Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 24(11): 1867-1878, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37798557

RESUMO

The capacity to survive and thrive in conditions of limited resources and high inflammation is a major driver of tumor malignancy. Here we identified slow-cycling ADAM12+PDGFRα+ mesenchymal stromal cells (MSCs) induced at the tumor margins in mouse models of melanoma, pancreatic cancer and prostate cancer. Using inducible lineage tracing and transcriptomics, we demonstrated that metabolically altered ADAM12+ MSCs induced pathological angiogenesis and immunosuppression by promoting macrophage efferocytosis and polarization through overexpression of genes such as Gas6, Lgals3 and Csf1. Genetic depletion of ADAM12+ cells restored a functional tumor vasculature, reduced hypoxia and acidosis and normalized CAFs, inducing infiltration of effector T cells and growth inhibition of melanomas and pancreatic neuroendocrine cancer, in a process dependent on TGF-ß. In human cancer, ADAM12 stratifies patients with high levels of hypoxia and innate resistance mechanisms, as well as factors associated with a poor prognosis and drug resistance such as AXL. Altogether, our data show that depletion of tumor-induced slow-cycling PDGFRα+ MSCs through ADAM12 restores antitumor immunity.


Assuntos
Células-Tronco Mesenquimais , Neoplasias , Masculino , Camundongos , Animais , Humanos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptores Proteína Tirosina Quinases , Macrófagos , Hipóxia , Linhagem Celular Tumoral , Proteína ADAM12/genética
2.
Immunity ; 48(1): 120-132.e8, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29343433

RESUMO

Group 3 innate lymphoid cells (ILC3s) sense environmental signals and are critical for tissue integrity in the intestine. Yet, which signals are sensed and what receptors control ILC3 function remain poorly understood. Here, we show that ILC3s with a lymphoid-tissue-inducer (LTi) phenotype expressed G-protein-coupled receptor 183 (GPR183) and migrated to its oxysterol ligand 7α,25-hydroxycholesterol (7α,25-OHC). In mice lacking Gpr183 or 7α,25-OHC, ILC3s failed to localize to cryptopatches (CPs) and isolated lymphoid follicles (ILFs). Gpr183 deficiency in ILC3s caused a defect in CP and ILF formation in the colon, but not in the small intestine. Localized oxysterol production by fibroblastic stromal cells provided an essential signal for colonic lymphoid tissue development, and inflammation-induced increased oxysterol production caused colitis through GPR183-mediated cell recruitment. Our findings show that GPR183 promotes lymphoid organ development and indicate that oxysterol-GPR183-dependent positioning within tissues controls ILC3 activity and intestinal homeostasis.


Assuntos
Colite/metabolismo , Linfócitos/metabolismo , Tecido Linfoide/metabolismo , Oxisteróis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Movimento Celular/genética , Colite/imunologia , Colite/patologia , Colo/imunologia , Colo/patologia , Citocinas/metabolismo , Citometria de Fluxo , Imunofluorescência , Ligantes , Linfócitos/patologia , Tecido Linfoide/patologia , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais
3.
Nucleic Acids Res ; 47(7): 3407-3421, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30753595

RESUMO

The proper tissue-specific regulation of gene expression is essential for development and homeostasis in metazoans. However, the illegitimate expression of normally tissue-restricted genes-like testis- or placenta-specific genes-is frequently observed in tumors; this promotes transformation, but also allows immunotherapy. Two important questions are: how is the expression of these genes controlled in healthy cells? And how is this altered in cancer? To address these questions, we used an unbiased approach to test the ability of 350 distinct genetic or epigenetic perturbations to induce the illegitimate expression of over 40 tissue-restricted genes in primary human cells. We find that almost all of these genes are remarkably resistant to reactivation by a single alteration in signaling pathways or chromatin regulation. However, a few genes differ and are more readily activated; one is the placenta-expressed gene ADAM12, which promotes invasion. Using cellular systems, an animal model, and bioinformatics, we find that a non-canonical but druggable TGF-ß/KAT2A/TAK1 axis controls ADAM12 induction in normal and cancer cells. More broadly, our data show that illegitimate gene expression in cancer is an heterogeneous phenomenon, with a few genes activatable by simple events, and most genes likely requiring a combination of events to become reactivated.


Assuntos
Regulação da Expressão Gênica/genética , Neoplasias/genética , Especificidade de Órgãos/genética , Transcrição Gênica/genética , Proteína ADAM12/genética , Proteína ADAM12/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Histona Acetiltransferases/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Transdução de Sinais/genética , Fator de Crescimento Transformador beta1/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo
4.
Proc Natl Acad Sci U S A ; 114(4): E506-E513, 2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-28074039

RESUMO

The intestinal epithelium is continuously renewed by intestinal epithelial stem cells (IESCs) positioned at the base of each crypt. Mesenchymal-derived factors are essential to maintain IESCs; however, the cellular composition and development of such mesenchymal niche remains unclear. Here, we identify pericryptal CD34+ Gp38+ αSMA- mesenchymal cells closely associated with Lgr5+ IESCs. We demonstrate that CD34+ Gp38+ cells are the major intestinal producers of the niche factors Wnt2b, Gremlin1, and R-spondin1, and are sufficient to promote maintenance of Lgr5+ IESCs in intestinal organoids, an effect mainly mediated by Gremlin1. CD34+ Gp38+ cells develop after birth in the intestinal submucosa and expand around the crypts during the third week of life in mice, independently of the microbiota. We further show that pericryptal CD34+gp38+ cells are rapidly activated by intestinal injury, up-regulating niche factors Gremlin1 and R-spondin1 as well as chemokines, proinflammatory cytokines, and growth factors with key roles in gut immunity and tissue repair, including IL-7, Ccl2, Ptgs2, and Amphiregulin. Our results indicate that CD34+ Gp38+ mesenchymal cells are programmed to develop in the intestine after birth to constitute a specialized microenvironment that maintains IESCs at homeostasis and contribute to intestinal inflammation and repair after injury.


Assuntos
Antígenos CD34/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Nicho de Células-Tronco , Animais , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Sulfato de Dextrana , Homeostase , Mucosa Intestinal/citologia , Camundongos Endogâmicos C57BL
5.
Int J Cancer ; 139(6): 1358-71, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27130719

RESUMO

Enhancing anti-tumor immunity and preventing tumor escape are efficient strategies to increase the efficacy of therapeutic cancer vaccines. However, the treatment of advanced tumors remains difficult, mainly due to the immunosuppressive tumor microenvironment. Regulatory T cells and myeloid-derived suppressor cells have been extensively studied, and their role in suppressing tumor immunity is now well established. In contrast, the role of B lymphocytes in tumor immunity remains unclear because B cells can promote tumor immunity or display regulatory functions to control excessive inflammation, mainly through IL-10 secretion. Here, in a mouse model of HPV-related cancer, we demonstrate that B cells accumulated in the draining lymph node of tumor-bearing mice, due to a prolonged survival, and showed a decreased expression of MHC class II and CD86 molecules and an increased expression of Ly6A/E, PD-L1 and CD39, suggesting potential immunoregulatory properties. However, B cells from tumor-bearing mice did not show an increased ability to secrete IL-10 and a deficiency in IL-10 production did not impair tumor growth. In contrast, in B cell-deficient µMT mice, tumor rejection occurred due to a strong T cell-dependent anti-tumor response. Genetic analysis based on single nucleotide polymorphisms identified genetic variants associated with tumor rejection in µMT mice, which could potentially affect reactive oxygen species production and NK cell activity. Our results demonstrate that B cells play a detrimental role in anti-tumor immunity and suggest that targeting B cells could enhance the anti-tumor response and improve the efficacy of therapeutic cancer vaccines.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Infecções por Papillomavirus/complicações , Neoplasias do Colo do Útero/etiologia , Neoplasias do Colo do Útero/metabolismo , Animais , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Subpopulações de Linfócitos B/patologia , Linfócitos B/patologia , Movimento Celular/imunologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Estudo de Associação Genômica Ampla , Interleucina-10/biossíntese , Linfonodos/imunologia , Linfonodos/patologia , Ativação Linfocitária , Camundongos , Papillomaviridae , Infecções por Papillomavirus/virologia , Fenótipo , Polimorfismo de Nucleotídeo Único , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Receptor Toll-Like 9/metabolismo , Neoplasias do Colo do Útero/patologia
6.
Mucosal Immunol ; 16(2): 221-231, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36708806

RESUMO

The intestinal barrier is a complex structure that allows the absorption of nutrients while ensuring protection against intestinal pathogens and balanced immunity. The development and maintenance of a functional intestinal barrier is a multifactorial process that is only partially understood. Here we review novel findings on the emerging role of mesenchymal cells in this process using insights gained from lineage tracing approaches, Cre-based gene deletion, and single-cell transcriptomics. The current evidence points toward a key organizer role for distinct mesenchymal lineages in intestinal development and homeostasis, regulating both epithelial and immune components of the intestinal barrier. We further discuss recent findings on functional mesenchymal heterogeneity and implications for intestinal regeneration and inflammatory intestinal pathologies.


Assuntos
Intestinos , Células-Tronco Mesenquimais , Intestinos/patologia , Mucosa Intestinal
7.
Sci Rep ; 13(1): 14960, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37696912

RESUMO

In this work, we investigated the oncogenic role of Streptococcus gallolyticus subsp. gallolyticus (SGG), a gut bacterium associated with colorectal cancer (CRC). We showed that SGG UCN34 accelerates colon tumor development in a chemically induced CRC murine model. Full proteome and phosphoproteome analysis of murine colons chronically colonized by SGG UCN34 revealed that 164 proteins and 725 phosphorylation sites were differentially regulated. Ingenuity Pathway Analysis (IPA) indicates a pro-tumoral shift specifically induced by SGG UCN34, as ~ 90% of proteins and phosphoproteins identified were associated with digestive cancer. Comprehensive analysis of the altered phosphoproteins using ROMA software revealed up-regulation of several cancer hallmark pathways such as MAPK, mTOR and integrin/ILK/actin, affecting epithelial and stromal colonic cells. Importantly, an independent analysis of protein arrays of human colon tumors colonized with SGG showed up-regulation of PI3K/Akt/mTOR and MAPK pathways, providing clinical relevance to our findings. To test SGG's capacity to induce pre-cancerous transformation of the murine colonic epithelium, we grew ex vivo organoids which revealed unusual structures with compact morphology. Taken together, our results demonstrate the oncogenic role of SGG UCN34 in a murine model of CRC associated with activation of multiple cancer-related signaling pathways.


Assuntos
Neoplasias do Colo , Streptococcus gallolyticus subspecies gallolyticus , Humanos , Animais , Camundongos , Modelos Animais de Doenças , Fosfatidilinositol 3-Quinases , Proteômica , Serina-Treonina Quinases TOR , Fosfoproteínas , Proteoma , Transdução de Sinais
8.
Proc Natl Acad Sci U S A ; 106(5): 1512-7, 2009 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-19164539

RESUMO

The thymus represents the "cradle" for T cell development, with thymic stroma providing multiple soluble and membrane cues to developing thymocytes. Although IL-7 is recognized as an essential factor for thymopoiesis, the "environmental niche" of thymic IL-7 activity remains poorly characterized in vivo. Using bacterial artificial chromosome transgenic mice in which YFP is under control of IL-7 promoter, we identify a subset of thymic epithelial cells (TECs) that co-express YFP and high levels of Il7 transcripts (IL-7(hi) cells). IL-7(hi) TECs arise during early fetal development, persist throughout life, and co-express homeostatic chemokines (Ccl19, Ccl25, Cxcl12) and cytokines (Il15) that are critical for normal thymopoiesis. In the adult thymus, IL-7(hi) cells localize to the cortico-medullary junction and display traits of both cortical and medullary TECs. Interestingly, the frequency of IL-7(hi) cells decreases with age, suggesting a mechanism for the age-related thymic involution that is associated with declining IL-7 levels. Our temporal-spatial analysis of IL-7-producing cells in the thymus in vivo suggests that thymic IL-7 levels are dynamically regulated under distinct physiological conditions. This IL-7 reporter mouse provides a valuable tool to further dissect the mechanisms that govern thymic IL-7 expression in vivo.


Assuntos
Interleucina-7/metabolismo , Timo/metabolismo , Animais , Células Cultivadas , Cromossomos Artificiais Bacterianos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Genes Reporter , Interleucina-7/genética , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Timo/citologia , Timo/imunologia
9.
Cell Stem Cell ; 29(5): 856-868.e5, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35523143

RESUMO

After birth, the intestine undergoes major changes to shift from an immature proliferative state to a functional intestinal barrier. By combining inducible lineage tracing and transcriptomics in mouse models, we identify a prodifferentiation PDGFRαHigh intestinal stromal lineage originating from postnatal LTßR+ perivascular stromal progenitors. The genetic blockage of this lineage increased the intestinal stem cell pool while decreasing epithelial and immune maturation at weaning age, leading to reduced postnatal growth and dysregulated repair responses. Ablating PDGFRα in the LTBR stromal lineage demonstrates that PDGFRα has a major impact on the lineage fate and function, inducing a transcriptomic switch from prostemness genes, such as Rspo3 and Grem1, to prodifferentiation factors, including BMPs, retinoic acid, and laminins, and on spatial organization within the crypt-villus and repair responses. Our results show that the PDGFRα-induced transcriptomic switch in intestinal stromal cells is required in the first weeks after birth to coordinate postnatal intestinal maturation and function.


Assuntos
Intestinos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Animais , Diferenciação Celular/fisiologia , Mecanismos de Defesa , Mucosa Intestinal , Receptor beta de Linfotoxina , Camundongos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Células-Tronco
10.
J Immunol ; 182(9): 5789-99, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380827

RESUMO

Stromal cells in lymphoid tissues regulate lymphocyte recruitment and survival through the expression of specific chemokines and cytokines. During inflammation, the same signals recruit lymphocytes to the site of injury; however, the "lymphoid" stromal (LS) cells producing these signals remain poorly characterized. We find that mouse inflammatory lesions and tumors develop gp38(+) LS cells, in recapitulation of the development of LS cells early during the ontogeny of lymphoid organs and the intestine, and express a set of genes that promotes the development of lymphocyte-permissive tissues. These gp38(+) LS cells are induced by a robust pathway that requires myeloid cells but not known Toll- or NOD-like receptors, the inflammasome, or adaptive immunity. Parabiosis and inducible genetic cell fate mapping experiments indicate that local precursors, presumably resident fibroblasts rather that circulating precursors, massively proliferate and give rise to LS cells during inflammation. Our results show that LS cells are both programmed during ontogeny and reinduced during inflammation.


Assuntos
Movimento Celular/imunologia , Inflamação/imunologia , Inflamação/patologia , Tecido Linfoide/imunologia , Tecido Linfoide/patologia , Animais , Quimiocinas/biossíntese , Citocinas/biossíntese , Inflamação/embriologia , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Mucosa Intestinal/embriologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Leishmania major/imunologia , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/patologia , Tecido Linfoide/embriologia , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Camundongos Transgênicos , Células Estromais/imunologia , Células Estromais/metabolismo , Células Estromais/patologia
11.
iScience ; 24(3): 102158, 2021 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-33665576

RESUMO

Innate lymphoid cells (ILCs) have been shown to be significantly affected in the small intestine lamina propria and secondary lymphoid organs (SLOs) of conventional lymphopenic mice. How ILCs are regulated by adaptive immunity in SLOs remains unclear. In T cell-deficient mice, ILC2s are significantly increased in the mesenteric lymph nodes (MLNs) at the expense of CCR6+ ILC3s, which are nonetheless increased in the peripheral lymph nodes (PLNs). Here, we show that T cells regulate lymph node-resident ILCs in a tissue- and subset-specific way. First, reducing microbial colonization from birth restored CCR6+ ILC3s in the MLNs of T cell-deficient mice. In contrast, T cell reconstitution resulted in the contraction of both MLNs ILC2s and PLNs ILC3s, whereas antagonizing microbial colonization from birth had no impact on these populations. Finally, the accumulation of MLNs ILC2s was partly regulated by T cells through stroma-derived IL-33.

12.
Curr Opin Immunol ; 64: 50-55, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32387900

RESUMO

The mesenchymal microenvironment is increasingly recognized as a major player in immunity. Here we focus on mesenchymal cells located within or in proximity to the blood vessels wall, which include pericytes, adventitial fibroblasts and mesenchymal stromal cells. We discuss recent evidence that these cells play a role in tissue homeostasis, immunity and inflammatory pathologies by multiple mechanisms, including vascular modulation, leucocyte migration, activation or survival in the perivascular space and differentiation into specialized 'effector' mesenchymal cells essential for tissue repair and immunity, such as myofibroblasts and lymphoid stromal cells. When dysregulated, these responses contribute to inflammatory and fibrotic diseases.


Assuntos
Células-Tronco Mesenquimais , Diferenciação Celular , Fibroblastos/fisiologia , Humanos , Células-Tronco Mesenquimais/fisiologia , Pericitos/patologia , Pericitos/fisiologia , Células Estromais
13.
J Clin Invest ; 128(1): 54-63, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29293094

RESUMO

The ability to repair tissues is essential for the survival of organisms. In chronic settings, the failure of the repair process to terminate results in overproduction of collagen, a pathology known as fibrosis, which compromises organ recovery and impairs function. The origin of the collagen-overproducing cell has been debated for years. Here we review recent insights gained from the use of lineage tracing approaches in several organs. The resulting evidence points toward specific subsets of tissue-resident mesenchymal cells, mainly localized in a perivascular position, as the major source for collagen-producing cells after injury. We discuss these findings in view of the functional heterogeneity of mesenchymal cells of the perivascular niche, which have essential vascular, immune, and regenerative functions that need to be preserved for efficient repair.


Assuntos
Colágeno/imunologia , Colágeno/metabolismo , Fibroblastos/imunologia , Fibroblastos/metabolismo , Células-Tronco Mesenquimais/imunologia , Animais , Vasos Sanguíneos/imunologia , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Fibroblastos/patologia , Fibrose , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia
14.
J Exp Med ; 215(11): 2936-2954, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30355616

RESUMO

The foodborne pathogen Listeria monocytogenes (Lm) crosses the intestinal villus epithelium via goblet cells (GCs) upon the interaction of Lm surface protein InlA with its receptor E-cadherin. Here, we show that Lm infection accelerates intestinal villus epithelium renewal while decreasing the number of GCs expressing luminally accessible E-cadherin, thereby locking Lm portal of entry. This novel innate immune response to an enteropathogen is triggered by the infection of Peyer's patch CX3CR1+ cells and the ensuing production of IL-23. It requires STAT3 phosphorylation in epithelial cells in response to IL-22 and IL-11 expressed by lamina propria gp38+ stromal cells. Lm-induced IFN-γ signaling and STAT1 phosphorylation in epithelial cells is also critical for Lm-associated intestinal epithelium response. GC depletion also leads to a decrease in colon mucus barrier thickness, thereby increasing host susceptibility to colitis. This study unveils a novel innate immune response to an enteropathogen, which implicates gp38+ stromal cells and locks intestinal villus invasion, but favors colitis.


Assuntos
Colite/imunologia , Mucosa Intestinal/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Glicoproteínas de Membrana/imunologia , Células Mieloides/imunologia , Nódulos Linfáticos Agregados/imunologia , Animais , Colite/genética , Colite/microbiologia , Colite/patologia , Citocinas/genética , Citocinas/imunologia , Imunidade Inata/genética , Imunidade nas Mucosas/genética , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Listeriose/genética , Listeriose/patologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Células Mieloides/microbiologia , Células Mieloides/patologia , Nódulos Linfáticos Agregados/microbiologia , Nódulos Linfáticos Agregados/patologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia , Células Estromais/imunologia , Células Estromais/microbiologia , Células Estromais/patologia
16.
Cancer Res ; 65(20): 9312-9, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16230393

RESUMO

ADAM9 is a membrane-anchored metalloprotease that is markedly up-regulated in several human carcinomas. Here, we show that ADAM9 is similarly up-regulated in mouse models for prostate, breast, and intestinal carcinoma. To assess whether ADAM9 is critical for the pathogenesis of prostate carcinoma, one of the most common cancers in men, we evaluated how loss of ADAM9 affects tumorigenesis in W(10) mice, a mouse model for this disease. In the absence of ADAM9, most tumors in 50-week-old W(10) mice were well differentiated, whereas littermate controls expressing wild-type ADAM9 had predominantly poorly differentiated, and in some cases significantly larger, tumors. Moreover, gain-of-function experiments in which ADAM9 was overexpressed in mouse prostate epithelium resulted in significant abnormalities, including epithelial hyperplasia at 4 to 6 months of age, and prostatic intraepithelial neoplasia after 1 year. A potential underlying mechanism for the role of ADAM9 in prostate cancer emerged from cell-based assays: ADAM9 can cleave and release epidermal growth factor and FGFR2iiib from cells, both of which have pivotal functions in the pathogenesis of this disease. Taken together, these results suggest that ADAM9 contributes to the pathogenesis of prostate cancer and potentially also other carcinomas, raising the possibility that ADAM9 might be a good target for antitumor drugs.


Assuntos
Proteínas ADAM/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias da Próstata/enzimologia , Proteínas ADAM/biossíntese , Proteínas ADAM/genética , Animais , Diferenciação Celular/fisiologia , Fator de Crescimento Epidérmico/metabolismo , Feminino , Fator 7 de Crescimento de Fibroblastos/metabolismo , Hibridização In Situ , Neoplasias Intestinais/enzimologia , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Masculino , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Regulação para Cima
17.
J Immunol Methods ; 421: 14-19, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25599879

RESUMO

Mesenchymal stromal cells in lymphoid organs, also called lymphoid stromal cells (LSCs), play a pivotal role in immunity by forming specialized microenvironments that provide signals for leukocyte migration, positioning, and survival. Best characterized in lymphoid organs, LSCs are also abundant in the intestinal mucosa, which harbors a rich repertoire of immune cells. However, the lack of efficient procedures for isolation and purification of LSCs from the intestine has been a major limitation to their characterization. Here we report a new method to efficiently isolate, in addition to immune cells, viable lymphoid stromal cells and other stromal subsets from the intestinal lamina propria for subsequent phenotypic and functional analysis.


Assuntos
Citometria de Fluxo/métodos , Imunofluorescência/métodos , Mucosa Intestinal/citologia , Células-Tronco Mesenquimais/citologia , Animais , Mucosa Intestinal/imunologia , Glicoproteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo
18.
Cell Host Microbe ; 13(6): 735-45, 2013 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-23768497

RESUMO

Neisseria meningitidis is a life-threatening human bacterial pathogen responsible for pneumonia, sepsis, and meningitis. Meningococcal strains with reduced susceptibility to penicillin G (Pen(I)) carry a mutated penicillin-binding protein (PBP2) resulting in a modified peptidoglycan structure. Despite their antibiotic resistance, Pen(I) strains have failed to expand clonally. We analyzed the biological consequences of PBP2 alteration among clinical meningococcal strains and found that peptidoglycan modifications of the Pen(I) strain resulted in diminished in vitro Nod1-dependent proinflammatory activity. In an influenza virus-meningococcal sequential mouse model mimicking human disease, wild-type meningococci induced a Nod1-dependent inflammatory response, colonizing the lungs and surviving in the blood. In contrast, isogenic Pen(I) strains were attenuated for such response and were out-competed by meningococci sensitive to penicillin G. Our results suggest that antibiotic resistance imposes a cost to the success of the pathogen and may potentially explain the lack of clonal expansion of Pen(I) strains.


Assuntos
Parede Celular/imunologia , Neisseria meningitidis/patogenicidade , Proteína Adaptadora de Sinalização NOD1/imunologia , Resistência às Penicilinas , Proteínas de Ligação às Penicilinas/genética , Animais , Parede Celular/metabolismo , Humanos , Camundongos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neisseria meningitidis/efeitos dos fármacos , Neisseria meningitidis/imunologia , Proteínas de Ligação às Penicilinas/metabolismo
19.
Nat Med ; 18(8): 1262-70, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22842476

RESUMO

Profibrotic cells that develop upon injury generate permanent scar tissue and impair organ recovery, though their origin and fate are unclear. Here we show that transient expression of ADAM12 (a disintegrin and metalloprotease 12) identifies a distinct proinflammatory subset of platelet-derived growth factor receptor-α-positive stromal cells that are activated upon acute injury in the muscle and dermis. By inducible genetic fate mapping, we demonstrate in vivo that injury-induced ADAM12(+) cells are specific progenitors of a major fraction of collagen-overproducing cells generated during scarring, which are progressively eliminated during healing. Genetic ablation of ADAM12(+) cells, or knockdown of ADAM12, is sufficient to limit generation of profibrotic cells and interstitial collagen accumulation. ADAM12(+) cells induced upon injury are developmentally distinct from muscle and skin lineage cells and are derived from fetal ADAM12(+) cells programmed during vascular wall development. Thus, our data identify injury-activated profibrotic progenitors residing in the perivascular space that can be targeted through ADAM12 to limit tissue scarring.


Assuntos
Proteínas ADAM/análise , Cicatriz/patologia , Derme/lesões , Músculo Esquelético/lesões , Miofibroblastos/patologia , Células Estromais/patologia , Proteínas ADAM/deficiência , Proteínas ADAM/genética , Proteína ADAM12 , Doença Aguda , Adipócitos/patologia , Animais , Vasos Sanguíneos/citologia , Linhagem da Célula , Proteínas Cardiotóxicas de Elapídeos/toxicidade , Colágeno/biossíntese , Cruzamentos Genéticos , Derme/metabolismo , Derme/patologia , Orelha Externa/lesões , Orelha Externa/metabolismo , Orelha Externa/patologia , Fibrose , Adjuvante de Freund/toxicidade , Técnicas de Silenciamento de Genes , Genes Reporter , Traumatismos da Perna/metabolismo , Traumatismos da Perna/patologia , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miofibroblastos/metabolismo , Parabiose , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/análise , Organismos Livres de Patógenos Específicos , Células Estromais/metabolismo , Cicatrização
20.
Autophagy ; 8(8): 1261-3, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22836517

RESUMO

It has been recognized that macroautophagy constitutes an important survival mechanism that allows both the maintenance of cellular homeostasis and the regulation of programmed cell death pathways (e.g., apoptosis). Although several pathogens have been described to induce autophagy, the prosurvival function of this process in infectious models remains poorly characterized. Our recent studies on chikungunya virus (CHIKV), the causative agent of major epidemics in India, Southeast Asia and southern Europe, reveal a novel mechanism by which autophagy limits the cytopathic effects of CHIKV by impinging upon virus-induced cell death pathways.


Assuntos
Autofagia , Vírus Chikungunya/fisiologia , Estresse do Retículo Endoplasmático , Estresse Oxidativo , Infecções por Alphavirus/patologia , Animais , Apoptose , Febre de Chikungunya , Humanos , Camundongos , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA