Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Immunity ; 49(3): 464-476.e4, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30193847

RESUMO

According to the established model of murine innate lymphoid cell (ILC) development, helper ILCs develop separately from natural killer (NK) cells. However, it is unclear how helper ILCs and NK cells develop in humans. Here we elucidated key steps of NK cell, ILC2, and ILC3 development within human tonsils using ex vivo molecular and functional profiling and lineage differentiation assays. We demonstrated that while tonsillar NK cells, ILC2s, and ILC3s originated from a common CD34-CD117+ ILC precursor pool, final steps of ILC2 development deviated independently and became mutually exclusive from those of NK cells and ILC3s, whose developmental pathways overlapped. Moreover, we identified a CD34-CD117+ ILC precursor population that expressed CD56 and gave rise to NK cells and ILC3s but not to ILC2s. These data support a model of human ILC development distinct from the mouse, whereby human NK cells and ILC3s share a common developmental pathway separate from ILC2s.


Assuntos
Células Matadoras Naturais/imunologia , Linfócitos/imunologia , Tonsila Palatina/imunologia , Animais , Antígenos CD34/metabolismo , Antígeno CD56/metabolismo , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Imunidade Inata , Ativação Linfocitária , Camundongos , Proteínas Proto-Oncogênicas c-kit/metabolismo
2.
Immunity ; 47(1): 159-170.e10, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28723548

RESUMO

Clearance of pathogens or tumor cells by antibodies traditionally requires both Fab and Fc domains of IgG. Here, we show the Fc domain of IgG alone mediates recognition and clearance of herpes simplex virus (HSV1)-infected cells. The human natural killer (NK) cell surface is naturally coated with IgG bound by its Fc domain to the Fcγ receptor CD16a. NK cells utilize the Fc domain of bound IgG to recognize gE, an HSV1-encoded glycoprotein that also binds the Fc domain of IgG but at a site distinct from CD16a. The bridge formed by the Fc domain between the HSV1-infected cell and the NK cell results in NK cell activation and lysis of the HSV1-infected cell in the absence of HSV1-specific antibody in vitro and prevents fatal HSV1 infection in vivo. This mechanism also explains how bacterial IgG-binding proteins regulate NK cell function and may be broadly applicable to Fcγ-receptor-bearing cells.


Assuntos
Anticorpos Antivirais/metabolismo , Herpes Simples/imunologia , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/metabolismo , Células Matadoras Naturais/imunologia , Simplexvirus/imunologia , Animais , Anticorpos Antivirais/imunologia , Células Cultivadas , Citotoxicidade Imunológica , Feminino , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ligação Proteica , Agregação de Receptores , Receptores de IgG/metabolismo , Transdução de Sinais , Proteínas Virais/imunologia
3.
Immunity ; 44(5): 1140-50, 2016 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-27178467

RESUMO

The current model of murine innate lymphoid cell (ILC) development holds that mouse ILCs are derived downstream of the common lymphoid progenitor through lineage-restricted progenitors. However, corresponding lineage-restricted progenitors in humans have yet to be discovered. Here we identified a progenitor population in human secondary lymphoid tissues (SLTs) that expressed the transcription factor RORγt and was unique in its ability to generate all known ILC subsets, including natural killer (NK) cells, but not other leukocyte populations. In contrast to murine fate-mapping data, which indicate that only ILC3s express Rorγt, these human progenitor cells as well as human peripheral blood NK cells and all mature ILC populations expressed RORγt. Thus, all human ILCs can be generated through an RORγt(+) developmental pathway from a common progenitor in SLTs. These findings help establish the developmental signals and pathways involved in human ILC development.


Assuntos
Células Matadoras Naturais/fisiologia , Linfonodos/imunologia , Subpopulações de Linfócitos/fisiologia , Células Progenitoras Linfoides/fisiologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Tonsila Palatina/imunologia , Adulto , Animais , Antígenos CD34/metabolismo , Diferenciação Celular , Linhagem Celular , Criança , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética
4.
Immunity ; 42(3): 457-70, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25769609

RESUMO

Little is known about the role of negative regulators in controlling natural killer (NK) cell development and effector functions. Foxo1 is a multifunctional transcription factor of the forkhead family. Using a mouse model of conditional deletion in NK cells, we found that Foxo1 negatively controlled NK cell differentiation and function. Immature NK cells expressed abundant Foxo1 and little Tbx21 relative to mature NK cells, but these two transcription factors reversed their expression as NK cells proceeded through development. Foxo1 promoted NK cell homing to lymph nodes by upregulating CD62L expression and inhibited late-stage maturation and effector functions by repressing Tbx21 expression. Loss of Foxo1 rescued the defect in late-stage NK cell maturation in heterozygous Tbx21(+/-) mice. Collectively, our data reveal a regulatory pathway by which the negative regulator Foxo1 and the positive regulator Tbx21 play opposing roles in controlling NK cell development and effector functions.


Assuntos
Fatores de Transcrição Forkhead/imunologia , Regulação Neoplásica da Expressão Gênica , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/genética , Melanoma Experimental/genética , Neoplasias Cutâneas/genética , Proteínas com Domínio T/imunologia , Animais , Diferenciação Celular , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Heterozigoto , Células Matadoras Naturais/patologia , Selectina L/genética , Selectina L/imunologia , Pulmão/imunologia , Pulmão/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Linfonodos/imunologia , Linfonodos/patologia , Depleção Linfocítica , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Transdução de Sinais , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/secundário , Proteínas com Domínio T/genética
5.
Mol Ther ; 27(5): 922-932, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-30833178

RESUMO

IL-15 is a proinflammatory cytokine that plays an essential role in the development and activation of natural killer (NK) cells. Adipose tissue acts as an endocrine organ that secretes cytokines and is an important reservoir for lymphocytes. We hypothesized that activation of the IL-15 signaling in adipose tissue will activate and expand the NK cell population and control tumor growth. We recently developed an adipocyte-targeting recombinant adeno-associated viral (rAAV) vector with minimal off-target transgene expression in the liver. Here, we used this rAAV system to deliver an IL-15/IL-15Rα complex to the abdominal fat by intraperitoneal (i.p.) injection. Adipose IL-15/IL-15Rα complex gene transfer led to the expansion of NK cells in the adipose tissue and spleen in normal mice without notable side effects. The i.p. injection of rAAV-IL-15/IL-15Rα complex significantly suppressed the growth of Lewis lung carcinoma implanted subcutaneously and exerted a significant survival advantage in a B16-F10 melanoma metastasis model. The antitumor effects were associated with the expansion of the NK cells in the blood, spleen, abdominal fat, and tumor, as well as the enhancement of NK cell maturity. Our proof-of-concept preclinical studies demonstrate the safety and efficacy of the adipocyte-specific IL-15/IL-15Rα complex vector as a novel cancer immune gene therapy.


Assuntos
Terapia Genética , Subunidade alfa de Receptor de Interleucina-15/genética , Interleucina-15/farmacologia , Neoplasias/terapia , Gordura Abdominal/efeitos dos fármacos , Gordura Abdominal/imunologia , Adipócitos/efeitos dos fármacos , Adipócitos/imunologia , Adipócitos/metabolismo , Animais , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/terapia , Proliferação de Células/genética , Dependovirus , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/farmacologia , Humanos , Interleucina-15/genética , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Melanoma Experimental/genética , Melanoma Experimental/terapia , Camundongos , Metástase Neoplásica , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Transdução de Sinais/genética
6.
Blood ; 121(23): 4663-71, 2013 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-23580661

RESUMO

MicroRNAs (miRNAs) bind to complementary sequences of target mRNAs, resulting in translational repression or target degradation and thus gene silencing. miRNAs are abundant in circulating blood, yet it is not known whether, as a class of regulatory molecules, they interact with human natural killer (NK) cells. Here we found that the treatment of human NK cells with several mature miRNAs in the presence of a low concentration of interleukin-12 induced CD69 expression, interferon-γ production, and degranulation marker CD107a expression. In vivo, infusion of several miRNAs alone in murine peripheral blood also resulted in comparable NK-cell activation, but not T-cell activation. Furthermore, miRNA administration significantly protected mice from tumor development in an NK cell-dependent manner. Mechanistically, we found that miRNA stimulation led to downstream activation of nuclear factor κB (NF-κB), an effect that was blunted by a block in Toll-like receptor 1(TLR1) signaling and attenuated in lymphoma patients. Knockdown of TLR1 resulted in less activation by miRNAs. Collectively, we show that miRNAs have a capacity to selectively activate innate immune effector cells that is, at least in part, via the TLR1-NF-κB signaling pathway. This may be important in the normal host defense against infection and/or malignant transformation.


Assuntos
Células Matadoras Naturais/imunologia , Linfoma/prevenção & controle , MicroRNAs/genética , Baço/imunologia , Receptores Toll-Like/metabolismo , Animais , Western Blotting , Células Cultivadas , Citometria de Fluxo , Humanos , Interferon gama/metabolismo , Interleucina-12/genética , Interleucina-12/metabolismo , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Ativação Linfocitária , Linfoma/genética , Linfoma/imunologia , Proteína 1 de Membrana Associada ao Lisossomo/genética , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , NF-kappa B/genética , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/metabolismo , Baço/patologia , Receptores Toll-Like/antagonistas & inibidores , Receptores Toll-Like/genética
7.
Mol Ther ; 22(9): 1678-87, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24895995

RESUMO

Glioblastoma is a devastating disease, and there is an urgent need to develop novel therapies, such as oncolytic HSV1 (OV) to effectively target tumor cells. OV therapy depends on tumor-specific replication leading to destruction of neoplastic tissues. Host responses that curtail virus replication limit its efficacy in vivo. We have previously shown that cysteine-rich 61 protein (CCN1) activates a type 1 IFN antiviral defense response in glioblastoma cells. Incorporating TCGA data, we found CCN1 expression to be a negative prognostic factor for glioblastoma patients. Based on this, we used neutralizing antibodies against CCN1 to investigate its effect on OV therapy. Use of an anti-CCN1 antibody in mice bearing glioblastomas treated with OV led to enhanced virus expression along with reduced immune cell infiltration. OV-induced CCN1 increases macrophage migration toward infected glioblastoma cells by directly binding macrophages and also by enhancing the proinflammatory activation of macrophages inducing MCP-1 expression in glioblastoma cells. Activation of macrophages by CCN1 also increases viral clearance. Neutralization of integrin αMß2 reversed CCN1-induced macrophage activation and migration, and reduced MCP-1 expression by glioblastoma cells. Our findings reveal that CCN1 plays a novel role in pathogen clearance; increasing macrophage infiltration and activation resulting in increased virus clearance in tumors.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Proteína Rica em Cisteína 61/genética , Proteína Rica em Cisteína 61/metabolismo , Glioblastoma/imunologia , Herpesvirus Humano 1/genética , Macrófagos/metabolismo , Animais , Anticorpos Monoclonais/uso terapêutico , Linhagem Celular Tumoral , Quimiocina CCL2/metabolismo , Feminino , Vetores Genéticos/administração & dosagem , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Ativação de Macrófagos , Camundongos , Transplante de Neoplasias , Vírus Oncolíticos/genética
8.
Blood Adv ; 8(2): 429-440, 2024 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-37871309

RESUMO

ABSTRACT: Enasidenib (ENA) is an inhibitor of isocitrate dehydrogenase 2 (IDH2) approved for the treatment of patients with IDH2-mutant relapsed/refractory acute myeloid leukemia (AML). In this phase 2/1b Beat AML substudy, we applied a risk-adapted approach to assess the efficacy of ENA monotherapy for patients aged ≥60 years with newly diagnosed IDH2-mutant AML in whom genomic profiling demonstrated that mutant IDH2 was in the dominant leukemic clone. Patients for whom ENA monotherapy did not induce a complete remission (CR) or CR with incomplete blood count recovery (CRi) enrolled in a phase 1b cohort with the addition of azacitidine. The phase 2 portion assessing the overall response to ENA alone demonstrated efficacy, with a composite complete response (cCR) rate (CR/CRi) of 46% in 60 evaluable patients. Seventeen patients subsequently transitioned to phase 1b combination therapy, with a cCR rate of 41% and 1 dose-limiting toxicity. Correlative studies highlight mechanisms of clonal elimination with differentiation therapy as well as therapeutic resistance. This study demonstrates both efficacy of ENA monotherapy in the upfront setting and feasibility and applicability of a risk-adapted approach to the upfront treatment of IDH2-mutant AML. This trial is registered at www.clinicaltrials.gov as #NCT03013998.


Assuntos
Aminopiridinas , Azacitidina , Leucemia Mieloide Aguda , Triazinas , Humanos , Azacitidina/efeitos adversos , Isocitrato Desidrogenase/genética , Mutação , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Resposta Patológica Completa
9.
J Exp Med ; 204(10): 2397-405, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17875674

RESUMO

Monokines (i.e., interleukin [IL]-12, -18, and -15) induce natural killer (NK) cells to produce interferon-gamma (IFN-gamma), which is a critical factor for immune surveillance of cancer and monocyte clearance of infection. We show that SET, which is a potent inhibitor of protein phosphatase type 2A (PP2A) activity, is highly expressed in human CD56bright NK cells, which produce more IFN-gamma than CD56dim NK cells. SET was up-regulated upon monokine stimulation of primary human NK cells. Furthermore, ectopic overexpression of SET significantly enhanced IFN-gamma gene expression in monokine-stimulated NK cells. In contrast, RNAi-mediated suppression of SET expression renders NK cells inefficient in producing high levels of IFN-gamma in response to monokine costimulation. Mechanistically, suppression of PP2A activity by SET is important for IFN-gamma gene expression in NK cells. In fact, treatment of primary human NK cells with the PP2A activator 1,9-dideoxy-forskolin, as well as administration of the drug to C57BL/6 mice, significantly reduced NK-dependent IFN-gamma production in response to monokine treatment. Further, SET knockdown or pharmacologic activation of PP2A diminished extracellular signal-regulated kinase 1/2, p65RelA, signal transducer and activator of transduction 4 (STAT4), and STAT5 activity in monokine-stimulated NK cells, potentially contributing to the reduction in IFN-gamma gene expression. Thus, SET expression is essential for suppressing PP2A phosphatase activity that would otherwise limit NK cell antitumoral and/or antiinflammatory functions by impairing NK cell production of IFN-gamma.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Interferon gama/biossíntese , Células Matadoras Naturais/metabolismo , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA , Ativação Enzimática , Regulação da Expressão Gênica , Chaperonas de Histonas , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Monocinas/farmacologia , Transdução de Sinais , Fatores de Transcrição/genética
10.
Blood ; 117(8): 2378-84, 2011 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-21156847

RESUMO

The ability of natural killer (NK) cells to kill malignant or infected cells depends on the integration of signals from different families of cell surface receptors, including cytokine receptors. How such signals then regulate NK-cell cytotoxicity is incompletely understood. Here we analyzed an endogenous inhibitor of protein phosphatase 2A (PP2A) activity called SET, and its role in regulating human NK-cell cytotoxicity and its mechanism of action in human NK cells. RNAi-mediated suppression of SET down-modulates NK-cell cytotoxicity, whereas ectopic overexpression of SET enhances cytotoxicity. SET knockdown inhibits both mRNA and protein granzyme B expression, as well as perforin expression, whereas SET overexpression enhances granzyme B expression. Treatment of NK cells with the PP2A activator 1,9-dideoxy-forskolin also inhibits both granzyme B expression and cytotoxicity. In addition, pretreatment with the PP2A inhibitor okadaic acid rescues declining granzyme B mRNA levels in SET knockdown cells. Down-modulation of SET expression or activation of PP2A also decreases human NK-cell antibody-dependent cellular cytotoxicity. Finally, the induction of granzyme B gene expression by interleukin-2 and interleukin-15 is inhibited by SET knockdown. These data provide evidence that granzyme B gene expression and therefore human NK-cell cytotoxicity can be regulated by the PP2A-SET interplay.


Assuntos
Granzimas/genética , Chaperonas de Histonas/fisiologia , Células Matadoras Naturais/metabolismo , Proteína Fosfatase 2/fisiologia , Fatores de Transcrição/fisiologia , Citotoxicidade Imunológica , Proteínas de Ligação a DNA , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Granzimas/biossíntese , Humanos , Células Matadoras Naturais/imunologia , Proteína Fosfatase 2/antagonistas & inibidores , RNA Interferente Pequeno/farmacologia
11.
Cancer Cell ; 8(5): 355-68, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16286244

RESUMO

The oncogenic BCR/ABL kinase activity induces and maintains chronic myelogenous leukemia (CML). We show here that, in BCR/ABL-transformed cells and CML blast crisis (CML-BC) progenitors, the phosphatase activity of the tumor suppressor PP2A is inhibited by the BCR/ABL-induced expression of the PP2A inhibitor SET. In imatinib-sensitive and -resistant (T315I included) BCR/ABL+ cell lines and CML-BC progenitors, molecular and/or pharmacological activation of PP2A promotes dephosphorylation of key regulators of cell proliferation and survival, suppresses BCR/ABL activity, and induces BCR/ABL degradation. Furthermore, PP2A activation results in growth suppression, enhanced apoptosis, restored differentiation, impaired clonogenic potential, and decreased in vivo leukemogenesis of imatinib-sensitive and -resistant BCR/ABL+ cells. Thus, functional inactivation of PP2A is essential for BCR/ABL leukemogenesis and, perhaps, required for blastic transformation.


Assuntos
Crise Blástica/metabolismo , Proteínas Cromossômicas não Histona/fisiologia , Proteínas de Fusão bcr-abl/fisiologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fosfoproteínas Fosfatases/fisiologia , Fatores de Transcrição/fisiologia , Animais , Antineoplásicos/farmacologia , Benzamidas , Linhagem Celular Transformada , Colforsina/farmacologia , Proteínas de Ligação a DNA , Inibidores Enzimáticos/metabolismo , Chaperonas de Histonas , Humanos , Mesilato de Imatinib , Técnicas In Vitro , Células K562 , Leucemia/prevenção & controle , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Camundongos , Camundongos SCID , Transplante de Neoplasias , Fosfoproteínas Fosfatases/antagonistas & inibidores , Piperazinas/farmacologia , Proteína Fosfatase 2 , Pirimidinas/farmacologia , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/fisiologia
12.
J Exp Med ; 203(4): 1033-43, 2006 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-16606675

RESUMO

Human natural killer (NK) cells originate from CD34(+) hematopoietic progenitor cells, but the discrete stages of NK cell differentiation in vivo have not been elucidated. We identify and functionally characterize, from human lymph nodes and tonsils, four NK cell developmental intermediates spanning the continuum of differentiation from a CD34(+) NK cell progenitor to a functionally mature NK cell. Analyses of each intermediate stage for CD34, CD117, and CD94 cell surface expression, lineage differentiation potentials, capacity for cytokine production and natural cytotoxicity, and ETS-1, GATA-3, and T-BET expression provide evidence for a new model of human NK cell differentiation in secondary lymphoid tissues.


Assuntos
Diferenciação Celular/imunologia , Células Matadoras Naturais/citologia , Biomarcadores , Linhagem da Célula/imunologia , Células Cultivadas , Citocinas/biossíntese , Testes Imunológicos de Citotoxicidade , Perfilação da Expressão Gênica , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Linfonodos/citologia , Linfonodos/imunologia , Linfonodos/metabolismo , Tonsila Palatina/citologia , Tonsila Palatina/imunologia , Tonsila Palatina/metabolismo
13.
Blood ; 115(2): 274-81, 2010 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-19897577

RESUMO

Human CD56(bright) natural killer (NK) cells possess little or no killer immunoglobulin-like receptors (KIRs), high interferon-gamma (IFN-gamma) production, but little cytotoxicity. CD56(dim) NK cells have high KIR expression, produce little IFN-gamma, yet display high cytotoxicity. We hypothesized that, if human NK maturation progresses from a CD56(bright) to a CD56(dim) phenotype, an intermediary NK cell must exist, which demonstrates more functional overlap than these 2 subsets, and we used CD94 expression to test our hypothesis. CD94(high)CD56(dim) NK cells express CD62L, CD2, and KIR at levels between CD56(bright) and CD94(low)CD56(dim) NK cells. CD94(high)CD56(dim) NK cells produce less monokine-induced IFN-gamma than CD56(bright) NK cells but much more than CD94(low)CD56(dim) NK cells because of differential interleukin-12-mediated STAT4 phosphorylation. CD94(high)CD56(dim) NK cells possess a higher level of granzyme B and perforin expression and CD94-mediated redirected killing than CD56(bright) NK cells but lower than CD94(low)CD56(dim) NK cells. Collectively, our data suggest that the density of CD94 surface expression on CD56(dim) NK cells identifies a functional and likely developmental intermediary between CD56(bright) and CD94(low)CD56(dim) NK cells. This supports the notion that, in vivo, human CD56(bright) NK cells progress through a continuum of differentiation that ends with a CD94(low)CD56(dim) phenotype.


Assuntos
Antígeno CD56/imunologia , Diferenciação Celular/imunologia , Regulação da Expressão Gênica/imunologia , Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos/imunologia , Subfamília D de Receptores Semelhantes a Lectina de Células NK/imunologia , Células Cultivadas , Humanos , Interferon gama/imunologia , Interleucina-12/imunologia , Células Matadoras Naturais/citologia , Selectina L/imunologia , Subpopulações de Linfócitos/citologia , Fosforilação/imunologia , Fator de Transcrição STAT4/imunologia
14.
J Immunol ; 184(6): 2769-75, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20142363

RESUMO

IL-15 is required for NK cell development and homeostasis in vivo. Because IL-15 is presented in trans via its high-affinity IL-15Ralpha-chain to cells expressing the IL-15Rbetagamma complex, we postulated that certain IL-15-bearing cells must be required for NK cell homeostasis. Using IL-15(WT/WT) and IL-15(-/-) mice, bone marrow chimeras with normal cellularity, and a selective depletion of CD11c(hi) dendritic cells (DCs), we demonstrate that ablation of the resting CD11c(hi) DC population results in a highly significant decrease in the absolute number of mature NK cells. In contrast, administration of Flt3 ligand increases the CD11c(hi) DC population, which, when expressing IL-15, significantly expands mature NK cells via enhanced survival and proliferation. In summary, a CD11c(hi) DC population expressing IL-15 is required to maintain NK cell homeostasis under conditions of normal cellularity and also is required to mediate Flt3 ligand-induced NK cell expansion in vivo.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Homeostase/imunologia , Células Matadoras Naturais/citologia , Proteínas de Membrana/fisiologia , Animais , Antígeno CD11c/biossíntese , Diferenciação Celular/imunologia , Proliferação de Células , Sobrevivência Celular/imunologia , Feminino , Humanos , Interleucina-15/deficiência , Interleucina-15/genética , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Ligantes , Proteínas de Membrana/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Recombinantes/administração & dosagem
15.
Blood ; 113(17): 4008-10, 2009 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-19244159

RESUMO

Considerable functional heterogeneity within human natural killer (NK) cells has been revealed through the characterization of distinct NK-cell subsets. Accordingly, a small subset of CD56(+)NKp44(+)NK cells, termed NK-22 cells, was recently described within secondary lymphoid tissue (SLT) as IL-22(-) when resting, with a minor fraction of this population becoming IL-22(+) when activated. Here we discover that the vast majority of stage 3 immature NK (iNK) cells in SLT constitutively and selectively express IL-22, a T(H)17 cytokine important for mucosal immunity, whereas earlier and later stages of NK developmental intermediates do not express IL-22. These iNK cells have a surface phenotype of CD34(-)CD117(+)CD161(+)CD94(-), largely lack expression of NKp44 and CD56, and do not produce IFN-gamma or possess cytolytic activity. In summary, stage 3 iNK cells are highly enriched for IL-22 and IL-26 messenger RNA, and IL-22 protein production, but do not express IL-17A or IL-17F.


Assuntos
Interleucina-17/imunologia , Interleucinas/imunologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Tecido Linfoide/imunologia , Linfopoese/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Células Matadoras Naturais/metabolismo , Tecido Linfoide/citologia , Tecido Linfoide/metabolismo , Fenótipo , RNA Mensageiro/genética , Interleucina 22
16.
Blood ; 113(22): 5558-67, 2009 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-19329776

RESUMO

Aberrant methylation of tumor suppressor genes can lead to their silencing in many cancers. TSC-22 is a gene silenced in several solid tumors, but its function and the mechanism(s) responsible for its silencing are largely unknown. Here we demonstrate that the TSC-22 promoter is methylated in primary mouse T or natural killer (NK) large granular lymphocyte (LGL) leukemia and this is associated with down-regulation or silencing of TSC-22 expression. The TSC-22 deregulation was reversed in vivo by a 5-aza-2'-deoxycytidine therapy of T or NK LGL leukemia, which significantly increased survival of the mice bearing this disease. Ectopic expression of TSC-22 in mouse leukemia or lymphoma cell lines resulted in delayed in vivo tumor formation. Targeted disruption of TSC-22 in wild-type mice enhanced proliferation and in vivo repopulation efficiency of hematopoietic precursor cells (HPCs). Collectively, our data suggest that TSC-22 normally contributes to the regulation of HPC function and is a putative tumor suppressor gene that is hypermethylated and silenced in T or NK LGL leukemia.


Assuntos
Movimento Celular/genética , Proliferação de Células , Células-Tronco Hematopoéticas/fisiologia , Leucemia Linfocítica Granular Grande/genética , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Animais , Células Cultivadas , Metilação de DNA , Epigênese Genética/fisiologia , Regulação Leucêmica da Expressão Gênica , Inativação Gênica/fisiologia , Genes Supressores de Tumor , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas
17.
J Immunol ; 183(8): 4968-74, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19801519

RESUMO

Understanding of heterogeneous NK subsets is important for the study of NK cell biology and development, and for the application of NK cell-based therapies in the treatment of disease. Here we demonstrate that the surface expression of CD94 can distinctively divide mouse NK cells into two approximately even CD94(low) and CD94(high) subsets in all tested organs and tissues. The CD94(high) NK subset has significantly greater capacity to proliferate, produce IFN-gamma, and lyse target cells than does the CD94(low) subset. The CD94(high) subset has exclusive expression of NKG2A/C/E, higher expression of CD117 and CD69, and lower expression of Ly49D (activating) and Ly49G2 (inhibitory). In vivo, purified mouse CD94(low) NK cells become CD94(high) NK cells, but not vice versa. Collectively, our data suggest that CD94 is an Ag that can be used to identify functionally distinct NK cell subsets in mice and could also be relevant to late-stage mouse NK cell development.


Assuntos
Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos/imunologia , Subfamília D de Receptores Semelhantes a Lectina de Células NK/imunologia , Transferência Adotiva , Animais , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Feminino , Interferon gama/biossíntese , Interferon gama/imunologia , Lectinas Tipo C , Camundongos , Camundongos Endogâmicos C57BL , Subfamília A de Receptores Semelhantes a Lectina de Células NK/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Proteínas Proto-Oncogênicas c-kit/imunologia
18.
J Immunol ; 181(6): 3784-92, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768831

RESUMO

TGF-beta can be a potent suppressor of lymphocyte effector cell functions and can mediate these effects via distinct molecular pathways. The role of TGF-beta in regulating CD16-mediated NK cell IFN-gamma production and antibody-dependent cellular cytotoxicity (ADCC) is unclear, as are the signaling pathways that may be utilized. Treatment of primary human NK cells with TGF-beta inhibited IFN-gamma production induced by CD16 activation with or without IL-12 or IL-2, and it did so without affecting the phosphorylation/activation of MAP kinases ERK and p38, as well as STAT4. TGF-beta treatment induced SMAD3 phosphorylation, and ectopic overexpression of SMAD3 resulted in a significant decrease in IFN-gamma gene expression following CD16 activation with or without IL-12 or IL-2. Likewise, NK cells obtained from smad3(-/-) mice produced more IFN-gamma in response to CD16 activation plus IL-12 when compared with NK cells obtained from wild-type mice. Coactivation of human NK cells via CD16 and IL-12 induced expression of T-BET, the positive regulator of IFN-gamma, and T-BET was suppressed by TGF-beta and by SMAD3 overexpression. An extended treatment of primary NK cells with TGF-beta was required to inhibit ADCC, and it did so by inhibiting granzyme A and granzyme B expression. This effect was accentuated in cells overexpressing SMAD3. Collectively, our results indicate that TGF-beta inhibits CD16-mediated human NK cell IFN-gamma production and ADCC, and these effects are mediated via SMAD3.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/imunologia , Tolerância Imunológica , Interferon gama/antagonistas & inibidores , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Receptores de IgG/fisiologia , Proteína Smad3/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Proteínas Ligadas por GPI , Regulação da Expressão Gênica/imunologia , Humanos , Tolerância Imunológica/genética , Interferon gama/biossíntese , Interleucina-12/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de IgG/antagonistas & inibidores , Proteína Smad3/biossíntese , Proteína Smad3/deficiência , Proteína Smad3/genética
19.
J Clin Invest ; 126(12): 4404-4416, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27775550

RESUMO

Natural killer (NK) cells can have potent antileukemic activity following haplo-mismatched, T cell-depleted stem cell transplantations for the treatment of acute myeloid leukemia (AML), but they are not successful in eradicating de novo AML. Here, we have used a mouse model of de novo AML to elucidate the mechanisms by which AML evades NK cell surveillance. NK cells in leukemic mice displayed a marked reduction in the cytolytic granules perforin and granzyme B. Further, as AML progressed, we noted the selective loss of an immature subset of NK cells in leukemic mice and in AML patients. This absence was not due to elimination by cell death or selective reduction in proliferation, but rather to the result of a block in NK cell differentiation. Indeed, NK cells from leukemic mice and humans with AML showed lower levels of TBET and EOMES, transcription factors that are critical for terminal NK cell differentiation. Further, the microRNA miR-29b, a regulator of T-bet and EOMES, was elevated in leukemic NK cells. Finally, deletion of miR-29b in NK cells reversed the depletion of this NK cell subset in leukemic mice. These results indicate that leukemic evasion of NK cell surveillance occurs through miR-mediated dysregulation of lymphocyte development, representing an additional mechanism of immune escape in cancer.


Assuntos
Imunidade Inata , Células Matadoras Naturais/imunologia , Leucemia Mieloide Aguda/imunologia , MicroRNAs/imunologia , RNA Neoplásico/imunologia , Evasão Tumoral , Animais , Linhagem Celular Tumoral , Granzimas/genética , Granzimas/imunologia , Humanos , Células Matadoras Naturais/patologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Transgênicos , MicroRNAs/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Perforina/genética , Perforina/imunologia , RNA Neoplásico/genética , Proteínas com Domínio T/genética , Proteínas com Domínio T/imunologia
20.
J Immunol Methods ; 296(1-2): 115-23, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15680156

RESUMO

Molecular characterization of human natural killer (NK) cells will require targeted gene delivery to inhibit and activate specific signaling pathways, yet to our knowledge, an effective means to deliver such products for long-term gene expression without disrupting normal cellular processes has not been described. In this study, we have developed a retroviral strategy to effectively express gene products in the NK cell, whereby its effector functions of cytotoxicity and cytokine production remain intact. Using an EBV/retroviral hybrid vector, we demonstrate infection of human peripheral blood NK cells with simultaneous expression of a marker for infection--the enhanced green fluorescent protein (EGFP)--along with various genes of interest. This technique results in successful infection of the CD56dim NK population that predominates among human peripheral blood NK and is the effector of antibody-dependent cellular cytotoxicity and natural killing. In addition, we demonstrate infection of the CD56bright NK subset as well as the NK-92 cell line. In summary, we have devised an efficient and reproducible methodology for the targeted delivery of gene products to human NK cells that should now provide opportunities to dissect the molecular processes critical to normal NK cell physiology.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos/genética , Herpesvirus Humano 4/genética , Células Matadoras Naturais/metabolismo , Retroviridae/genética , Antígeno CD56/imunologia , Células Cultivadas , Expressão Gênica , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Humanos , Interferon gama/análise , Interferon gama/metabolismo , Células Matadoras Naturais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA