Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Protoc ; 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38906985

RESUMO

Tissues are dynamic and complex biological systems composed of specialized cell types that interact with each other for proper biological function. To comprehensively characterize and understand the cell circuitry underlying biological processes within tissues, it is crucial to preserve their spatial information. Here we report a simple mounting technique to maximize the area of the tissue to be analyzed, encompassing the whole length of the murine gastrointestinal (GI) tract, from mouth to rectum. Using this method, analysis of the whole murine GI tract can be performed in a single slide not only by means of histological staining, immunohistochemistry and in situ hybridization but also by multiplexed antibody staining and spatial transcriptomic approaches. We demonstrate the utility of our method in generating a comprehensive gene and protein expression profile of the whole GI tract by combining the versatile tissue-rolling technique with a cutting-edge transcriptomics method (Visium) and two cutting-edge proteomics methods (ChipCytometry and CODEX-PhenoCycler) in a systematic and easy-to-follow step-by-step procedure. The entire process, including tissue rolling, processing and sectioning, can be achieved within 2-3 d for all three methods. For Visium spatial transcriptomics, an additional 2 d are needed, whereas for spatial proteomics assays (ChipCytometry and CODEX-PhenoCycler), another 3-4 d might be considered. The whole process can be accomplished by researchers with skills in performing murine surgery, and standard histological and molecular biology methods.

2.
Nat Commun ; 15(1): 1752, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38409190

RESUMO

Stromal cells support epithelial cell and immune cell homeostasis and play an important role in inflammatory bowel disease (IBD) pathogenesis. Here, we quantify the stromal response to inflammation in pediatric IBD and reveal subset-specific inflammatory responses across colon segments and intestinal layers. Using data from a murine dynamic gut injury model and human ex vivo transcriptomic, protein and spatial analyses, we report that PDGFRA+CD142-/low fibroblasts and monocytes/macrophages co-localize in the intestine. In primary human fibroblast-monocyte co-cultures, intestinal PDGFRA+CD142-/low fibroblasts foster monocyte transition to CCR2+CD206+ macrophages through granulocyte-macrophage colony-stimulating factor (GM-CSF). Monocyte-derived CCR2+CD206+ cells from co-cultures have a phenotype similar to intestinal CCR2+CD206+ macrophages from newly diagnosed pediatric IBD patients, with high levels of PD-L1 and low levels of GM-CSF receptor. The study describes subset-specific changes in stromal responses to inflammation and suggests that the intestinal stroma guides intestinal macrophage differentiation.


Assuntos
Doenças Inflamatórias Intestinais , Monócitos , Humanos , Animais , Camundongos , Criança , Monócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Macrófagos/metabolismo , Inflamação/metabolismo , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/metabolismo , Diferenciação Celular
3.
EBioMedicine ; 94: 104691, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37480626

RESUMO

BACKGROUND: Radiotherapy is effective in the treatment of cancer but also causes damage to non-cancerous tissue. Pelvic radiotherapy may produce chronic and debilitating bowel symptoms, yet the underlying pathophysiology is still undefined. Most notably, although pelvic radiotherapy causes an acute intestinal inflammation there is no consensus on whether the late-phase pathophysiology contains an inflammatory component or not. To address this knowledge gap, we examined the potential presence of a chronic inflammation in mucosal biopsies from irradiated pelvic cancer survivors. METHODS: We biopsied 24 cancer survivors two to 20 years after pelvic radiotherapy, and four non-irradiated controls. Using tandem mass tag (TMT) mass spectrometry and mRNA sequencing (mRNA-seq), we charted proteomic and transcriptomic profiles of the mucosal tissue previously exposed to a high or a low/no dose of radiation. Changes in the immune cell populations were determined with flow cytometry. The integrity of the protective mucus layers were determined by permeability analysis and 16S rRNA bacterial detection. FINDINGS: 942 proteins were differentially expressed in mucosa previously exposed to a high radiation dose compared to a low radiation dose. The data suggested a chronic low-grade inflammation with neutrophil activity, which was confirmed by mRNA-seq and flow cytometry and further supported by findings of a weakened mucus barrier with bacterial infiltration. INTERPRETATION: Our results challenge the idea that pelvic radiotherapy causes an acute intestinal inflammation that either heals or turns fibrotic without progression to chronic inflammation. This provides a rationale for exploring novel strategies to mitigate chronic bowel symptoms in pelvic cancer survivors. FUNDING: This study was supported by the King Gustav V Jubilee Clinic Cancer Foundation (CB), The Adlerbertska Research Foundation (CB), The Swedish Cancer Society (GS), The Swedish State under the ALF agreement (GS and CB), Mary von Sydow's foundation (MA and VP).

4.
Immunity ; 55(12): 2336-2351.e12, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36462502

RESUMO

Therapeutic promotion of intestinal regeneration holds great promise, but defining the cellular mechanisms that influence tissue regeneration remains an unmet challenge. To gain insight into the process of mucosal healing, we longitudinally examined the immune cell composition during intestinal damage and regeneration. B cells were the dominant cell type in the healing colon, and single-cell RNA sequencing (scRNA-seq) revealed expansion of an IFN-induced B cell subset during experimental mucosal healing that predominantly located in damaged areas and associated with colitis severity. B cell depletion accelerated recovery upon injury, decreased epithelial ulceration, and enhanced gene expression programs associated with tissue remodeling. scRNA-seq from the epithelial and stromal compartments combined with spatial transcriptomics and multiplex immunostaining showed that B cells decreased interactions between stromal and epithelial cells during mucosal healing. Activated B cells disrupted the epithelial-stromal cross talk required for organoid survival. Thus, B cell expansion during injury impairs epithelial-stromal cell interactions required for mucosal healing, with implications for the treatment of IBD.


Assuntos
Colite , Mucosa Intestinal , Animais , Cicatrização , Células Epiteliais/metabolismo , Epitélio , Modelos Animais de Doenças
5.
Nat Rev Gastroenterol Hepatol ; 19(8): 493-507, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35440774

RESUMO

Almost all currently available treatments for inflammatory bowel disease (IBD) act by inhibiting inflammation, often blocking specific inflammatory molecules. However, given the infectious and neoplastic disease burden associated with chronic immunosuppressive therapy, the goal of attaining mucosal healing without immunosuppression is attractive. The absence of treatments that directly promote mucosal healing and regeneration in IBD could be linked to the lack of understanding of the underlying pathways. The range of potential strategies to achieve mucosal healing is diverse. However, the targeting of regenerative mechanisms has not yet been achieved for IBD. Stem cells provide hope as a regenerative treatment and are used in limited clinical situations. Growth factors are available for the treatment of short bowel syndrome but have not yet been applied in IBD. The therapeutic application of organoid culture and stem cell therapy to generate new intestinal tissue could provide a novel mechanism to restore barrier function in IBD. Furthermore, blocking key effectors of barrier dysfunction (such as MLCK or damage-associated molecular pattern molecules) has shown promise in experimental IBD. Here, we review the diversity of molecular targets available to directly promote mucosal healing, experimental models to identify new potential pathways and some of the anticipated potential therapies for IBD.


Assuntos
Doenças Inflamatórias Intestinais , Mucosa Intestinal , Humanos , Terapia de Imunossupressão , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal/metabolismo , Intestinos , Cicatrização
6.
Nat Commun ; 13(1): 828, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149721

RESUMO

The intestinal barrier is composed of a complex cell network defining highly compartmentalized and specialized structures. Here, we use spatial transcriptomics to define how the transcriptomic landscape is spatially organized in the steady state and healing murine colon. At steady state conditions, we demonstrate a previously unappreciated molecular regionalization of the colon, which dramatically changes during mucosal healing. Here, we identified spatially-organized transcriptional programs defining compartmentalized mucosal healing, and regions with dominant wired pathways. Furthermore, we showed that decreased p53 activation defined areas with increased presence of proliferating epithelial stem cells. Finally, we mapped transcriptomics modules associated with human diseases demonstrating the translational potential of our dataset. Overall, we provide a publicly available resource defining principles of transcriptomic regionalization of the colon during mucosal healing and a framework to develop and progress further hypotheses.


Assuntos
Intestinos/metabolismo , Transcriptoma , Cicatrização , Animais , Colo/metabolismo , Colo/patologia , Modelos Animais de Doenças , Células Epiteliais , Feminino , Mucosa Intestinal/metabolismo , Intestinos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes Neurológicos , Transdução de Sinais
7.
Cancer Res Commun ; 2(9): 1075-1088, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36922937

RESUMO

The tumor suppressor protein p53 is mutated in close to 50% of human tumors and is dysregulated in many others, for instance by silencing or loss of p14ARF. Under steady-state conditions, the two E3 ligases MDM2/MDM4 interact with and inhibit the transcriptional activity of p53. Inhibition of p53-MDM2/4 interaction to reactivate p53 in tumors with wild-type (WT) p53 has therefore been considered a therapeutic strategy. Moreover, studies indicate that p53 reactivation may synergize with radiation and increase tumor immunogenicity. In vivo studies of most MDM2 inhibitors have utilized immunodeficient xenograft mouse models, preventing detailed studies of action of these molecules on the immune response. The mouse melanoma cell line B16-F10 carries functional, WT p53 but does not express the MDM2 regulator p19ARF. In this study, we tested a p53-MDM2 protein-protein interaction inhibitor, the small molecule Navtemadlin, which is currently being tested in phase II clinical trials. Using mass spectrometry-based proteomics and imaging flow cytometry, we identified specific protein expression patterns following Navtemadlin treatment of B16-F10 melanoma cells compared with their p53 CRISPR-inactivated control cells. In vitro, Navtemadlin induced a significant, p53-dependent, growth arrest but little apoptosis in B16-F10 cells. When combined with radiotherapy, Navtemadlin showed synergistic effects and increased apoptosis. In vivo, Navtemadlin treatment significantly reduced the growth of B16-F10 melanoma cells implanted in C57Bl/6 mice. Our data highlight the utility of a syngeneic B16-F10 p53+/+ mouse melanoma model for assessing existing and novel p53-MDM2/MDM4 inhibitors and in identifying new combination therapies that can efficiently eliminate tumors in vivo. Significance: The MDM2 inhibitor Navtemadlin arrests mouse tumor growth and potentiates radiotherapy. Our results support a threshold model for apoptosis induction that requires a high, prolonged p53 signaling for cancer cells to become apoptotic.


Assuntos
Antineoplásicos , Melanoma Experimental , Humanos , Animais , Camundongos , Proteínas Proto-Oncogênicas c-mdm2 , Proteína Supressora de Tumor p53/genética , Ubiquitina-Proteína Ligases/metabolismo , Melanoma Experimental/tratamento farmacológico , Modelos Animais de Doenças , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Ciclo Celular/metabolismo
8.
Immunity ; 54(2): 259-275.e7, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33382972

RESUMO

The study of human macrophages and their ontogeny is an important unresolved issue. Here, we use a humanized mouse model expressing human cytokines to dissect the development of lung macrophages from human hematopoiesis in vivo. Human CD34+ hematopoietic stem and progenitor cells (HSPCs) generated three macrophage populations, occupying separate anatomical niches in the lung. Intravascular cell labeling, cell transplantation, and fate-mapping studies established that classical CD14+ blood monocytes derived from HSPCs migrated into lung tissue and gave rise to human interstitial and alveolar macrophages. In contrast, non-classical CD16+ blood monocytes preferentially generated macrophages resident in the lung vasculature (pulmonary intravascular macrophages). Finally, single-cell RNA sequencing defined intermediate differentiation stages in human lung macrophage development from blood monocytes. This study identifies distinct developmental pathways from circulating monocytes to lung macrophages and reveals how cellular origin contributes to human macrophage identity, diversity, and localization in vivo.


Assuntos
Células-Tronco Hematopoéticas/imunologia , Pulmão/imunologia , Macrófagos Alveolares/imunologia , Monócitos/imunologia , Antígenos CD34/metabolismo , Biodiversidade , Diferenciação Celular , Movimento Celular , Células Cultivadas , Sangue Fetal/citologia , Humanos , Receptores de Lipopolissacarídeos/metabolismo , Pulmão/irrigação sanguínea , Receptores de IgG/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Nicho de Células-Tronco
9.
Cell Rep ; 32(5): 107979, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32755573

RESUMO

Single-nucleotide polymorphisms in the gene encoding G protein-coupled receptor 35 (GPR35) are associated with increased risk of inflammatory bowel disease. However, the mechanisms by which GPR35 modulates intestinal immune homeostasis remain undefined. Here, integrating zebrafish and mouse experimental models, we demonstrate that intestinal Gpr35 expression is microbiota dependent and enhanced upon inflammation. Moreover, murine GPR35+ colonic macrophages are characterized by enhanced production of pro-inflammatory cytokines. We identify lysophosphatidic acid (LPA) as a potential endogenous ligand produced during intestinal inflammation, acting through GPR35 to induce tumor necrosis factor (Tnf) expression in macrophages. Mice lacking Gpr35 in CX3CR1+ macrophages aggravate colitis when exposed to dextran sodium sulfate, which is associated with decreased transcript levels of the corticosterone-generating gene Cyp11b1 and macrophage-derived Tnf. Administration of TNF in these mice restores Cyp11b1 expression and intestinal corticosterone production and ameliorates DSS-induced colitis. Our findings indicate that LPA signals through GPR35 in CX3CR1+ macrophages to maintain TNF-mediated intestinal homeostasis.


Assuntos
Receptor 1 de Quimiocina CX3C/metabolismo , Homeostase , Intestinos/fisiologia , Lisofosfolipídeos/metabolismo , Macrófagos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Proteínas de Peixe-Zebra/metabolismo , Animais , Colite/induzido quimicamente , Colite/patologia , Sulfato de Dextrana , Microbioma Gastrointestinal , Deleção de Genes , Humanos , Inflamação/patologia , Doenças Inflamatórias Intestinais/patologia , Camundongos Endogâmicos C57BL , Diester Fosfórico Hidrolases/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Peixe-Zebra
10.
J Crohns Colitis ; 14(12): 1748-1758, 2020 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-32556102

RESUMO

BACKGROUND AND AIMS: Ulcerative colitis [UC] is associated with excessive neutrophil infiltration and collateral tissue damage, but the link is not yet completely understood. Since c-MET receptor tyrosine kinase [MET] is required for neutrophil chemoattraction and cytotoxicity in response to its ligand hepatocyte growth factor [HGF], we aimed to identify the function of HGF-MET signalling in neutrophils in UC patients and in mice during intestinal inflammation. METHODS: Serum and colonic biopsies from healthy controls and UC patients with active [Mayo endoscopic subscore 2-3] and inactive [Mayo endoscopic subscore 0-1] disease were collected to assess the level of serum and colonic HGF. Disease progression and immune cell infiltration were assessed during dextran sodium sulphate [DSS] colitis in wild-type and MRP8-Cre MET-LoxP mice. RESULTS: Increased mucosal HGF expression was detected in patients with active UC, and in mice during the inflammatory phase of DSS colitis. Similarly, serum HGF was significantly increased in active UC patients and positively correlated with C-reactive protein and blood neutrophil counts. Flow cytometric analysis also demonstrated an upregulation of colonic MET+ neutrophils during DSS colitis. Genetic ablation of MET in neutrophils reduced the severity of DSS-induced colitis. Concomitantly, there was a decreased number of TH17 cells, which could be due to a decreased production of IL-1ß by MET-deficient neutrophils. CONCLUSIONS: These data highlight the central role of neutrophilic HGF-MET signalling in exacerbating damage during intestinal inflammation. Hence, selective blockade of this pathway in neutrophils could be considered as a novel therapeutic approach in UC.


Assuntos
Colite Ulcerativa/genética , Fator de Crescimento de Hepatócito/farmacologia , Proteínas Proto-Oncogênicas c-met/farmacologia , Transdução de Sinais/fisiologia , Exacerbação dos Sintomas , Animais , Bélgica , Colite Ulcerativa/fisiopatologia , Colo/metabolismo , Colo/patologia , Colo/fisiopatologia , Modelos Animais de Doenças , Citometria de Fluxo/métodos , Citometria de Fluxo/estatística & dados numéricos , Fator de Crescimento de Hepatócito/genética , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-met/genética , Transdução de Sinais/imunologia
11.
Nat Commun ; 11(1): 2049, 2020 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-32341343

RESUMO

ILCs and T helper cells have been shown to exert bi-directional regulation in mice. However, how crosstalk between ILCs and CD4+ T cells influences immune function in humans is unknown. Here we show that human intestinal ILCs co-localize with T cells in healthy and colorectal cancer tissue and display elevated HLA-DR expression in tumor and tumor-adjacent areas. Although mostly lacking co-stimulatory molecules ex vivo, intestinal and peripheral blood (PB) ILCs acquire antigen-presenting characteristics triggered by inflammasome-associated cytokines IL-1ß and IL-18. IL-1ß drives the expression of HLA-DR and co-stimulatory molecules on PB ILCs in an NF-κB-dependent manner, priming them as efficient inducers of cytomegalovirus-specific memory CD4+ T-cell responses. This effect is strongly inhibited by the anti-inflammatory cytokine TGF-ß. Our results suggest that circulating and tissue-resident ILCs have the intrinsic capacity to respond to the immediate cytokine milieu and regulate local CD4+ T-cell responses, with potential implications for anti-tumor immunity and inflammation.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Citocinas/imunologia , Imunidade Inata , Linfócitos/imunologia , Animais , Apresentação de Antígeno , Linfócitos T CD4-Positivos/imunologia , Neoplasias do Colo/imunologia , Humanos , Inflamassomos/imunologia , Interleucina-18/imunologia , Interleucina-1beta/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Leucócitos Mononucleares/imunologia , Camundongos
12.
J Autoimmun ; 98: 86-94, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30616979

RESUMO

CTLA-4 is required for CD4+Foxp3+ regulatory T (Treg) cell function, but its mode of action remains incompletely defined. Herein we generated Ctla-4ex2fl/flFoxp3-Cre mice with Treg cells exclusively expressing a naturally occurring, ligand-independent isoform of CTLA-4 (liCTLA-4) that cannot interact with the costimulatory molecules CD80 and CD86. The mice did not exhibit any signs of effector T cell activation early in life, however, at 6 months of age they exhibited excessive T cell activation and inflammation in lungs. In contrast, mice with Treg cells completely lacking CTLA-4 developed lymphoproliferative disease characterized by multi-organ inflammation early in life. In vitro, Treg cells exclusively expressing liCTLA-4 inhibited CD80 and CD86 expression on dendritic cells (DC). Conversely, Treg cells required the extra-cellular part of CTLA-4 to up-regulate expression of the co-inhibitory molecule PD-L2 on DCs. Transcriptomic analysis of suppressed DCs revealed that Treg cells induced a specific immunosuppressive program in DCs.


Assuntos
Antígeno CTLA-4/metabolismo , Células Dendríticas/imunologia , Transtornos Linfoproliferativos/imunologia , Pneumonia/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos CD4/metabolismo , Antígeno CTLA-4/genética , Diferenciação Celular , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Ativação Linfocitária , Transtornos Linfoproliferativos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pneumonia/genética , Proteína 2 Ligante de Morte Celular Programada 1/genética , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Isoformas de Proteínas/genética
13.
Dis Model Mech ; 11(11)2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30266803

RESUMO

Xenografts of the hematopoietic system are extremely useful as disease models and for translational research. Zebrafish xenografts have been widely used to monitor blood cancer cell dissemination and homing due to the optical clarity of embryos and larvae, which allow unrestricted in vivo visualization of migratory events. Here, we have developed a xenotransplantation technique that transiently generates hundreds of hematopoietic tissue chimeric embryos by transplanting murine bone marrow cells into zebrafish blastulae. In contrast to previous methods, this procedure allows mammalian cell integration into the fish developmental hematopoietic program, which results in chimeric animals containing distinct phenotypes of murine blood cells in both circulation and the hematopoietic niche. Murine cells in chimeric animals express antigens related to (i) hematopoietic stem and progenitor cells, (ii) active cell proliferation and (iii) myeloid cell lineages. We verified the utility of this method by monitoring zebrafish chimeras during development using in vivo non-invasive imaging to show novel murine cell behaviors, such as homing to primitive and definitive hematopoietic tissues, dynamic hematopoietic cell and hematopoietic niche interactions, and response to bacterial infection. Overall, transplantation into the zebrafish blastula provides a useful method that simplifies the generation of numerous chimeric animals and expands the range of murine cell behaviors that can be studied in zebrafish chimeras. In addition, integration of murine cells into the host hematopoietic system during development suggests highly conserved molecular mechanisms of hematopoiesis between zebrafish and mammals.This article has an associated First Person interview with the first author of the paper.


Assuntos
Quimera/embriologia , Embrião de Mamíferos/fisiologia , Embrião não Mamífero/fisiologia , Hematopoese , Interações Hospedeiro-Patógeno , Peixe-Zebra/embriologia , Animais , Infecções Bacterianas/patologia , Blástula/transplante , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Fusão Celular , Linhagem da Célula , Movimento Celular , Rastreamento de Células , Corantes/metabolismo , Feminino , Larva/citologia , Masculino , Camundongos Endogâmicos C57BL , Células Mieloides/citologia , Transplante Heterólogo , Peixe-Zebra/microbiologia
14.
PLoS One ; 13(9): e0204181, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30235302

RESUMO

The gut homing receptor integrin α4ß7 is essential for the migration of pro-inflammatory T cells into the gut mucosa. Since intestinal neoplasia has been associated with chronic inflammation, we investigated whether interfering with gut-homing affects intestinal tumorigenesis. Using chemically induced and spontaneous intestinal tumor models we showed that lack of ß7 integrin significantly impairs tumor growth without affecting tumor frequencies, with a mild translatable effect on overall survival. This correlates with human data showing lower MAdCAM-1 expression and disease-free survival in colorectal cancer patients. Thus, paradoxically in contrast to extra-intestinal tumors, blocking migration of immune cells into the gut might have a positive therapeutic effect on intestinal neoplasia.


Assuntos
Cadeias beta de Integrinas/metabolismo , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Animais , Carcinogênese/metabolismo , Carcinogênese/patologia , Proliferação de Células , Inflamação/patologia , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sobrevida , Linfócitos T/imunologia , Carga Tumoral
15.
Sci Rep ; 8(1): 154, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29317685

RESUMO

A common helper-like innate lymphoid precursor (CHILP) restricted to the innate lymphoid cells (ILC) lineage has been recently characterized. While specific requirements of transcription factors for CHILPs development has been partially described, their ability to sense cytokines and react to peripheral inflammation remains unaddressed. Here, we found that systemic increase in Flt3L levels correlated with the expansion of Lineage (Lin)negα4ß7+ precursors in the adult murine bone marrow. Expanded Linnegα4ß7+ precursors were bona fide CHILPs as seen by their ability to differentiate into all helper ILCs subsets but cNK in vivo. Interestingly, Flt3L-expanded CHILPs transferred into lymphopenic mice preferentially reconstituted the small intestine. While we did not observe changes in serum Flt3L during DSS-induced colitis in mice or plasma from inflammatory bowel disease (IBD) patients, elevated Flt3L levels were detected in acute malaria patients. Interestingly, while CHILP numbers were stable during the course of DSS-induced colitis, they expanded following increased serum Flt3L levels in malaria-infected mice, hence suggesting a role of the Flt3L-ILC axis in malaria. Collectively, our results indicate that Flt3L expands CHILPs in the bone marrow, which might be associated with specific inflammatory conditions.


Assuntos
Imunidade Inata/genética , Subpopulações de Linfócitos/metabolismo , Células Progenitoras Linfoides/metabolismo , Proteínas de Membrana/genética , Animais , Biomarcadores , Células da Medula Óssea/metabolismo , Modelos Animais de Doenças , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Integrinas/metabolismo , Subpopulações de Linfócitos/imunologia , Células Progenitoras Linfoides/imunologia , Melanoma Experimental , Proteínas de Membrana/sangue , Proteínas de Membrana/metabolismo , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo
16.
J Allergy Clin Immunol ; 141(1): 279-292, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28433688

RESUMO

BACKGROUND: Vitamin D deficiency is a risk factor for inflammatory bowel disease (IBD). The IL-23-driven tissue-resident group 3 innate lymphoid cells (ILC3s) play essential roles in intestinal immunity, and targeting IL-23/12 is a promising approach in IBD therapy. OBJECTIVE: We set out to define the role of 1α,25-dihydroxy vitamin D3 (1,25D) in regulating functional responses of human mucosal ILC3s to IL-23 plus IL-1ß stimulation. METHODS: Transcriptomes of sorted tonsillar ILC3s were assessed by using microarray analysis. ILC3 cytokine production, proliferation, and differentiation were determined by means of flow cytometry, ELISA, and multiplex immunoassay. Intestinal cell suspensions and ILC3s sorted from gut biopsy specimens of patients with IBD were also analyzed along with plasma 25-hydroxy vitamin D3 (25D) detection. RESULTS: ILC3s stimulated with IL-23 plus IL-1ß upregulated the vitamin D receptor and responded to 1,25D with downregulation of the IL-23 receptor pathway. Consequently, 1,25D suppressed IL-22, IL-17F, and GM-CSF production from tonsillar and gut ILC3s. In parallel, 1,25D upregulated genes linked to the IL-1ß signaling pathway, as well as the IL-1ß-inducible cytokines IL-6, IL-8, and macrophage inflammatory protein 1α/ß. The 1,25D-triggered skewing in ILC3 function was not accompanied or caused by changes in viability, proliferation, or phenotype. Finally, we confirmed low 25D plasma levels in patients with IBD with active inflammation. CONCLUSION: In light of the beneficial targeting of IL-23/12 in patients with IBD, 1,25D appears as an interesting therapeutic agent that inhibits the IL-23 receptor pathway, providing a novel mechanism for how ILC3s could be manipulated to regulate intestinal inflammation.


Assuntos
Linfócitos/imunologia , Linfócitos/metabolismo , Mucosa/citologia , Mucosa/imunologia , Receptores de Interleucina/metabolismo , Transdução de Sinais/imunologia , Vitamina D/farmacologia , Biomarcadores , Proliferação de Células , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunofenotipagem , Ativação Linfocitária/imunologia , Mucosa/efeitos dos fármacos , Mucosa/metabolismo , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo
17.
Mol Cell Proteomics ; 16(3): 502-511, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28111361

RESUMO

Extracellular vesicles (EVs) are membrane-coated objects such as exosomes and microvesicles, released by many cell-types. Their presence in body fluids and the variable surface composition and content render them attractive potential biomarkers. The ability to determine their cellular origin could greatly move the field forward. We used multiplex proximity extension assays (PEA) to identify with high specificity and sensitivity the protein profiles of exosomes of different origins, including seven cell lines and two different body fluids. By comparing cells and exosomes, we successfully identified the cells originating the exosomes. Furthermore, by principal component analysis of protein patterns human milk EVs and prostasomes released from prostate acinar cells clustered with cell lines from breast and prostate tissues, respectively. Milk exosomes uniquely expressed CXCL5, MIA, and KLK6, whereas prostasomes carried NKX31, GSTP1, and SRC, highlighting that EVs originating from different origins express distinct proteins. In conclusion, PEA provides a powerful protein screening tool in exosome research, for purposes of identifying the cell source of exosomes, or new biomarkers in diseases such as cancer and inflammation.


Assuntos
Biomarcadores/metabolismo , Líquidos Corporais/metabolismo , Micropartículas Derivadas de Células/metabolismo , Exossomos/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Linhagem Celular , Feminino , Humanos , Células K562 , Células MCF-7 , Masculino , Leite Humano/metabolismo , Especificidade de Órgãos , Análise de Componente Principal , Próstata/metabolismo
18.
Immunity ; 44(6): 1392-405, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27287411

RESUMO

Although numerous polymorphisms have been associated with inflammatory bowel disease (IBD), identifying the function of these genetic factors has proved challenging. Here we identified a role for nine genes in IBD susceptibility loci in antibacterial autophagy and characterized a role for one of these genes, GPR65, in maintaining lysosome function. Mice lacking Gpr65, a proton-sensing G protein-coupled receptor, showed increased susceptibly to bacteria-induced colitis. Epithelial cells and macrophages lacking GPR65 exhibited impaired clearance of intracellular bacteria and accumulation of aberrant lysosomes. Similarly, IBD patient cells and epithelial cells expressing an IBD-associated missense variant, GPR65 I231L, displayed aberrant lysosomal pH resulting in lysosomal dysfunction, impaired bacterial restriction, and altered lipid droplet formation. The GPR65 I231L polymorphism was sufficient to confer decreased GPR65 signaling. Collectively, these data establish a role for GPR65 in IBD susceptibility and identify lysosomal dysfunction as a potentially causative element in IBD pathogenesis with effects on cellular homeostasis and defense.


Assuntos
Colite/imunologia , Células Epiteliais/imunologia , Doenças Inflamatórias Intestinais/genética , Lisossomos/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Infecções por Salmonella/imunologia , Salmonella enterica/imunologia , Salmonella typhimurium/imunologia , Animais , Predisposição Genética para Doença , Células HeLa , Humanos , Doenças Inflamatórias Intestinais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagossomos/fisiologia , Polimorfismo Genético , Receptores Acoplados a Proteínas G/genética , Risco
19.
Nat Commun ; 6: 7838, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26194095

RESUMO

The phagocyte oxidative burst, mediated by Nox2 NADPH oxidase-derived reactive oxygen species, confers host defense against a broad spectrum of bacterial and fungal pathogens. Loss-of-function mutations that impair function of the Nox2 complex result in a life-threatening immunodeficiency, and genetic variants of Nox2 subunits have been implicated in pathogenesis of inflammatory bowel disease (IBD). Thus, alterations in the oxidative burst can profoundly impact host defense, yet little is known about regulatory mechanisms that fine-tune this response. Here we report the discovery of regulatory nodes controlling oxidative burst by functional screening of genes within loci linked to human inflammatory disease. Implementing a multi-omics approach, we define transcriptional, metabolic and ubiquitin-cycling nodes controlled by Rbpj, Pfkl and Rnf145, respectively. Furthermore, we implicate Rnf145 in proteostasis of the Nox2 complex by endoplasmic reticulum-associated degradation. Consequently, ablation of Rnf145 in murine macrophages enhances bacterial clearance, and rescues the oxidative burst defects associated with Ncf4 haploinsufficiency.


Assuntos
Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Fagócitos/metabolismo , Explosão Respiratória , Animais , Sequência de Bases , Linhagem Celular , Genômica , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Camundongos , Dados de Sequência Molecular , NADPH Oxidase 2 , Fosfofrutoquinase-1/metabolismo , Staphylococcus aureus
20.
Cell Rep ; 11(12): 1905-18, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26095365

RESUMO

The polymorphism ATG16L1 T300A, associated with increased risk of Crohn's disease, impairs pathogen defense mechanisms including selective autophagy, but specific pathway interactions altered by the risk allele remain unknown. Here, we use perturbational profiling of human peripheral blood cells to reveal that CLEC12A is regulated in an ATG16L1-T300A-dependent manner. Antibacterial autophagy is impaired in CLEC12A-deficient cells, and this effect is exacerbated in the presence of the ATG16L1(∗)300A risk allele. Clec12a(-/-) mice are more susceptible to Salmonella infection, supporting a role for CLEC12A in antibacterial defense pathways in vivo. CLEC12A is recruited to sites of bacterial entry, bacteria-autophagosome complexes, and sites of sterile membrane damage. Integrated genomics identified a functional interaction between CLEC12A and an E3-ubiquitin ligase complex that functions in antibacterial autophagy. These data identify CLEC12A as early adaptor molecule for antibacterial autophagy and highlight perturbational profiling as a method to elucidate defense pathways in complex genetic disease.


Assuntos
Proteínas de Transporte/genética , Doença de Crohn/genética , Lectinas Tipo C/genética , Receptores Mitogênicos/genética , Infecções por Salmonella/genética , Alelos , Animais , Autofagia/genética , Proteínas Relacionadas à Autofagia , Doença de Crohn/microbiologia , Doença de Crohn/patologia , Predisposição Genética para Doença , Genômica , Humanos , Lectinas Tipo C/biossíntese , Camundongos , Receptores Mitogênicos/biossíntese , Fatores de Risco , Salmonella/patogenicidade , Infecções por Salmonella/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA