Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Nat Immunol ; 21(10): 1205-1218, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32839608

RESUMO

Immune-modulating therapies have revolutionized the treatment of chronic diseases, particularly cancer. However, their success is restricted and there is a need to identify new therapeutic targets. Here, we show that natural killer cell granule protein 7 (NKG7) is a regulator of lymphocyte granule exocytosis and downstream inflammation in a broad range of diseases. NKG7 expressed by CD4+ and CD8+ T cells played key roles in promoting inflammation during visceral leishmaniasis and malaria-two important parasitic diseases. Additionally, NKG7 expressed by natural killer cells was critical for controlling cancer initiation, growth and metastasis. NKG7 function in natural killer and CD8+ T cells was linked with their ability to regulate the translocation of CD107a to the cell surface and kill cellular targets, while NKG7 also had a major impact on CD4+ T cell activation following infection. Thus, we report a novel therapeutic target expressed on a range of immune cells with functions in different immune responses.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Inflamação/imunologia , Células Matadoras Naturais/imunologia , Leishmania donovani/fisiologia , Leishmaniose Visceral/imunologia , Malária/imunologia , Proteínas de Membrana/metabolismo , Plasmodium/fisiologia , Animais , Células Cultivadas , Citotoxicidade Imunológica , Modelos Animais de Doenças , Exocitose , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Vesículas Secretórias/metabolismo
4.
Nat Immunol ; 18(9): 1004-1015, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28759001

RESUMO

Avoiding destruction by immune cells is a hallmark of cancer, yet how tumors ultimately evade control by natural killer (NK) cells remains incompletely defined. Using global transcriptomic and flow-cytometry analyses and genetically engineered mouse models, we identified the cytokine-TGF-ß-signaling-dependent conversion of NK cells (CD49a-CD49b+Eomes+) into intermediate type 1 innate lymphoid cell (intILC1) (CD49a+CD49b+Eomes+) populations and ILC1 (CD49a+CD49b-Eomesint) populations in the tumor microenvironment. Strikingly, intILC1s and ILC1s were unable to control local tumor growth and metastasis, whereas NK cells favored tumor immunosurveillance. Experiments with an antibody that neutralizes the cytokine TNF suggested that escape from the innate immune system was partially mediated by TNF-producing ILC1s. Our findings provide new insight into the plasticity of group 1 ILCs in the tumor microenvironment and suggest that the TGF-ß-driven conversion of NK cells into ILC1s is a previously unknown mechanism by which tumors escape surveillance by the innate immune system.


Assuntos
Reprogramação Celular/imunologia , Fibrossarcoma/imunologia , Neoplasias Gastrointestinais/imunologia , Tumores do Estroma Gastrointestinal/imunologia , Imunidade Inata/imunologia , Células Matadoras Naturais/imunologia , Neoplasias Experimentais/imunologia , Evasão Tumoral/imunologia , Animais , Estudos de Casos e Controles , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Células Matadoras Naturais/citologia , Linfócitos/citologia , Linfócitos/imunologia , Camundongos , Análise de Sequência de RNA , Transdução de Sinais , Fator de Crescimento Transformador beta/imunologia
5.
Immunity ; 52(6): 895-897, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32553176

RESUMO

The regulatory mechanisms controlling natural killer (NK) cells in the tumor microenvironment remain unknown. In this issue, Ni. et al. provide evidence that the transcription factor HIF-1α acts as a key negative regulator of NK cell metabolic fitness in the tumor microenvironment that critically impedes NK cell anti-tumor immune responses.


Assuntos
Neoplasias , Fatores de Transcrição , Humanos , Células Matadoras Naturais , Análise de Sequência de RNA , Microambiente Tumoral
6.
Immunity ; 53(4): 805-823.e15, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-33053330

RESUMO

The activating receptor CD226 is expressed on lymphocytes, monocytes, and platelets and promotes anti-tumor immunity in pre-clinical models. Here, we examined the role of CD226 in the function of tumor-infiltrating lymphocytes (TILs) and resistance to immunotherapy. In murine tumors, a large proportion of CD8+ TILs had decreased surface expression of CD226 and exhibited features of dysfunction, whereas CD226hi TILs were highly functional. This correlation was seen also in TILs isolated from HNSCC patients. Mutation of CD226 at tyrosine 319 (Y319) led to increased CD226 surface expression, enhanced anti-tumor immunity and improved efficacy of immune checkpoint blockade (ICB). Mechanistically, tumor-derived CD155, the ligand for CD226, initiated phosphorylation of Y319 by Src kinases, thereby enabling ubiquitination of CD226 by CBL-B, internalization, and proteasomal degradation. In pre-treatment samples from melanoma patients, CD226+CD8+ T cells correlated with improved progression-free survival following ICB. Our findings argue for the development of therapies aimed at maintaining the expression of CD226.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Linfócitos T CD8-Positivos/imunologia , Receptores Virais/imunologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Humanos , Inibidores de Checkpoint Imunológico/imunologia , Imunoterapia/métodos , Células Jurkat , Linfócitos do Interstício Tumoral/imunologia , Masculino , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos C57BL
7.
Immunity ; 47(4): 789-802.e9, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-29045907

RESUMO

Inhibitors of the receptor tyrosine kinase c-MET are currently used in the clinic to target oncogenic signaling in tumor cells. We found that concomitant c-MET inhibition promoted adoptive T cell transfer and checkpoint immunotherapies in murine cancer models by increasing effector T cell infiltration in tumors. This therapeutic effect was independent of tumor cell-intrinsic c-MET dependence. Mechanistically, c-MET inhibition impaired the reactive mobilization and recruitment of neutrophils into tumors and draining lymph nodes in response to cytotoxic immunotherapies. In the absence of c-MET inhibition, neutrophils recruited to T cell-inflamed microenvironments rapidly acquired immunosuppressive properties, restraining T cell expansion and effector functions. In cancer patients, high serum levels of the c-MET ligand HGF correlated with increasing neutrophil counts and poor responses to checkpoint blockade therapies. Our findings reveal a role for the HGF/c-MET pathway in neutrophil recruitment and function and suggest that c-MET inhibitor co-treatment may improve responses to cancer immunotherapy in settings beyond c-MET-dependent tumors.


Assuntos
Imunoterapia/métodos , Neoplasias Experimentais/terapia , Neutrófilos/imunologia , Proteínas Proto-Oncogênicas c-met/imunologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Estimativa de Kaplan-Meier , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Neutrófilos/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
8.
Immunol Cell Biol ; 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38873699

RESUMO

Antibiotic resistance is a major public health threat, and alternatives to antibiotic therapy are urgently needed. Immunotherapy, particularly the blockade of inhibitory immune checkpoints, is a leading treatment option in cancer and autoimmunity. In this study, we used a murine model of Salmonella Typhimurium infection to investigate whether immune checkpoint blockade could be applied to bacterial infection. We found that the immune checkpoint T-cell immunoglobulin and ITIM domain (TIGIT) was significantly upregulated on lymphocytes during infection, particularly on CD4+ T cells, drastically limiting their proinflammatory function. Blockade of TIGIT in vivo using monoclonal antibodies was able to enhance immunity and improve bacterial clearance. The efficacy of anti-TIGIT was dependent on the capacity of the antibody to bind to Fc (fragment crystallizable) receptors, giving important insights into the mechanism of anti-TIGIT therapy. This research suggests that targeting immune checkpoints, such as TIGIT, has the potential to enhance immune responses toward bacteria and restore antibacterial treatment options in the face of antibiotic resistance.

9.
Haematologica ; 109(3): 787-798, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-37767564

RESUMO

T-cell-engaging bispecific antibodies (T-BsAb) have produced impressive clinical responses in patients with relapsed/refractory B-cell malignancies, although treatment failure remains a major clinical challenge. Growing evidence suggests that a complex interplay between immune cells and tumor cells is implicated in the mechanism of action and therefore, understanding immune regulatory mechanisms might provide a clue for how to improve the efficacy of T-BsAb therapy. Here, we investigated the functional impact of regulatory T (Treg) cells on anti-tumor immunity elicited by T-BsAb therapy. In a preclinical model of myeloma, the activation and expansion of Treg cells in the bone marrow were observed in response to anti-B-cell maturation antigen (BCMA) T-BsAb therapy. T-BsAb triggered the generation of induced Treg cells from human conventional CD4 cells after co-culture with tumor cells. Moreover, T-BsAb directly activated freshly isolated circulating Treg cells, leading to the production of interleukin-10 and inhibition of T-BsAb-mediated CD8 T-cell responses. The activation of Treg cells was also seen in bone marrow samples from myeloma patients after ex vivo treatment with T-BsAb, further supporting that T-BsAb have an impact on Treg homeostasis. Importantly, transient ablation of Treg cells in combination with T-BsAb therapy dramatically improved effector lymphocyte activities and disease control in the preclinical myeloma model, leading to prolonged survival. Together, this information suggests that therapy-induced activation of Treg cells critically regulates anti-tumor immunity elicited by T-BsAb therapy, with important implications for improving the efficacy of such treatment.


Assuntos
Anticorpos Biespecíficos , Mieloma Múltiplo , Humanos , Linfócitos T Reguladores , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos
10.
Haematologica ; 109(7): 2131-2143, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38268493

RESUMO

T-cell-engaging bispecific antibody (T-BsAb, also known as BiTE) therapy has emerged as a powerful therapeutic modality against multiple myeloma. Given that T-BsAb therapy redirects endogenous T cells to eliminate tumor cells, reinvigorating dysfunctional T cells may be a potential approach to improve the efficacy of T-BsAb. While various immunostimulatory cytokines can potentiate effector T-cell functions, the optimal cytokine treatment for T-BsAb therapy is yet to be established, partly due to a concern of cytokine release syndrome driven by aberrant interferon (IFN)-γ production. Here, we functionally screen immunostimulatory cytokines to determine an ideal combination partner for T-BsAb therapy. This approach reveals interleukin (IL)-21 as a potential immunostimulatory cytokine with the ability to augment T-BsAb-mediated release of granzyme B and perforin, without increasing IFN-γ release. Transcriptome profiling and functional characterization strongly support that IL-21 selectively targets the cytotoxic granule exocytosis pathway, but not pro-inflammatory responses. Notably, IL-21 modulates multiple steps of cytotoxic effector functions including upregulation of co-activating CD226 receptor, increasing cytotoxic granules, and promoting cytotoxic granule delivery at the immunological synapse. Indeed, T-BsAb-mediated myeloma killing is cytotoxic granule-dependent, and IL-21 priming significantly augments cytotoxic activities. Furthermore, in vivo IL-21 treatment induces cytotoxic effector reprogramming in bone marrow T cells, showing synergistic anti-myeloma effects in combination with T-BsAb therapy. Together, harnessing the cytotoxic granule exocytosis pathway by IL-21 may be a potential approach to achieve better responses by T-BsAb therapy.


Assuntos
Anticorpos Biespecíficos , Exocitose , Mieloma Múltiplo , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Humanos , Camundongos , Animais , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/terapia , Mieloma Múltiplo/patologia , Citotoxicidade Imunológica , Interleucinas/metabolismo , Linhagem Celular Tumoral , Citocinas/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Granzimas/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/efeitos dos fármacos
11.
Blood ; 136(24): 2731-2740, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-32645135

RESUMO

Avoiding immune destruction is a hallmark of cancer. Over the past few years, significant advances have been made in understanding immune dysfunction and immunosuppression in multiple myeloma (MM), and various immunotherapeutic approaches have delivered improved clinical responses. However, it is still challenging to completely eliminate malignant plasma cells (PCs) and achieve complete cure. The interplay between the immune system and malignant PCs is implicated throughout all stages of PC dyscrasias, including asymptomatic states called monoclonal gammopathy of undetermined significance and smoldering myeloma. Although the immune system effectively eliminates malignant PCs, or at least induces functional dormancy at early stages, malignant PCs eventually evade immune elimination, leading to progression to active MM, in which dysfunctional effector lymphocytes, tumor-educated immunosuppressive cells, and soluble mediators coordinately act as a barrier for antimyeloma immunity. An in-depth understanding of this dynamic process, called cancer immunoediting, will provide important insights into the immunopathology of PC dyscrasias and MM immunotherapy. Moreover, a growing body of evidence suggests that, together with nonhematopoietic stromal cells, bone marrow (BM) immune cells with unique functions support the survival of normal and malignant PCs in the BM niche, highlighting the diverse roles of immune cells beyond antimyeloma immunity. Together, the immune system critically acts as a rheostat that fine-tunes the balance between dormancy and disease progression in PC dyscrasias.


Assuntos
Imunoterapia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/terapia , Plasmócitos/imunologia , Humanos , Mieloma Múltiplo/patologia , Plasmócitos/patologia
12.
Proteomics ; 21(19): e2100152, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34390184

RESUMO

Mass spectrometry-based proteomics revolutionized global proteomic profiling. Although high molecular weight abundant proteins are readily sampled in global proteomics studies, less abundant low molecular weight proteins are often underrepresented. This includes biologically important classes of low molecular weight proteins including ligands, growth factors, peptide hormones and cytokines. Although extensive fractionation can facilitate achieving better coverage of proteome, it requires additional infrastructure, mass spectrometry time and labour. There is need for a simple method that can selectively deplete high molecular weight abundant proteins and enrich for low molecular weight less abundant proteins to improve their coverage in proteomics studies. We present a simple organic-solvent based protein precipitation method that selectively depletes high molecular weight proteins and enriches low molecular weight proteins in the soluble fraction. Using this strategy, we demonstrate identification of low molecular weight proteins that are generally underrepresented in proteomics datasets. In addition, we show the utility of this approach in identifying functional cleavage products from precursor proteins and low molecular weight short open reading frame proteins encoded by non-coding regions such as lncRNAs and UTRs. As the method does not require additional infrastructure, it can complement existing proteomics workflows to increase detection and coverage of low molecular weight proteins that are less abundant.


Assuntos
Peptídeos , Proteômica , Peso Molecular , Proteoma , Solventes
13.
Immunol Cell Biol ; 99(1): 65-83, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32748462

RESUMO

Type 2 innate lymphoid cells (ILC2s) are important producers of type 2 cytokines whose role in hematological cancers remains unclear. ILC2s are a heterogeneous population encompassing distinct subsets with different tissue localization and cytokine responsiveness. In this study, we investigated the role of bone marrow (BM) ILC2s and interleukin (IL)-33-stimulated ILC2s in multiple myeloma, a plasma cell malignancy that develops in the BM. We found that myeloma growth was associated with phenotypic and functional alterations of BM ILC2s, characterized by an increased expression of maturation markers and reduced cytokine response to IL-2/IL-33. We identified a population of KLRG1hi ILC2s that preferentially accumulated in the liver and spleen of Il2rg-/- Rag2-/- mice reconstituted with BM ILC2s. A similar population of KLRG1hi ILC2s was observed in the blood, liver and spleen of IL-33-treated wild-type mice. The presence of KLRG1hi ILC2s in ILC2-reconstituted Il2rg-/- Rag2-/- mice or in IL-33-treated wild-type mice was associated with increased eosinophil numbers but had no effect on myeloma progression. Interestingly, while decreased myeloma growth was observed following treatment of Rag-deficient mice with the type 1 cytokines IL-12 and IL-18, this protection was reversed when mice received a combined treatment of IL-33 together with IL-12 and IL-18. In summary, our data indicate that IL-33 treatment induces a population of circulating inflammatory KLRG1hi ILC2s and inhibits type 1 immunity against multiple myeloma. These results argue against therapeutic administration of IL-33 to myeloma patients.


Assuntos
Imunidade Inata , Mieloma Múltiplo , Animais , Citocinas , Humanos , Interleucina-33 , Lectinas Tipo C , Linfócitos , Camundongos , Mieloma Múltiplo/tratamento farmacológico , Receptores Imunológicos
14.
Rinsho Ketsueki ; 62(8): 1186-1194, 2021.
Artigo em Japonês | MEDLINE | ID: mdl-34497206

RESUMO

The cancer immunoediting theory states that selective pressure by the immune system can sculpt tumor phenotypes and genotypes during all stages of tumor development. Multiple myeloma development has been recognized as a multistage process that includes premalignant expansion of plasma cells, symptomatic progression, and recurrent relapses. We discuss the dynamic crosstalk between the immune system and malignant plasma cells in light of the cancer immunoediting theory. Myeloma progression triggers dramatic alterations in the bone marrow, including bone destruction, inflammation, and angiogenesis, thereby generating an immunosuppressive milieu. We also present an overview of the immune microenvironment in multiple myeloma.


Assuntos
Mieloma Múltiplo , Medula Óssea , Humanos , Recidiva Local de Neoplasia , Plasmócitos , Microambiente Tumoral
15.
Immunol Cell Biol ; 98(6): 434-436, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32535941

RESUMO

A recent paper by Zhou et al. describes a new engineered decoy-resistant interleukin-18 with potent antitumor activity and translational potential for cancer immunotherapy.


Assuntos
Interleucina-18 , Neoplasias , Humanos , Fatores Imunológicos , Imunoterapia , Neoplasias/terapia
16.
Blood ; 132(16): 1689-1694, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-29986909

RESUMO

Immune-based therapies hold promise for the treatment of multiple myeloma (MM), but so far, immune checkpoint blockade targeting programmed cell death protein 1 has not proven effective as single agent in this disease. T-cell immunoglobulin and ITIM domains (TIGIT) is another immune checkpoint receptor known to negatively regulate T-cell functions. In this study, we investigated the therapeutic potential of TIGIT blockade to unleash immune responses against MM. We observed that, in both mice and humans, MM progression was associated with high levels of TIGIT expression on CD8+ T cells. TIGIT+ CD8+ T cells from MM patients exhibited a dysfunctional phenotype characterized by decreased proliferation and inability to produce cytokines in response to anti-CD3/CD28/CD2 or myeloma antigen stimulation. Moreover, when challenged with Vk*MYC mouse MM cells, TIGIT-deficient mice showed decreased serum monoclonal immunoglobulin protein levels associated with reduced tumor burden and prolonged survival, indicating that TIGIT limits antimyeloma immune responses. Importantly, blocking TIGIT using monoclonal antibodies increased the effector function of MM patient CD8+ T cells and suppressed MM development. Altogether our data provide evidence for an immune-inhibitory role of TIGIT in MM and support the development of TIGIT-blocking strategies for the treatment of MM patients.


Assuntos
Anticorpos Monoclonais/farmacologia , Linfócitos T CD8-Positivos/imunologia , Mieloma Múltiplo/prevenção & controle , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptores Imunológicos/antagonistas & inibidores , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Células Cultivadas , Humanos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mieloma Múltiplo/etiologia , Mieloma Múltiplo/patologia , Receptor de Morte Celular Programada 1/imunologia , Receptores Imunológicos/metabolismo , Receptores Imunológicos/fisiologia
17.
Immunol Cell Biol ; 95(4): 325-332, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27999432

RESUMO

Recent advances in cancer immunotherapy, particularly immune checkpoint blockade therapy have dramatically changed the therapeutic strategy against advanced malignancies. Still, only a subset of patients shows a good response to any single therapy. Moreover, it remains largely unsolved how we can maintain durable clinical responses, or how we can successfully treat a broader range of cancers by immunotherapy. Growing evidence suggests that the major barrier to more successful cancer immunotherapy is the tumour microenvironment (TME), where chronic inflammation has a predominant role in tumour survival and proliferation, angiogenesis and immunosuppression. Over the past decades, our understanding of cancer-related inflammation has significantly evolved, and now we have various therapeutic options tailored to the TME. These therapeutic strategies include inhibiting inflammatory mediators or their downstream signalling molecules, blocking the recruitment of myeloid cells, modulating immunosuppressive functions in myeloid cells and re-educating the TME. In this review, we discuss the role of cancer-related inflammation as a potential target in the era of immunotherapy.


Assuntos
Imunoterapia/métodos , Inflamação/terapia , Células Mieloides/imunologia , Neoplasias/terapia , Microambiente Tumoral , Animais , Humanos , Terapia de Imunossupressão , Inflamação/imunologia , Neoplasias/imunologia , Transdução de Sinais , Evasão Tumoral
18.
Cell Mol Life Sci ; 73(8): 1569-89, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26801219

RESUMO

Multiple myeloma (MM) is a tumor of terminally differentiated B cells that arises in the bone marrow. Immune interactions appear as key determinants of MM progression. While myeloid cells foster myeloma-promoting inflammation, Natural Killer cells and T lymphocytes mediate protective anti-myeloma responses. The profound immune deregulation occurring in MM patients may be involved in the transition from a premalignant to a malignant stage of the disease. In the last decades, the advent of stem cell transplantation and new therapeutic agents including proteasome inhibitors and immunoregulatory drugs has dramatically improved patient outcomes, suggesting potentially key roles for innate and adaptive immunity in disease control. Nevertheless, MM remains largely incurable for the vast majority of patients. A better understanding of the complex interplay between myeloma cells and their immune environment should pave the way for designing better immunotherapies with the potential of very long term disease control. Here, we review the immunological microenvironment in myeloma. We discuss the role of naturally arising anti-myeloma immune responses and their potential corruption in MM patients. Finally, we detail the numerous promising immune-targeting strategies approved or in clinical trials for the treatment of MM.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/terapia , Medula Óssea/imunologia , Células da Medula Óssea/imunologia , Humanos , Vigilância Imunológica/imunologia , Evasão Tumoral/imunologia , Microambiente Tumoral/imunologia
19.
Proc Natl Acad Sci U S A ; 110(23): 9421-6, 2013 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-23690625

RESUMO

The natural killer group 2 membrane D (NKG2D) activating receptor plays crucial roles not only in host defense against tumors and viral infections, but also in autoimmune diseases. After NKG2D-mediated activation, Natural killer (NK) cells must be regulated to avoid potentially harmful reactivity. However, the negative regulation of these activated NK cells is poorly understood. Here, we reveal that the engagement of NKG2D by its ligand elicits not only target cell lysis, but also NK cell fratricide. Conventional mouse NK cells underwent cell death when cocultured with RMA cells expressing the NKG2D ligand retinoic acid early-inducible protein 1 (Rae-1), but not with RMA cells lacking MHC class I. NK cells from mice deficient for DAP10 and DAP12 or perforin did not undergo death, highlighting the importance of the NKG2D pathway for NK cell death. However, NKG2D does not transmit direct death signals in NK cells. Rather, the interaction between NKG2D and Rae-1 allowed NK cells to acquire tumor-derived Rae-1 by a membrane transfer process known as "trogocytosis," which was associated with clathrin-dependent NKG2D endocytosis. NK cells dressed with Rae-1 were lysed by neighboring NK cells through the NKG2D-induced perforin pathway in vitro and in vivo. These results provide the unique NKG2D function in negative regulation of activated NK cells.


Assuntos
Morte Celular/imunologia , Células Matadoras Naturais/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Transdução de Sinais/imunologia , Animais , Linhagem Celular Tumoral , Primers do DNA/genética , Endocitose/fisiologia , Citometria de Fluxo , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Perforina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Int Immunol ; 26(11): 597-606, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24860119

RESUMO

The natural killer group 2 membrane D (NKG2D) receptor is an NK-activating receptor that plays an important role in host defense against tumors and viral infections. Although the marmoset is an important and reliable animal model, especially for the study of human-specific viral infections, functional characterization of NKG2D on marmoset NK cells has not previously been conducted. In the present study, we investigated a subpopulation of marmoset NK cells that express NKG2D and exhibit cytolytic potential. On the basis of their CD16 and CD56 expression patterns, marmoset NK cells can be classified into three subpopulations: CD16(+) CD56(-), CD16(-) CD56(+) and CD16(-) CD56(-) cells. NKG2D expression on marmoset CD16(+) CD56(-) and CD16(-) CD56(+) splenocytes was confirmed using an NKG2D ligand composed of an MHC class I chain-related molecule A (MICA)-Fc fusion protein. When marmoset splenocytes were cultured with IL-2 for 4 days, NKG2D expression was retained on CD16(+) CD56(-) and CD16(-) CD56(+). In addition, CD16(+) CD56(+) cells within the marmoset NK population appeared which expressed NKG2D after IL-2 stimulation. IL-2-activated marmoset NK cells showed strong cytolytic activity against K562 target cells and target cells stably expressing MICA. Further, the cytolytic activity of marmoset splenocytes was significantly reduced after addition of MICA-Fc fusion protein. Thus, NKG2D functions as an activating receptor on marmoset NK cells that possesses cytotoxic potential, and phenotypic profiles of marmoset NK cell subpopulations are similar to those seen in humans.


Assuntos
Callithrix/imunologia , Callithrix/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Callithrix/genética , Linhagem Celular , Membrana Celular/metabolismo , Clonagem Molecular , Reações Cruzadas/imunologia , Antígenos de Histocompatibilidade Classe I/química , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/metabolismo , Dados de Sequência Molecular , Subfamília K de Receptores Semelhantes a Lectina de Células NK/antagonistas & inibidores , Subfamília K de Receptores Semelhantes a Lectina de Células NK/química , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Fenótipo , Proteínas Recombinantes de Fusão/farmacologia , Alinhamento de Sequência , Baço/citologia , Baço/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA