Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Front Immunol ; 13: 854312, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35757763

RESUMO

Natural killer (NK) cells play roles in viral clearance and early surveillance against malignant transformation, yet our knowledge of the underlying mechanisms controlling their development and functions remain incomplete. To reveal cell fate-determining pathways in NK cell progenitors (NKP), we utilized an unbiased approach and generated comprehensive gene expression profiles of NK cell progenitors. We found that the NK cell program was gradually established in the CLP to preNKP and preNKP to rNKP transitions. In line with FOXO1 and FOXO3 being co-expressed through the NK developmental trajectory, the loss of both perturbed the establishment of the NK cell program and caused stalling in both NK cell development and maturation. In addition, we found that the combined loss of FOXO1 and FOXO3 caused specific changes to the composition of the non-cytotoxic innate lymphoid cell (ILC) subsets in bone marrow, spleen, and thymus. By combining transcriptome and chromatin profiling, we revealed that FOXO TFs ensure proper NK cell development at various lineage-commitment stages through orchestrating distinct molecular mechanisms. Combined FOXO1 and FOXO3 deficiency in common and innate lymphoid cell progenitors resulted in reduced expression of genes associated with NK cell development including ETS-1 and their downstream target genes. Lastly, we found that FOXO1 and FOXO3 controlled the survival of committed NK cells via gene regulation of IL-15Rß (CD122) on rNKPs and bone marrow NK cells. Overall, we revealed that FOXO1 and FOXO3 function in a coordinated manner to regulate essential developmental genes at multiple stages during murine NK cell and ILC lineage commitment.


Assuntos
Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Células Matadoras Naturais , Células Progenitoras Linfoides , Animais , Diferenciação Celular/imunologia , Proteína Forkhead Box O1/imunologia , Proteína Forkhead Box O3/imunologia , Imunidade Inata , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/imunologia , Camundongos , Camundongos Endogâmicos C57BL
2.
J Immunol ; 208(5): 1066-1075, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35140132

RESUMO

BATF3-deficient mice that lack CD8+ dendritic cells (DCs) showed an exacerbation of chronic graft-versus-host disease (cGVHD), including T follicular helper (Tfh) cell and autoantibody responses, whereas mice carrying the Sle2c2 lupus-suppressive locus with a mutation in the G-CSFR showed an expansion of CD8+ DCs and a poor mobilization of plasmacytoid DCs (pDCs) and responded poorly to cGVHD induction. Here, we investigated the contribution of CD8+ DCs and pDCs to the humoral response to protein immunization, where CD8neg DCs are thought to represent the major inducers. Both BATF3-/- and Sle2c2 mice had reduced humoral and germinal center (GC) responses compared with C57BL/6 (B6) controls. We showed that B6-derived CD4+ DCs are the major early producers of IL-6, followed by CD4-CD8- DCs. Surprisingly, IL-6 production and CD80 expression also increased in CD8+ DCs after immunization, and B6-derived CD8+ DCs rescued Ag-specific adaptive responses in BATF3-/- mice. In addition, inflammatory pDCs (ipDCs) produced more IL-6 than all conventional DCs combined. Interestingly, G-CSFR is highly expressed on pDCs. G-CSF expanded pDC and CD8+ DC numbers and IL-6 production by ipDCs and CD4+ DCs, and it improved the quality of Ab response, increasing the localization of Ag-specific T cells to the GC. Finally, G-CSF activated STAT3 in early G-CSFR+ common lymphoid progenitors of cDCs/pDCs but not in mature cells. In conclusion, we showed a multilayered role of DC subsets in priming Tfh cells in protein immunization, and we unveiled the importance of G-CSFR signaling in the development and function pDCs.


Assuntos
Células Dendríticas/imunologia , Doença Enxerto-Hospedeiro/imunologia , Células Progenitoras Linfoides/citologia , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Células T Auxiliares Foliculares/imunologia , Transferência Adotiva , Animais , Autoanticorpos/imunologia , Antígeno B7-1/biossíntese , Fatores de Transcrição de Zíper de Leucina Básica/genética , Antígenos CD4/biossíntese , Antígenos CD8/biossíntese , Diferenciação Celular/imunologia , Células Dendríticas/transplante , Feminino , Fator Estimulador de Colônias de Granulócitos/metabolismo , Interleucina-6/biossíntese , Ativação Linfocitária/imunologia , Células Progenitoras Linfoides/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Fator Estimulador de Colônias de Granulócitos/genética , Proteínas Repressoras/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/imunologia
3.
Cell Rep ; 38(3): 110266, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35045305

RESUMO

Production of effector CD8+ T cells during persistent infection requires a stable pool of stem-like cells that can give rise to effector cells via a proliferative intermediate population. In infection models marked by T cell exhaustion, this process can be transiently induced by checkpoint blockade but occurs spontaneously in mice chronically infected with the protozoan intracellular parasite Toxoplasma gondii. We observe distinct locations for parasite-specific T cell subsets, implying a link between differentiation and anatomical niches in the spleen. Loss of the chemokine receptor CXCR3 on T cells does not prevent white pulp-to-red pulp migration but reduces interactions with CXCR3 ligand-producing dendritic cells (DCs) and impairs memory-to-intermediate transition, leading to a buildup of memory T cells in the red pulp. Thus, CXCR3 increases T cell exposure to differentiation-inducing signals during red pulp migration, providing a dynamic mechanism for modulating effector differentiation in response to environmental signals.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Células Progenitoras Linfoides/imunologia , Receptores CXCR3/imunologia , Baço/imunologia , Animais , Camundongos , Infecção Persistente/imunologia , Toxoplasmose Animal/imunologia
4.
J Exp Med ; 219(2)2022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-34958351

RESUMO

During dendritic cell (DC) development, Myc expression in progenitors is replaced by Mycl in mature DCs, but when and how this transition occurs is unknown. We evaluated DC development using reporters for MYC, MYCL, and cell cycle proteins Geminin and CDT1 in wild-type and various mutant mice. For classical type 1 dendritic cells (cDC1s) and plasmacytoid DCs (pDCs), the transition occurred upon their initial specification from common dendritic cell progenitors (CDPs) or common lymphoid progenitors (CLPs), respectively. This transition required high levels of IRF8 and interaction with PU.1, suggesting the use of EICEs within Mycl enhancers. In pDCs, maximal MYCL induction also required the +41kb Irf8 enhancer that controls pDC IRF8 expression. IRF8 also contributed to repression of MYC. While MYC is expressed only in rapidly dividing DC progenitors, MYCL is most highly expressed in DCs that have exited the cell cycle. Thus, IRF8 levels coordinate the Myc-Mycl transition during DC development.


Assuntos
Diferenciação Celular/genética , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Genes myc , Fatores Reguladores de Interferon/genética , Animais , Proteínas de Ciclo Celular/genética , Elementos Facilitadores Genéticos , Genes Reporter , Imunofenotipagem , Fatores Reguladores de Interferon/metabolismo , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/imunologia , Células Progenitoras Linfoides/metabolismo , Camundongos , Camundongos Knockout , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo
5.
Mol Biol Rep ; 48(1): 817-822, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33438082

RESUMO

Acute lymphoid leukemia (ALL) is a type of hematological neoplasm that affects the precursor cells of strains B, T  and NK, with a higher incidence in the pediatric range. The pathophysiology of ALL is characterized by chromosomal abnormalities and genetic alterations involved in the differentiation and proliferation of lymphoid precursor cells. Despite the lack of information in the literature, it is believed that leukemogenesis originates from a complex interaction between environmental and genetic factors, which combined lead to cellular modifications. Environmental factors have been evaluated as possible predisposing factors in the development of ALL but there are still conflicting results in the world literature. In this context, the aim of the present review is to discuss the major exogenous factors regarding ALL.


Assuntos
Carcinogênese/imunologia , Regulação Leucêmica da Expressão Gênica/imunologia , Interação Gene-Ambiente , Células Progenitoras Linfoides/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Adulto , Linfócitos B/imunologia , Linfócitos B/patologia , Carcinogênese/genética , Carcinogênese/patologia , Diferenciação Celular , Proliferação de Células , Criança , Aberrações Cromossômicas , Citocinas/genética , Citocinas/imunologia , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Células Progenitoras Linfoides/patologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Linfócitos T/imunologia , Linfócitos T/patologia
6.
Front Immunol ; 12: 825813, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35095929

RESUMO

Protection against pathogen re-infection is mediated, in large part, by two humoral cellular compartments, namely, long-lived plasma cells and memory B cells. Recent data have reinforced the importance of memory B cells, particularly in response to re-infection of different viral subtypes or in response with viral escape mutants. In regard to memory B cell generation, considerable advancements have been made in recent years in elucidating its basic mechanism, which seems to well explain why the memory B cells pool can deal with variant viruses. Despite such progress, efforts to develop vaccines that induce broadly protective memory B cells to fight against rapidly mutating pathogens such as influenza virus and HIV have not yet been successful. Here, we discuss recent advances regarding the key signals and factors regulating germinal center-derived memory B cell development and activation and highlight the challenges for successful vaccine development.


Assuntos
Memória Imunológica , Células B de Memória/imunologia , Células B de Memória/metabolismo , Anticorpos Neutralizantes/imunologia , Formação de Anticorpos/genética , Formação de Anticorpos/imunologia , Comunicação Celular/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Seleção Clonal Mediada por Antígeno , Feminino , Centro Germinativo/citologia , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/imunologia , Células Progenitoras Linfoides/metabolismo , Masculino , Células B de Memória/citologia , Receptores de Antígenos de Linfócitos B/genética , Receptores de Antígenos de Linfócitos B/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
7.
Cancer Immunol Immunother ; 70(5): 1305-1321, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33140189

RESUMO

Allogeneic natural killer (NK) cell transfer is a potential immunotherapy to eliminate and control cancer. A promising source are CD34 + hematopoietic progenitor cells (HPCs), since large numbers of cytotoxic NK cells can be generated. Effective boosting of NK cell function can be achieved by interleukin (IL)-15. However, its in vivo half-life is short and potent trans-presentation by IL-15 receptor α (IL-15Rα) is absent. Therefore, ImmunityBio developed IL-15 superagonist N-803, which combines IL-15 with an activating mutation, an IL-15Rα sushi domain for trans-presentation, and IgG1-Fc for increased half-life. Here, we investigated whether and how N-803 improves HPC-NK cell functionality in leukemia and ovarian cancer (OC) models in vitro and in vivo in OC-bearing immunodeficient mice. We used flow cytometry-based assays, enzyme-linked immunosorbent assay, microscopy-based serial killing assays, and bioluminescence imaging, for in vitro and in vivo experiments. N-803 increased HPC-NK cell proliferation and interferon (IFN)γ production. On leukemia cells, co-culture with HPC-NK cells and N-803 increased ICAM-1 expression. Furthermore, N-803 improved HPC-NK cell-mediated (serial) leukemia killing. Treating OC spheroids with HPC-NK cells and N-803 increased IFNγ-induced CXCL10 secretion, and target killing after prolonged exposure. In immunodeficient mice bearing human OC, N-803 supported HPC-NK cell persistence in combination with total human immunoglobulins to prevent Fc-mediated HPC-NK cell depletion. Moreover, this combination treatment decreased tumor growth. In conclusion,  N-803 is a promising IL-15-based compound that boosts HPC-NK cell expansion and functionality in vitro and in vivo. Adding N-803 to HPC-NK cell therapy could improve cancer immunotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Interleucina-15/agonistas , Células Matadoras Naturais/imunologia , Leucemia/terapia , Células Progenitoras Linfoides/imunologia , Neoplasias Ovarianas/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Antígenos CD34/metabolismo , Antineoplásicos/farmacologia , Diferenciação Celular , Linhagem Celular Tumoral , Testes Imunológicos de Citotoxicidade , Modelos Animais de Doenças , Feminino , Humanos , Interferon gama/metabolismo , Células Matadoras Naturais/transplante , Leucemia/imunologia , Células Progenitoras Linfoides/transplante , Camundongos , Camundongos SCID , Neoplasias Ovarianas/imunologia , Proteínas Recombinantes de Fusão/farmacologia
8.
Nat Immunol ; 21(12): 1574-1584, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33077975

RESUMO

A classical view of blood cell development is that multipotent hematopoietic stem and progenitor cells (HSPCs) become lineage-restricted at defined stages. Lin-c-Kit+Sca-1+Flt3+ cells, termed lymphoid-primed multipotent progenitors (LMPPs), have lost megakaryocyte and erythroid potential but are heterogeneous in their fate. Here, through single-cell RNA sequencing, we identify the expression of Dach1 and associated genes in this fraction as being coexpressed with myeloid/stem genes but inversely correlated with lymphoid genes. Through generation of Dach1-GFP reporter mice, we identify a transcriptionally and functionally unique Dach1-GFP- subpopulation within LMPPs with lymphoid potential with low to negligible classic myeloid potential. We term these 'lymphoid-primed progenitors' (LPPs). These findings define an early definitive branch point of lymphoid development in hematopoiesis and a means for prospective isolation of LPPs.


Assuntos
Biomarcadores , Proteínas do Olho/metabolismo , Genômica , Células Progenitoras Linfoides/metabolismo , Análise de Célula Única , Animais , Células Cultivadas , Biologia Computacional/métodos , Proteínas do Olho/genética , Perfilação da Expressão Gênica , Genômica/métodos , Hematopoese/genética , Sequenciamento de Nucleotídeos em Larga Escala , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteômica , Análise de Célula Única/métodos
9.
Nat Immunol ; 21(12): 1552-1562, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33046887

RESUMO

T cell memory relies on the generation of antigen-specific progenitors with stem-like properties. However, the identity of these progenitors has remained unclear, precluding a full understanding of the differentiation trajectories that underpin the heterogeneity of antigen-experienced T cells. We used a systematic approach guided by single-cell RNA-sequencing data to map the organizational structure of the human CD8+ memory T cell pool under physiological conditions. We identified two previously unrecognized subsets of clonally, epigenetically, functionally, phenotypically and transcriptionally distinct stem-like CD8+ memory T cells. Progenitors lacking the inhibitory receptors programmed death-1 (PD-1) and T cell immunoreceptor with Ig and ITIM domains (TIGIT) were committed to a functional lineage, whereas progenitors expressing PD-1 and TIGIT were committed to a dysfunctional, exhausted-like lineage. Collectively, these data reveal the existence of parallel differentiation programs in the human CD8+ memory T cell pool, with potentially broad implications for the development of immunotherapies and vaccines.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Memória Imunológica , Células Progenitoras Linfoides/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Animais , Biomarcadores , Diferenciação Celular/imunologia , Biologia Computacional/métodos , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imunofenotipagem , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/imunologia , Camundongos , Homeostase do Telômero
10.
Immunity ; 53(4): 775-792.e9, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-33002412

RESUMO

Innate lymphoid cells (ILCs) are generated early during ontogeny and persist predominantly as tissue-resident cells. Here, we examined how ILCs are maintained and renewed within tissues. We generated a single cell atlas of lung ILC2s and found that Il18r1+ ILCs comprise circulating and tissue-resident ILC progenitors (ILCP) and effector-cells with heterogeneous expression of the transcription factors Tcf7 and Zbtb16, and CD103. Our analyses revealed a continuous differentiation trajectory from Il18r1+ ST2- ILCPs to Il18r- ST2+ ILC2s, which was experimentally validated. Upon helminth infection, recruited and BM-derived cells generated the entire spectrum of ILC2s in parabiotic and shield chimeric mice, consistent with their potential role in the renewal of tissue ILC2s. Our findings identify local ILCPs and reveal ILCP in situ differentiation and tissue adaptation as a mechanism of ILC maintenance and phenotypic diversification. Local niches, rather than progenitor origin, or the developmental window during ontogeny, may dominantly imprint ILC phenotypes in adult tissues.


Assuntos
Imunidade Inata/imunologia , Linfócitos/imunologia , Células Progenitoras Linfoides/imunologia , Animais , Diferenciação Celular/imunologia , Células Cultivadas , Feminino , Humanos , Subunidade alfa de Receptor de Interleucina-18/imunologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína com Dedos de Zinco da Leucemia Promielocítica/imunologia , Transdução de Sinais/imunologia , Análise de Célula Única/métodos , Fator 1 de Transcrição de Linfócitos T/imunologia , Fatores de Transcrição/imunologia
11.
J Immunol ; 204(9): 2600-2611, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32213568

RESUMO

Accurately tuned expression levels of the transcription factor GATA-3 are crucial at several stages of T cell and innate lymphoid cell development and differentiation. Moreover, several lines of evidence suggest that Gata3 expression might provide a reliable molecular marker for the identification of elusive progenitor cell subsets at the earliest stages of T lineage commitment. To be able to faithfully monitor Gata3 expression noninvasively at the single-cell level, we have generated a novel strain of knock-in reporter mice, termed GATIR, by inserting an expression cassette encoding a bright fluorescent marker into the 3'-untranslated region of the endogenous Gata3 locus. Importantly, in contrast to three previously published strains of Gata3 reporter mice, GATIR mice preserve physiological Gata3 expression on the targeted allele. In this study, we show that GATIR mice faithfully reflect endogenous Gata3 expression without disturbing the development of GATA-3-dependent lymphoid cell populations. We further show that GATIR mice provide an ideal tool for noninvasive monitoring of Th2 polarization and straightforward identification of innate lymphoid cell 2 progenitor populations. Finally, as our reporter is non-gene-destructive, GATIR mice can be bred to homozygosity, not feasible with previously published strains of Gata3 reporter mice harboring disrupted alleles. The availability of hetero- and homozygous Gata3 reporter mice with an exceptionally bright fluorescent marker, allowed us to visualize allelic Gata3 expression in individual cells simply by flow cytometry. The unambiguous results obtained provide compelling evidence against previously postulated monoallelic Gata3 expression in early T lineage and hematopoietic stem cell subsets.


Assuntos
Fator de Transcrição GATA3/genética , Genes Reporter/genética , Regiões 3' não Traduzidas/genética , Regiões 3' não Traduzidas/imunologia , Alelos , Animais , Biomarcadores/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Feminino , Citometria de Fluxo/métodos , Corantes Fluorescentes/metabolismo , Fator de Transcrição GATA3/imunologia , Técnicas de Introdução de Genes/métodos , Genes Reporter/imunologia , Células-Tronco Hematopoéticas/imunologia , Imunidade Inata/genética , Imunidade Inata/imunologia , Linfócitos/imunologia , Células Progenitoras Linfoides/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia
12.
Methods Mol Biol ; 2121: 7-22, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32147782

RESUMO

Understanding the origins and developmental trajectory of innate lymphoid cell (ILC) progenitors has been of substantial interest to the fields of ILC biology and immunology. While mature ILC are rare lymphocytes, ILC progenitors represent an even smaller fraction of cells, providing additional challenges in studying them. Moreover, though the approaches to studying these cells are conceptually straightforward, the technical nuances that underlie them can substantially affect the quality of the data. Herein, we provide a detailed protocol for assessing the frequency of ILC progenitors in the bone marrow, their phenotype, and their potential to develop into mature ILC. These methods make up the foundation of in vivo investigations into ILC development, and we hope these thorough protocols and associated notes facilitate additional, high-quality inquiries into this fascinating field.


Assuntos
Transferência Adotiva/métodos , Células da Medula Óssea , Células Matadoras Naturais/citologia , Fígado/citologia , Linfócitos/citologia , Células Progenitoras Linfoides/citologia , Linfopoese/imunologia , Animais , Medula Óssea , Células da Medula Óssea/citologia , Linhagem da Célula , Feminino , Citometria de Fluxo , Células Matadoras Naturais/imunologia , Fígado/imunologia , Linfócitos/imunologia , Células Progenitoras Linfoides/imunologia , Células Progenitoras Linfoides/metabolismo , Camundongos , Camundongos Endogâmicos BALB C
13.
J Immunol ; 204(9): 2447-2454, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32198141

RESUMO

The caudal hematopoietic tissue in zebrafish, the equivalent to the fetal liver in mammals, is an intermediate hematopoietic niche for the maintenance and differentiation of hematopoietic stem and progenitor cells before homing to the thymus and kidney marrow. As one of the ultimate hematopoietic organs, the thymus sustains T lymphopoiesis, which is essential for adaptive immune system. However, the mechanism of prethymic T lymphoid progenitors migrating to the thymus remains elusive. In this study, we identify an Rho GTPase Rac2 as a modulator of T lymphoid progenitor homing to the thymus in zebrafish. rac2-Deficient embryos show the inability of T lymphoid progenitors homing to the thymus because of defective cell-autonomous motility. Mechanistically, we demonstrate that Rac2 regulates homing of T lymphoid progenitor through Pak1-mediated AKT pathway. Taken together, our work reveals an important function of Rac2 in directing T lymphoid progenitor migration to the thymus during zebrafish embryogenesis.


Assuntos
Movimento Celular/imunologia , Desenvolvimento Embrionário/imunologia , Células Progenitoras Linfoides/metabolismo , Timo/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Animais Geneticamente Modificados/imunologia , Animais Geneticamente Modificados/metabolismo , Medula Óssea/imunologia , Medula Óssea/metabolismo , Diferenciação Celular/imunologia , Células Progenitoras Linfoides/imunologia , Linfopoese/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/imunologia , Timo/imunologia , Peixe-Zebra/imunologia , Proteínas de Peixe-Zebra/imunologia , Quinases Ativadas por p21/imunologia , Quinases Ativadas por p21/metabolismo , Proteínas rac de Ligação ao GTP/imunologia , Proteínas rho de Ligação ao GTP/imunologia , Proteínas rho de Ligação ao GTP/metabolismo
14.
Eur J Immunol ; 50(7): 959-971, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32090320

RESUMO

The transcription factor Hhex (hematopoietically expressed homeobox gene) is critical for development of multiple lymphoid lineages beyond the common lymphoid progenitor. In addition, Hhex regulates hematopoietic stem cell (HSC) self-renewal, emergency hematopoiesis, and acute myeloid leukemia initiation and maintenance. Hhex mediates its effects on HSCs and acute myeloid leukemia stem cells via repression of the Cdkn2a tumor suppressor locus. However, we report here that loss of Cdkn2a does not rescue the failure of lymphoid development caused by loss of Hhex. As loss of Hhex causes apoptosis of lymphoid progenitors associated with impaired Bcl2 expression and defective Stat5b signaling, we tested the effects of rescuing these pathways using transgenic mice. Expression of the anti-apoptotic factor Bcl2, but not activated Stat5, rescued the development of T-, B-, and NK-cell lineages in the absence of Hhex. These results indicate that Bcl2 expression, but not Stat5b signaling or loss of Cdkn2a, can overcome the lymphoid deficiencies caused by the absence of Hhex, suggesting that the primary role of this transcription factor is to promote survival of lymphoid progenitors during early lymphoid development.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/imunologia , Proteínas de Homeodomínio/imunologia , Células Progenitoras Linfoides/imunologia , Fator de Transcrição STAT5/imunologia , Transdução de Sinais/imunologia , Fatores de Transcrição/imunologia , Animais , Apoptose/genética , Apoptose/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Inibidor p16 de Quinase Dependente de Ciclina/genética , Proteínas de Homeodomínio/genética , Células Progenitoras Linfoides/citologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Fator de Transcrição STAT5/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética
15.
Annu Rev Immunol ; 38: 229-247, 2020 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-31928469

RESUMO

Neonatal CD4+ and CD8+ T cells have historically been characterized as immature or defective. However, recent studies prompt a reinterpretation of the functions of neonatal T cells. Rather than a population of cells always falling short of expectations set by their adult counterparts, neonatal T cells are gaining recognition as a distinct population of lymphocytes well suited for the rapidly changing environment in early life. In this review, I will highlight new evidence indicating that neonatal T cells are not inert or less potent versions of adult T cells but instead are a broadly reactive layer of T cells poised to quickly develop into regulatory or effector cells, depending on the needs of the host. In this way, neonatal T cells are well adapted to provide fast-acting immune protection against foreign pathogens, while also sustaining tolerance to self-antigens.


Assuntos
Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Imunidade Adaptativa , Animais , Biomarcadores , Diferenciação Celular/imunologia , Interações Hospedeiro-Patógeno , Humanos , Memória Imunológica , Ativação Linfocitária/imunologia , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/imunologia , Células Progenitoras Linfoides/metabolismo , Fenótipo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Subpopulações de Linfócitos T/citologia
16.
Adv Immunol ; 143: 99-119, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31607369

RESUMO

Rapid advances have been made to uncover the mechanisms that regulate dendritic cell (DC) development, and in turn, how models of development can be employed to define dendritic cell function. Models of DC development have been used to define the unique functions of DC subsets during immune responses to distinct pathogens. More recently, models of DC function have expanded to include their homeostatic and inflammatory physiology, modes of communication with various innate and adaptive immune lineages, and specialized functions across different lymphoid organs. New models of DC development call for revisions of previously accepted paradigms with respect to the ontogeny of plasmacytoid DC (pDC) and classical DC (cDC) subsets. By far, development of the cDC1 subset is best understood, and models have now been developed that can separate deficiencies in development from deficiencies in function. Such models are lacking for pDCs and cDC2s, limiting the depth of our understanding of their unique and essential roles during immune responses. If novel immunotherapies aim to harness the functions of human DCs, understanding of DC development will be essential to develop models DC function. Here we review emerging models of DC development and function.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Células Progenitoras Linfoides/imunologia , Células Progenitoras Mieloides/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Células Dendríticas/citologia , Humanos , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Células Progenitoras Linfoides/metabolismo , Camundongos , Modelos Imunológicos , Células Progenitoras Mieloides/metabolismo , Homeobox 2 de Ligação a E-box com Dedos de Zinco/genética , Homeobox 2 de Ligação a E-box com Dedos de Zinco/metabolismo
17.
Front Immunol ; 10: 2045, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31555276

RESUMO

NK cells are generated from hematopoietic stem cells (HSC) residing in the bone marrow (BM), similar to other blood cells. Development toward mature NK cells occurs largely outside the BM through travel of CD34+ and other progenitor intermediates toward secondary lymphoid organs. The BM harbors multipotent CD34+ common lymphoid progenitors (CLPs) that generate T, B, NK, and Dendritic Cells and are devoid of erythroid, myeloid, and megakaryocytic potential. Over recent years, there has been a quest for single-lineage progenitors predominantly with the objective of manipulation and intervention in mind, which has led to the identification of unipotent NK cell progenitors devoid of other lymphoid lineage potential. Research efforts for the study of lymphopoiesis have almost exclusively concentrated on healthy donor tissues and on repopulation/transplant models. This has led to the widely accepted assumption that lymphopoiesis during disease states reflects the findings of these models. However, compelling evidences in animal models show that inflammation plays a fundamental role in the regulation of HSC maturation and release in the BM niches through several mechanisms including modulation of the CXCL12-CXCR4 expression. Indeed, recent findings during systemic inflammation in patients provide evidence that a so-far overlooked CLP exists in the BM (Lin-CD34+DNAM-1brightCXCR4+) and that it overwhelmingly exits the BM during systemic inflammation. These "inflammatory" precursors have a developmental trajectory toward surprisingly functional NK and T cells as reviewed here and mirror the steady state maintenance of the NK cell pool by CD34+DNAM-1-CXCR4- precursors. Our understanding of NK cell precursor development may benefit from including a distinct "inflammatory" progenitor modeling of lymphoid precursors, allowing rapid deployment of specialized Lin-CD34+DNAM-1brightCXCR4+ -derived resources from the BM.


Assuntos
Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Homeostase , Células Progenitoras Linfoides/imunologia , Células Progenitoras Linfoides/metabolismo , Animais , Biomarcadores , Células da Medula Óssea/citologia , Diferenciação Celular/imunologia , Suscetibilidade a Doenças , Humanos , Imunofenotipagem , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Células Progenitoras Linfoides/citologia , Nicho de Células-Tronco
18.
J Immunol ; 202(10): 2837-2842, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30962294

RESUMO

Lymphoid specification is the process by which hematopoietic stem cells (HSCs) and their progeny become restricted to differentiation through the lymphoid lineages. The basic helix-loop-helix transcription factors E2A and Lyl1 form a complex that promotes lymphoid specification. In this study, we demonstrate that Tal1, a Lyl1-related basic helix-loop-helix transcription factor that promotes T acute lymphoblastic leukemia and is required for HSC specification, erythropoiesis, and megakaryopoiesis, is a negative regulator of murine lymphoid specification. We demonstrate that Tal1 limits the expression of multiple E2A target genes in HSCs and controls the balance of myeloid versus T lymphocyte differentiation potential in lymphomyeloid-primed progenitors. Our data provide insight into the mechanisms controlling lymphocyte specification and may reveal a basis for the unique functions of Tal1 and Lyl1 in T acute lymphoblastic leukemia.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/imunologia , Diferenciação Celular/imunologia , Células Progenitoras Linfoides/imunologia , Células Progenitoras Mieloides/imunologia , Proteínas de Neoplasias/imunologia , Proteína 1 de Leucemia Linfocítica Aguda de Células T/imunologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Proteína 1 de Leucemia Linfocítica Aguda de Células T/genética
19.
Int Immunol ; 31(8): 489-498, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-30783658

RESUMO

Innate lymphoid cells (ILCs), composed of heterogeneous populations of lymphoid cells, contribute critically to immune surveillance at mucosal surfaces. ILC subsets develop from common lymphoid progenitors through stepwise lineage specification. However, the composition and temporal regulation of the transcription factor network governing such a process remain incompletely understood. Here, we report that deletion of the transcription factor interferon regulatory factor 2 (IRF-2), known also for its importance in the maturation of conventional NK cells, resulted in an impaired generation of ILC1, ILC2 and ILC3 subsets with lymphoid tissue inducer (LTi)-like cells hardly affected. In IRF-2-deficient mice, PD-1hi ILC precursors (ILCPs) that generate these three ILCs but not LTi-like cells were present at normal frequency, while their sub-population expressing high amounts of PLZF, another marker for ILCPs, was severely reduced. Notably, these IRF-2-deficient ILCPs contained normal quantities of PLZF-encoding Zbtb16 messages, and PLZF expression in developing invariant NKT cells within the thymus was unaffected in these mutant mice. These results point to a unique, cell-type selective role for IRF-2 in ILC development, acting at a discrete step critical for the generation of functionally competent ILCPs.


Assuntos
Imunidade Inata/imunologia , Fator Regulador 2 de Interferon/imunologia , Linfócitos/imunologia , Células Progenitoras Linfoides/imunologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
20.
Methods Mol Biol ; 1884: 73-86, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30465196

RESUMO

Single-cell RNA sequencing (scRNA-seq) is a powerful tool to study immune cells, which enables an unbiased way to discover novel cell populations, biological meaningful cellular heterogeneity, and cell lineage development trajectories. Advances in scRNA-seq technologies and computational data analysis have driven a revolution in our understanding of the immune system in health and disease. Technically, the key step for scRNA-seq analysis is making a high-quality cDNA library for sequencing. Here, we describe a plate-based protocol to prepare single-cell cDNA library of bone marrow innate lymphoid precursors for next generation sequencing-based transcriptome analysis.


Assuntos
Células Progenitoras Linfoides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Separação Celular/instrumentação , Separação Celular/métodos , Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Corantes Fluorescentes/química , Perfilação da Expressão Gênica/instrumentação , Perfilação da Expressão Gênica/métodos , Biblioteca Gênica , Linfócitos/imunologia , Células Progenitoras Linfoides/imunologia , Camundongos , RNA/genética , RNA/isolamento & purificação , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/instrumentação , Análise de Sequência de RNA/instrumentação , Análise de Célula Única/instrumentação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...