loose nodule formation and greater activation of tissue cells. In the diffuse lesions, the distribution of epithelial cells, distribution of myofibroblasts, and architecture of the basement membrane were disrupted. Consistent with the development of diffuse lesions, genes that were differentially expressed in CD45(-) tissue cells from the lung of wild-type and Ccr2(-/-) mice were highly enriched in human diffuse, progressive PF. In gene ontology network analyses, many of these genes were associated with tissue remodeling and included genes not previously associated with PF, such as Mmp14, Thbs2, and Fgfr4. Overall, these results indicate that monocyte-derived macrophages prevent transition from nodular to diffuse silica-induced PF, potentially by regulating tissue cell responses.


Asunto(s)
Macrófagos Alveolares/patología , Fibrosis Pulmonar/patología , Receptores CCR2/metabolismo , Animales , Bleomicina/efectos adversos , Modelos Animales de Enfermedad , Células Epiteliales/patología , Perfilación de la Expresión Génica , Humanos , Hidroxiprolina/análisis , Pulmón/efectos de los fármacos , Pulmón/patología , Macrófagos Alveolares/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Monocitos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fibrosis Pulmonar/inducido químicamente , Receptores CCR2/genética , Dióxido de Silicio/efectos adversos
8.
Cancer Immunol Res ; 3(6): 631-40, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25711759

RESUMEN

Depletion of CD4(+) cells in tumor-bearing mice has strong antitumor effects. However, the mechanisms underlying these effects and the therapeutic benefits of CD4(+) cell depletion relative to other immunotherapies have not been fully evaluated. Here, we investigated the antitumor effects of an anti-CD4-depleting mAb as a monotherapy or in combination with immune checkpoint mAbs. In B16F10, Colon 26, or Lewis lung carcinoma subcutaneous tumor models, administration of the anti-CD4 mAb alone had strong antitumor effects that were superior to those elicited by CD25(+) Treg depletion or other immune checkpoint mAbs, and which were completely reversed by CD8(+) cell depletion. CD4(+) cell depletion led to the proliferation of tumor-specific CD8(+) T cells in the draining lymph node and increased infiltration of PD-1(+)CD8(+) T cells into the tumor, with a shift toward type I immunity within the tumor. Combination treatment with the anti-CD4 mAb and immune checkpoint mAbs, particularly anti-PD-1 or anti-PD-L1 mAbs, synergistically suppressed tumor growth and greatly prolonged survival. To our knowledge, this work represents the first report of robust synergy between anti-CD4 and anti-PD-1 or anti-PD-L1 mAb therapies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Antígenos CD4 , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/administración & dosificación , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Depleción Linfocítica , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Melanoma Experimental , Ratones , Transducción de Señal/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
9.
J Immunol ; 194(1): 398-406, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25422510

RESUMEN

Graft-versus-host disease (GVHD) is a major risk factor for prolonged humoral immunodeficiency and vaccine unresponsiveness after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, the underlying mechanisms for this immunodeficiency are poorly understood. In this article, we describe previously overlooked impacts of GVHD on lymph node (LN) stromal cells involved in humoral immune responses. In major- and minor-mismatched mouse allo-HSCT models, recipients with CD8(+) T cell-mediated GVHD suffered severe and irreversible damage to LN structure. These mice were susceptible to pathogenic infection and failed to mount humoral immune responses despite the presence of peripheral T and B cells. These humoral immune defects were associated with the early loss of fibroblastic reticular cells, most notably the CD157(+) cell subset, as well as structural defects in high endothelial venules. The disruption to these LN stromal cells was dependent on alloantigens expressed by nonhematopoietic cells. Blockade of the Fas-FasL pathway prevented damage to CD157(+) fibroblastic reticular cells and ameliorated LN GVHD. However, blockade of CD62L- or CCR7-dependent migration of CD8(+) T cells to the LN was insufficient to prevent stromal cell injury. Overall, our results highlight GVHD-associated loss of functional stromal cells and LN GVHD as a possible explanation for the prolonged susceptibility to infectious disease that is experienced by allo-HSCT patients.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas , Ganglios Linfáticos/patología , ADP-Ribosil Ciclasa/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Antígenos CD/metabolismo , Movimiento Celular/inmunología , Células Cultivadas , Toxina del Cólera/inmunología , Células Endoteliales/citología , Proteína Ligando Fas/antagonistas & inhibidores , Proteínas Ligadas a GPI/metabolismo , Inmunidad Humoral/inmunología , Síndromes de Inmunodeficiencia/inmunología , Selectina L/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Receptores CCR7/antagonistas & inhibidores , Receptores CCR7/genética , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Células del Estroma/inmunología , Trasplante Homólogo , Receptor fas/antagonistas & inhibidores
10.
Sci Rep ; 4: 6030, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-25112380

RESUMEN

Skin-derived dendritic cells (DCs) play a crucial role in the maintenance of immune homeostasis due to their role in antigen trafficking from the skin to the draining lymph nodes (dLNs). To quantify the spatiotemporal regulation of skin-derived DCs in vivo, we generated knock-in mice expressing the photoconvertible fluorescent protein KikGR. By exposing the skin or dLN of these mice to violet light, we were able to label and track the migration and turnover of endogenous skin-derived DCs. Langerhans cells and CD103(+)DCs, including Langerin(+)CD103(+)dermal DCs (DDCs), remained in the dLN for 4-4.5 days after migration from the skin, while CD103(-)DDCs persisted for only two days. Application of a skin irritant (chemical stress) induced a transient >10-fold increase in CD103(-)DDC migration from the skin to the dLN. Tape stripping (mechanical injury) induced a long-lasting four-fold increase in CD103(-)DDC migration to the dLN and accelerated the trafficking of exogenous protein antigens by these cells. Both stresses increased the turnover of CD103(-)DDCs within the dLN, causing these cells to die within one day of arrival. Therefore, CD103(-)DDCs act as sentinels against skin invasion that respond with increased cellular migration and antigen trafficking from the skin to the dLNs.


Asunto(s)
Células Dendríticas/citología , Ganglios Linfáticos/citología , Piel/citología , Animales , Antígenos CD/metabolismo , Movimiento Celular , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Dermatitis Irritante/inmunología , Dermatitis Irritante/patología , Técnicas de Sustitución del Gen , Cadenas alfa de Integrinas/metabolismo , Células de Langerhans/citología , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Luz , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Proteínas/genética , Receptores CCR7/deficiencia , Receptores CCR7/genética , Receptores CCR7/metabolismo , Piel/inmunología , Piel/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(21): 7771-6, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24825888

RESUMEN

Myeloid cells such as monocytes and monocyte-derived macrophages promote tumor progression. Recent reports suggest that extramedullary hematopoiesis sustains a sizable reservoir of tumor-infiltrating monocytes in the spleen. However, the influence of the spleen on tumor development and the extent to which spleen monocytes populate the tumor relative to bone marrow (BM) monocytes remain controversial. Here, we used mice expressing the photoconvertible protein Kikume Green-Red to track the redistribution of monocytes from the BM and spleen, and mice expressing fluorescent ubiquitination-based cell-cycle indicator proteins to monitor active hematopoiesis in these tissues. In mice bearing late-stage tumors, the BM, besides being the major site of monocyte production, supplied the expansion of the spleen reservoir, replacing 9% of spleen monocytes every hour. Deployment of monocytes was equally rapid from the BM and the spleen. However, BM monocytes were younger than those in the spleen and were 2.7 times more likely to migrate into the tumor from the circulation. Partly as a result of this intrinsic difference in migration potential, spleen monocytes made only a minor contribution to the tumor-infiltrating monocyte population. At least 27% of tumor monocytes had traveled from the BM in the last 24 h, compared with only 2% from the spleen. These observations highlight the importance of the BM as the primary hematopoietic tissue and monocyte reservoir in tumor-bearing mice, despite the changes that occur in the spleen monocyte reservoir during tumor development.


Asunto(s)
Células de la Médula Ósea/inmunología , Carcinogénesis/inmunología , Movimiento Celular/inmunología , Hematopoyesis/fisiología , Monocitos/inmunología , Bazo/citología , Análisis de Varianza , Animales , Fluorescencia , Masculino , Ratones , Ratones Endogámicos C57BL , Bazo/inmunología
12.
Am J Pathol ; 183(3): 758-73, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23886891

RESUMEN

Pulmonary fibrosis is characterized by accumulation of activated fibroblasts that produce excessive amounts of extracellular matrix components such as collagen type I. However, the dynamics and activation signatures of fibroblasts during fibrogenesis remain poorly understood, especially in vivo. We examined changes in lung tissue cell populations and in the phenotype of activated fibroblasts after acute injury in a model of bleomycin-induced pulmonary fibrosis. Despite clustering of collagen type I-producing fibroblasts in fibrotic regions, flow cytometry-based quantitative analysis of whole lungs revealed that the number of fibroblasts in the lungs remained constant. At the peak of inflammation, fibroblast proliferation and apoptosis were both increased, suggesting that the clustering was not merely a result of proliferation, but also of fibroblast migration from nearby alveolar walls. Parabiosis experiments demonstrated that fibroblasts were not supplied from the circulation. Comprehensive gene expression analysis of freshly isolated fibroblasts revealed a detailed activation signature associated with fibrogenesis, including changes in genes responsible for migration and extracellular matrix construction. The Spp1 gene, which encodes osteopontin, was highly up-regulated and was an identifying characteristic of activated fibroblasts present at the sites of remodeling. Osteopontin may serve as a useful marker of profibrotic fibroblasts. These results provide insights into the cellular and molecular mechanisms underlying pulmonary fibrosis and provide a foundation for development of specific antifibrotic therapies.


Asunto(s)
Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Animales , Apoptosis , Biomarcadores/metabolismo , Bleomicina , Células de la Médula Ósea/patología , Proliferación Celular , Colágeno Tipo I/metabolismo , Perfilación de la Expresión Génica , Ontología de Genes , Proteínas Fluorescentes Verdes/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Osteopontina/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Regulación hacia Arriba/genética
13.
J Immunol ; 190(8): 4076-91, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23509353

RESUMEN

Memory CD4(+) T cells are central regulators of both humoral and cellular immune responses. T cell differentiation results in specific changes in chromatin structure and DNA methylation of cytokine genes. Although the methylation status of a limited number of gene loci in T cells has been examined, the genome-wide DNA methylation status of memory CD4(+) T cells remains unexplored. To further elucidate the molecular signature of memory T cells, we conducted methylome and transcriptome analyses of memory CD4(+) T cells generated using T cells from TCR-transgenic mice. The resulting genome-wide DNA methylation profile revealed 1144 differentially methylated regions (DMRs) across the murine genome during the process of T cell differentiation, 552 of which were associated with gene loci. Interestingly, the majority of these DMRs were located in introns. These DMRs included genes such as CXCR6, Tbox21, Chsy1, and Cish, which are associated with cytokine production, homing to bone marrow, and immune responses. Methylation changes in memory T cells exposed to specific Ag appeared to regulate enhancer activity rather than promoter activity of immunologically relevant genes. In addition, methylation profiles differed between memory T cell subsets, demonstrating a link between T cell methylation status and T cell differentiation. By comparing DMRs between naive and Ag-specific memory T cells, this study provides new insights into the functional status of memory T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Metilación de ADN/genética , Epítopos de Linfocito T/metabolismo , Memoria Inmunológica/genética , Animales , Linfocitos T CD4-Positivos/citología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Epítopos de Linfocito T/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Transcriptoma
14.
Front Immunol ; 3: 71, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22566952

RESUMEN

Organ fibrosis is a pathological condition associated with chronic inflammatory diseases. In fibrosis, excessive deposition of extracellular matrix (ECM) severely impairs tissue architecture and function, eventually resulting in organ failure. This process is mediated primarily by the induction of myofibroblasts, which produce large amounts of collagen I, the main component of the ECM. Accordingly, the origin, developmental pathways, and mechanisms of myofibroblast regulation are attracting increasing attention as potential therapeutic targets. The fibrotic cascade, from initial epithelial damage to eventual myofibroblast induction, is mediated by complex biological processes such as macrophage infiltration, a shift from Th1 to Th2 phenotype, and by inflammatory mediators such as transforming growth factor-ß. Here, we review the current understanding of the cellular and molecular mechanisms underlying organ fibrosis.

15.
Int Immunopharmacol ; 11(7): 783-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21406269

RESUMEN

Tumor growth is often associated with the aberrant systemic accumulation of myeloid-derived suppressor cells (MDSCs), which are a heterogenous population of cells composed of polymorphonuclear neutrophils, monocytes, macrophages, dendritic cells and early myeloid precursors. These MDSCs are thought to suppress anti-tumor T cell responses in both tumor tissues and secondary lymphoid tissues. Accumulation of MDSCs in these target tissues is a dynamic process associated with medullary and extramedullary myelopoiesis and subsequent cellular migration. Here, we review the current understanding of the cellular, molecular, hematological and anatomical principles of MDSC development and migration in tumor-bearing mice. We also discuss the therapeutic potential of chemokines that influence the balance between MDSC subpopulations.


Asunto(s)
Quimiocinas/inmunología , Células Mieloides/inmunología , Mielopoyesis , Neoplasias Experimentales/inmunología , Linfocitos T/inmunología , Animales , Comunicación Celular , Linaje de la Célula , Movimiento Celular , Humanos , Terapia de Inmunosupresión , Ratones , Células Mieloides/patología , Neoplasias Experimentales/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...