Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cell Death Differ ; 31(3): 265-279, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38383888

RESUMO

PirB is an inhibitory cell surface receptor particularly prominent on myeloid cells. PirB curtails the phenotypes of activated macrophages during inflammation or tumorigenesis, but its functions in macrophage homeostasis are obscure. To elucidate PirB-related functions in macrophages at steady-state, we generated and compared single-cell RNA-sequencing (scRNAseq) datasets obtained from myeloid cell subsets of wild type (WT) and PirB-deficient knockout (PirB KO) mice. To facilitate this analysis, we developed a novel approach to clustering parameter optimization called "Cluster Similarity Scoring and Distinction Index" (CaSSiDI). We demonstrate that CaSSiDI is an adaptable computational framework that facilitates tandem analysis of two scRNAseq datasets by optimizing clustering parameters. We further show that CaSSiDI offers more advantages than a standard Seurat analysis because it allows direct comparison of two or more independently clustered datasets, thereby alleviating the need for batch-correction while identifying the most similar and different clusters. Using CaSSiDI, we found that PirB is a novel regulator of Cebpb expression that controls the generation of Ly6Clo patrolling monocytes and the expansion properties of peritoneal macrophages. PirB's effect on Cebpb is tissue-specific since it was not observed in splenic red pulp macrophages (RPMs). However, CaSSiDI revealed a segregation of the WT RPM population into a CD68loIrf8+ "neuronal-primed" subset and an CD68hiFtl1+ "iron-loaded" subset. Our results establish the utility of CaSSiDI for single-cell assay analyses and the determination of optimal clustering parameters. Our application of CaSSiDI in this study has revealed previously unknown roles for PirB in myeloid cell populations. In particular, we have discovered homeostatic functions for PirB that are related to Cebpb expression in distinct macrophage subsets.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT , Macrófagos , Receptores Imunológicos , Análise de Célula Única , Animais , Camundongos , Macrófagos/metabolismo , Monócitos/metabolismo , Células Mieloides/metabolismo , Receptores de Superfície Celular , Receptores Imunológicos/metabolismo , Análise de Célula Única/métodos , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo
2.
Nat Cancer ; 4(10): 1437-1454, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37640929

RESUMO

Cholinergic nerves are involved in tumor progression and dissemination. In contrast to other visceral tissues, cholinergic innervation in the hepatic parenchyma is poorly detected. It remains unclear whether there is any form of cholinergic regulation of liver cancer. Here, we show that cholinergic T cells curtail the development of liver cancer by supporting antitumor immune responses. In a mouse multihit model of hepatocellular carcinoma (HCC), we observed activation of the adaptive immune response and induction of two populations of CD4+ T cells expressing choline acetyltransferase (ChAT), including regulatory T cells and dysfunctional PD-1+ T cells. Tumor antigens drove the clonal expansion of these cholinergic T cells in HCC. Genetic ablation of Chat in T cells led to an increased prevalence of preneoplastic cells and exacerbated liver cancer due to compromised antitumor immunity. Mechanistically, the cholinergic activity intrinsic in T cells constrained Ca2+-NFAT signaling induced by T cell antigen receptor engagement. Without this cholinergic modulation, hyperactivated CD25+ T regulatory cells and dysregulated PD-1+ T cells impaired HCC immunosurveillance. Our results unveil a previously unappreciated role for cholinergic T cells in liver cancer immunobiology.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Camundongos , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Receptor de Morte Celular Programada 1/genética , Monitorização Imunológica , Linfócitos T Reguladores/patologia
3.
Cell Death Differ ; 30(2): 407-416, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36528755

RESUMO

Experimental autoimmune encephalomyelitis (EAE) is a mouse model of multiple sclerosis (MS) in which Th17 cells have a crucial but unclear function. Here we show that choline acetyltransferase (ChAT), which synthesizes acetylcholine (ACh), is a critical driver of pathogenicity in EAE. Mice with ChAT-deficient Th17 cells resist disease progression and show reduced brain-infiltrating immune cells. ChAT expression in Th17 cells is linked to strong TCR signaling, expression of the transcription factor Bhlhe40, and increased Il2, Il17, Il22, and Il23r mRNA levels. ChAT expression in Th17 cells is independent of IL21r signaling but dampened by TGFß, implicating ChAT in controlling the dichotomous nature of Th17 cells. Our study establishes a cholinergic program in which ACh signaling primes chronic activation of Th17 cells, and thereby constitutes a pathogenic determinant of EAE. Our work may point to novel targets for therapeutic immunomodulation in MS.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Camundongos , Animais , Células Th17 , Virulência , Colinérgicos , Esclerose Múltipla/genética , Acetilcolina/metabolismo , Camundongos Endogâmicos C57BL , Diferenciação Celular
4.
Sci Immunol ; 7(67): eabf7777, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-34995099

RESUMO

Resident macrophages orchestrate homeostatic, inflammatory, and reparative activities. It is appreciated that different tissues instruct specialized macrophage functions. However, individual tissues contain heterogeneous subpopulations, and how these subpopulations are related is unclear. We asked whether common transcriptional and functional elements could reveal an underlying framework across tissues. Using single-cell RNA sequencing and random forest modeling, we observed that four genes could predict three macrophage subsets that were present in murine heart, liver, lung, kidney, and brain. Parabiotic and genetic fate mapping studies revealed that these core markers predicted three unique life cycles across 17 tissues. TLF+ (expressing TIMD4 and/or LYVE1 and/or FOLR2) macrophages were maintained through self-renewal with minimal monocyte input; CCR2+ (TIMD4−LYVE1−FOLR2−) macrophages were almost entirely replaced by monocytes, and MHC-IIhi macrophages (TIMD4−LYVE1−FOLR2−CCR2−), while receiving modest monocyte contribution, were not continually replaced. Rather, monocyte-derived macrophages contributed to the resident macrophage population until they reached a defined upper limit after which they did not outcompete pre-existing resident macrophages. Developmentally, TLF+ macrophages were first to emerge in the yolk sac and early fetal organs. Fate mapping studies in the mouse and human single-cell RNA sequencing indicated that TLF+ macrophages originated from both yolk sac and fetal monocyte precursors. Furthermore, TLF+ macrophages were the most transcriptionally conserved subset across mouse tissues and between mice and humans, despite organ- and species-specific transcriptional differences. Here, we define the existence of three murine macrophage subpopulations based on common life cycle properties and core gene signatures and provide a common starting point to understand tissue macrophage heterogeneity.


Assuntos
Receptor 2 de Folato/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Macrófagos/imunologia , Proteínas de Membrana/imunologia , Receptores CCR2/imunologia , Proteínas de Transporte Vesicular/imunologia , Animais , Estágios do Ciclo de Vida/imunologia , Ativação de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores CCR2/deficiência
5.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34210797

RESUMO

While modulatory effects of gut microbes on neurological phenotypes have been reported, the mechanisms remain largely unknown. Here, we demonstrate that indole, a tryptophan metabolite produced by tryptophanase-expressing gut microbes, elicits neurogenic effects in the adult mouse hippocampus. Neurogenesis is reduced in germ-free (GF) mice and in GF mice monocolonized with a single-gene tnaA knockout (KO) mutant Escherichia coli unable to produce indole. External administration of systemic indole increases adult neurogenesis in the dentate gyrus in these mouse models and in specific pathogen-free (SPF) control mice. Indole-treated mice display elevated synaptic markers postsynaptic density protein 95 and synaptophysin, suggesting synaptic maturation effects in vivo. By contrast, neurogenesis is not induced by indole in aryl hydrocarbon receptor KO (AhR-/-) mice or in ex vivo neurospheres derived from them. Neural progenitor cells exposed to indole exit the cell cycle, terminally differentiate, and mature into neurons that display longer and more branched neurites. These effects are not observed with kynurenine, another AhR ligand. The indole-AhR-mediated signaling pathway elevated the expression of ß-catenin, Neurog2, and VEGF-α genes, thus identifying a molecular pathway connecting gut microbiota composition and their metabolic function to neurogenesis in the adult hippocampus. Our data have implications for the understanding of mechanisms of brain aging and for potential next-generation therapeutic opportunities.


Assuntos
Envelhecimento/metabolismo , Microbioma Gastrointestinal , Neurogênese , Receptores de Hidrocarboneto Arílico/metabolismo , Triptofano/metabolismo , Animais , Indóis/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Células-Tronco Neurais/metabolismo
7.
Nat Commun ; 10(1): 2678, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31213601

RESUMO

Myeloid cells contribute to tumor progression, but how the constellation of receptors they express regulates their functions within the tumor microenvironment (TME) is unclear. We demonstrate that Fcmr (Toso), the putative receptor for soluble IgM, modulates myeloid cell responses to cancer. In a syngeneic melanoma model, Fcmr ablation in myeloid cells suppressed tumor growth and extended mouse survival. Fcmr deficiency increased myeloid cell population density in this malignancy and enhanced anti-tumor immunity. Single-cell RNA sequencing of Fcmr-deficient tumor-associated mononuclear phagocytes revealed a unique subset with enhanced antigen processing/presenting properties. Conversely, Fcmr activity negatively regulated the activation and migratory capacity of myeloid cells in vivo, and T cell activation by bone marrow-derived dendritic cells in vitro. Therapeutic targeting of Fcmr during oncogenesis decreased tumor growth when used as a single agent or in combination with anti-PD-1. Thus, Fcmr regulates myeloid cell activation within the TME and may be a potential therapeutic target.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proteínas de Transporte/metabolismo , Melanoma Experimental/imunologia , Proteínas de Membrana/metabolismo , Monócitos/imunologia , Neoplasias Cutâneas/imunologia , Animais , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinogênese/efeitos dos fármacos , Carcinogênese/imunologia , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Linhagem Celular Tumoral/transplante , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Feminino , Ativação Linfocitária/imunologia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/mortalidade , Melanoma Experimental/patologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/mortalidade , Neoplasias Cutâneas/patologia , Análise de Sobrevida , Linfócitos T/imunologia , Microambiente Tumoral/imunologia
8.
Curr Opin Immunol ; 48: 61-67, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28865357

RESUMO

The recent successes of immune check point targeting therapies in treating cancer patients has driven a resurgence of interest in targeting these pathways in chronically infected patients. While still in early stages, basic and clinical data suggest that blockade of CTLA-4 and PD-1 can be beneficial in the treatment of chronic HIV, HBV, and HCV infection, as well as other chronic maladies. Furthermore, novel inhibitory receptors such as Tim-3, LAG-3, and TIGIT are the potential next wave of check points that can be manipulated for the treatment of chronic infection. Blockade of these pathways influences more than simply T cell responses, and may provide new therapeutic options for chronically infected patients.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígeno CTLA-4/imunologia , Receptores Coestimuladores e Inibidores de Linfócitos T/imunologia , HIV-1/imunologia , Hepacivirus/imunologia , Vírus da Hepatite B/imunologia , Imunoterapia/métodos , Receptor de Morte Celular Programada 1/imunologia , Viroses/terapia , Animais , Antígenos CD/metabolismo , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Humanos , Terapia de Alvo Molecular , Receptores Imunológicos/metabolismo , Viroses/imunologia , Proteína do Gene 3 de Ativação de Linfócitos
9.
Cell Stem Cell ; 19(2): 205-216, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27184401

RESUMO

The E3 ubiquitin ligase Mule is often overexpressed in human colorectal cancers, but its role in gut tumorigenesis is unknown. Here, we show in vivo that Mule controls murine intestinal stem and progenitor cell proliferation by modulating Wnt signaling via c-Myc. Mule also regulates protein levels of the receptor tyrosine kinase EphB3 by targeting it for proteasomal and lysosomal degradation. In the intestine, EphB/ephrinB interactions position cells along the crypt-villus axis and compartmentalize incipient colorectal tumors. Our study thus unveils an important new avenue by which Mule acts as an intestinal tumor suppressor by regulation of the intestinal stem cell niche.


Assuntos
Efrina-B3/metabolismo , Intestinos/citologia , Lisossomos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Nicho de Células-Tronco , Ubiquitina-Proteína Ligases/metabolismo , Via de Sinalização Wnt , Polipose Adenomatosa do Colo/patologia , Alelos , Animais , Carcinogênese/metabolismo , Carcinogênese/patologia , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Endocitose , Células HEK293 , Humanos , Camundongos Knockout , Modelos Biológicos , Mutação/genética , Celulas de Paneth/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Supressoras de Tumor , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/deficiência
10.
Immunity ; 44(3): 527-541, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26982363

RESUMO

Lymphopoiesis requires the activation of lineage-specific genes embedded in naive, inaccessible chromatin or in primed, accessible chromatin. The mechanisms responsible for de novo gain of chromatin accessibility, known as "pioneer" function, remain poorly defined. Here, we showed that the EBF1 C-terminal domain (CTD) is required for the regulation of a specific gene set involved in B cell fate decision and differentiation, independently of activation and repression functions. Using genome-wide analysis of DNaseI hypersensitivity and DNA methylation in multipotent Ebf1(-/-) progenitors and derivative EBF1wt- or EBF1ΔC-expressing cells, we found that the CTD promoted chromatin accessibility and DNA demethylation in previously naive chromatin. The CTD allowed EBF1 to bind at inaccessible genomic regions that offer limited co-occupancy by other transcription factors, whereas the CTD was dispensable for EBF1 binding at regions that are occupied by multiple transcription factors. Thus, the CTD enables EBF1 to confer permissive lineage-specific changes in progenitor chromatin landscape.


Assuntos
Linfócitos B/fisiologia , Cromatina/metabolismo , Células Progenitoras Linfoides/fisiologia , Transativadores/metabolismo , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Células Cultivadas , Metilação de DNA/genética , Redes Reguladoras de Genes/genética , Linfopoese , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Estrutura Terciária de Proteína/genética , Transativadores/genética
11.
Immunity ; 43(1): 9-11, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26200008

RESUMO

Passenger mutations specific to particular mouse strains can distort experimental outcomes. In this issue of Immunity, Vanden Berghe et al. (2015) demonstrate that passenger mutations are frequent in most genetically engineered congenic mice and persist even after extensive backcrossing.


Assuntos
Variação Genética/genética , Genoma/genética , Camundongos Endogâmicos C57BL/genética , Animais
12.
Nat Immunol ; 14(8): 867-75, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23812095

RESUMO

The transcription factors EBF1 and Pax5 have been linked to activation of the B cell lineage program and irreversible loss of alternative lineage potential (commitment), respectively. Here we conditionally deleted Ebf1 in committed pro-B cells after transfer into alymphoid mice. We found that those cells converted into innate lymphoid cells (ILCs) and T cells with variable-diversity-joining (VDJ) rearrangements of loci encoding both B cell and T cell antigen receptors. As intermediates in lineage conversion, Ebf1-deficient CD19(+) cells expressing Pax5 and transcriptional regulators of the ILC and T cell fates were detectable. In particular, genes encoding the transcription factors Id2 and TCF-1 were bound and repressed by EBF1. Thus, both EBF1 and Pax5 are required for B lineage commitment by repressing distinct and common determinants of alternative cell fates.


Assuntos
Linfócitos B/imunologia , Transativadores/imunologia , Transferência Adotiva , Animais , Linfócitos B/citologia , Diferenciação Celular/imunologia , Linhagem da Célula , DNA/química , DNA/genética , Regulação da Expressão Gênica , Linfopoese/imunologia , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Transativadores/genética , Recombinação V(D)J/genética , Recombinação V(D)J/imunologia
13.
Dev Cell ; 23(4): 866-71, 2012 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-23079603

RESUMO

Satb1 and Satb2 have been recently described as regulators of embryonic stem (ES) cell pluripotency and as silencing factors in X chromosome inactivation. The influence of the pluripotency machinery on X chromosome inactivation and the lack of an X chromosome inactivation defect in Satb1(-/-) and Satb2(-/-) mice raise the question of whether or not Satb proteins are directly and/or redundantly involved in this process. Here, we analyzed X chromosome inactivation in fibroblastic cells that were derived from female Satb1(-/-)Satb2(-/-) embryos. By fluorescence in situ hybridization to visualize Xist RNA and by immunohistochemistry to detect H3K27me3 histone modifications, we found that female Satb1(-/-)Satb2(-/-) fibroblastic cells contain proper Barr bodies. Moreover, we did not detect an upregulation of X-linked genes, suggesting that Satb proteins are dispensable for X chromosome inactivation in mice.


Assuntos
Proteínas de Ligação à Região de Interação com a Matriz/deficiência , Fatores de Transcrição/deficiência , Inativação do Cromossomo X , Animais , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Camundongos , Fatores de Transcrição/metabolismo , Inativação do Cromossomo X/genética
14.
Genes Dev ; 26(7): 668-82, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22431510

RESUMO

The transcription factor Ebf1 is an important determinant of early B lymphopoiesis. To gain insight into the functions of Ebf1 at distinct stages of differentiation, we conditionally inactivated Ebf1. We found that Ebf1 is required for the proliferation, survival, and signaling of pro-B cells and peripheral B-cell subsets, including B1 cells and marginal zone B cells. The proliferation defect of Ebf1-deficient pro-B cells and the impaired expression of multiple cell cycle regulators are overcome by transformation with v-Abl. The survival defect of transformed Ebf1(fl/fl) pro-B cells can be rescued by the forced expression of the Ebf1 targets c-Myb or Bcl-x(L). In mature B cells, Ebf1 deficiency interferes with signaling via the B-cell-activating factor receptor (BAFF-R)- and B-cell receptor (BCR)-dependent Akt pathways. Moreover, Ebf1 is required for germinal center formation and class switch recombination. Genome-wide analyses of Ebf1-mediated gene expression and chromatin binding indicate that Ebf1 regulates both common and distinct sets of genes in early and late stage B cells. By regulating important components of transcription factor and signaling networks, Ebf1 appears to be involved in the coordination of cell proliferation, survival, and differentiation at multiple stages of B lymphopoiesis.


Assuntos
Linfócitos B/citologia , Linfócitos B/metabolismo , Diferenciação Celular , Proliferação de Células , Transdução de Sinais , Transativadores/metabolismo , Animais , Linfócitos B/imunologia , Sobrevivência Celular , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Cadeias Pesadas de Imunoglobulinas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transativadores/genética , Transativadores/imunologia , Transcrição Gênica
15.
Genes Dev ; 23(22): 2625-38, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19933152

RESUMO

Satb1 and the closely related Satb2 proteins regulate gene expression and higher-order chromatin structure of multigene clusters in vivo. In examining the role of Satb proteins in murine embryonic stem (ES) cells, we find that Satb1(-/-) cells display an impaired differentiation potential and augmented expression of the pluripotency determinants Nanog, Klf4, and Tbx3. Metastable states of self-renewal and differentiation competence have been attributed to heterogeneity of ES cells in the expression of Nanog. Satb1(-/-) cultures have a higher proportion of Nanog(high) cells, and an increased potential to reprogram human B lymphocytes in cell fusion experiments. Moreover, Satb1-deficient ES cells show an increased expression of Satb2, and we find that forced Satb2 expression in wild-type ES cells antagonizes differentiation-associated silencing of Nanog and enhances the induction of NANOG in cell fusions with human B lymphocytes. An antagonistic function of Satb1 and Satb2 is also supported by the almost normal differentiation potential of Satb1(-/-)Satb2(-/-) ES cells. Taken together with the finding that both Satb1 and Satb2 bind the Nanog locus in vivo, our data suggest that the balance of Satb1 and Satb2 contributes to the plasticity of Nanog expression and ES cell pluripotency.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linfócitos B/metabolismo , Linhagem Celular , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Proteína Homeobox Nanog
16.
J Immunol ; 181(8): 5213-7, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18832674

RESUMO

T cell development, originally thought to be completed in the thymus, has recently been shown to continue for several weeks in the lymphoid periphery. The forces that drive this peripheral maturation are unclear. The use of mice transgenic for GFP driven by the RAG2 promoter has enabled the ready identification and analysis of recent thymic emigrants. Here, we show that recent thymic emigrant maturation is a progressive process and is promoted by T cell exit from the thymus. Further, we show that this maturation occurs within secondary lymphoid organs and does not require extensive lymphocyte recirculation.


Assuntos
Movimento Celular/imunologia , Timo/imunologia , Animais , Movimento Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/imunologia , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/imunologia , Timo/citologia , Transgenes/genética , Transgenes/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA