Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Front Oncol ; 14: 1371307, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38863623

RESUMO

Background: Triple-negative breast cancer (TNBC) cells are a highly formidable cancer to treat. Nonetheless, by continued investigation into the molecular biology underlying the complex regulation of TNBC cell activity, vulnerabilities can be exposed as potential therapeutic targets at the molecular level. We previously revealed that lysyl oxidase-like 4 (LOXL4) promotes the invasiveness of TNBC cells via cell surface annexin A2 as a novel binding substrate of LOXL4, which promotes the abundant localization of integrin-ß1 at the cancer plasma membrane. However, it has yet to be uncovered how the LOXL4-mediated abundance of integrin-ß1 hastens the invasive outgrowth of TNBC cells at the molecular level. Methods: LOXL4-overexpressing stable clones were established from MDA-MB-231 cells and subjected to molecular analyses, real-time qPCR and zymography to clarify their invasiveness, signal transduction, and matrix metalloprotease (MMP) activity, respectively. Results: Our results show that LOXL4 potently promotes the induction of matrix metalloprotease 9 (MMP9) via activation of nuclear factor-κB (NF-κB). Our molecular analysis revealed that TNF receptor-associated factor 4 (TRAF4) and TGF-ß activated kinase 1 (TAK1) were required for the activation of NF-κB through Iκß kinase kinase (IKKα/ß) phosphorylation. Conclusion: Our results demonstrate that the newly identified LOXL4-mediated axis, integrin-ß1-TRAF4-TAK1-IKKα/ß-Iκßα-NF-κB-MMP9, is crucial for TNBC cell invasiveness.

2.
Rinsho Ketsueki ; 65(5): 428-438, 2024.
Artigo em Japonês | MEDLINE | ID: mdl-38825523

RESUMO

Over the past decade, new therapeutic modalities have markedly improved clinical outcomes for patients with multiple myeloma. Recently, immunotherapy using both bispecific antibodies (BsAb) and chimeric antigen receptor T cells (CAR-T cells) has induced further anti-myeloma responses. Different agents must be combined to overcome the heterogeneity of myeloma cell clones, and new modalities for the treatment of refractory myeloma must also be developed to strengthen therapeutic effects. We have developed a novel BiTE (bispecific T-cell engager)-based modality, referred to as bridging-BiTE (B-BiTE). B-BiTE is able to bind to both an Fc domain of a human immunoglobulin G monoclonal antibody (mAb) and the human CD3 molecule. This enables rapid generation of a mAb/B-BiTE complex and safely induces dual-lymphoid activation of both human T cells and NK cells against myeloma cells. Importantly, sequential immunotherapy using two different mAb/B-BiTE complexes can produce deep and durable anti-myeloma responses. To further advance treatment of multiple myeloma, it is important to determine how to combine and sequence immunotherapy with other agents while considering management of unique adverse events caused by activated immune cells.


Assuntos
Anticorpos Biespecíficos , Imunoterapia , Mieloma Múltiplo , Anticorpos Biespecíficos/uso terapêutico , Mieloma Múltiplo/terapia , Mieloma Múltiplo/imunologia , Humanos , Imunoterapia/métodos , Linfócitos T/imunologia
3.
Int J Hematol ; 120(1): 23-33, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38613724

RESUMO

Despite the development of various therapeutic agents, multiple myeloma remains incurable. Recently, T-cell redirected immunotherapy has become a promising strategy for the treatment of refractory myeloma. Clinical trials using chimeric antigen receptor (CAR)-T cells and bispecific antibodies have demonstrated successful anti-myeloma responses in triple-class-refractory patients. However, unique and unwanted immune effects associated with on-target/off-target reactivity of activated immune cells need to be considered and properly managed. This review summarizes recent advances in bispecific antibodies for the treatment of refractory myeloma. It outlines the history of their development, along with a discussion of their mechanisms of action and their current and potential future role in myeloma therapy. As more evidence emerges to inform the timing of CAR-T-cell therapy, the results of clinical trials and off-the-shelf nature of bispecifics also suggest the timing of their treatment. These findings will promote further development and application of bispecifics for refractory myeloma in combination with other appropriate agents.


Assuntos
Anticorpos Biespecíficos , Imunoterapia Adotiva , Mieloma Múltiplo , Mieloma Múltiplo/terapia , Mieloma Múltiplo/imunologia , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Biespecíficos/imunologia , Humanos , Imunoterapia Adotiva/métodos , Receptores de Antígenos Quiméricos/imunologia , Ensaios Clínicos como Assunto , Linfócitos T/imunologia
4.
Front Oncol ; 14: 1371342, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38595825

RESUMO

Background: Our earlier research revealed that the secreted lysyl oxidase-like 4 (LOXL4) that is highly elevated in triple-negative breast cancer (TNBC) acts as a catalyst to lock annexin A2 on the cell membrane surface, which accelerates invasive outgrowth of the cancer through the binding of integrin-ß1 on the cell surface. However, whether this machinery is subject to the LOXL4-mediated intrusive regulation remains uncertain. Methods: Cell invasion was assessed using a transwell-based assay, protein-protein interactions by an immunoprecipitation-Western blotting technique and immunocytochemistry, and plasmin activity in the cell membrane by gelatin zymography. Results: We revealed that cell surface annexin A2 acts as a receptor of plasminogen via interaction with S100A10, a key cell surface annexin A2-binding factor, and S100A11. We found that the cell surface annexin A2/S100A11 complex leads to mature active plasmin from bound plasminogen, which actively stimulates gelatin digestion, followed by increased invasion. Conclusion: We have refined our understanding of the role of LOXL4 in TNBC cell invasion: namely, LOXL4 mediates the upregulation of annexin A2 at the cell surface, the upregulated annexin 2 binds S100A11 and S100A10, and the resulting annexin A2/S100A11 complex acts as a receptor of plasminogen, readily converting it into active-form plasmin and thereby enhancing invasion.

5.
Blood ; 142(21): 1789-1805, 2023 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-37738633

RESUMO

Immunotherapy using bispecific antibodies including bispecific T-cell engager (BiTE) has the potential to enhance the efficacy of treatment for relapsed/refractory multiple myeloma. However, myeloma may still recur after treatment because of downregulation of a target antigen and/or myeloma cell heterogeneity. To strengthen immunotherapy for myeloma while overcoming its characteristics, we have newly developed a BiTE-based modality, referred to as bridging-BiTE (B-BiTE). B-BiTE was able to bind to both a human immunoglobulin G-Fc domain and the CD3 molecule. Clinically available monoclonal antibodies (mAbs) were bound with B-BiTE before administration, and the mAb/B-BiTE complex induced antitumor T-cell responses successfully while preserving and supporting natural killer cell reactivity, resulting in enhanced antimyeloma effects via dual-lymphoid activation. In contrast, any unwanted off-target immune-cell reactivity mediated by mAb/B-BiTE complexes or B-BiTE itself appeared not to be observed in vitro and in vivo. Importantly, sequential immunotherapy using 2 different mAb/B-BiTE complexes appeared to circumvent myeloma cell antigen escape, and further augmented immune responses to myeloma relative to those induced by mAb/B-BiTE monotherapy or sequential therapy with 2 mAbs in the absence of B-BiTE. Therefore, this modality facilitates easy and prompt generation of a broad panel of bispecific antibodies that can induce deep and durable antitumor responses in the presence of clinically available mAbs, supporting further advancement of reinforced immunotherapy for multiple myeloma and other refractory hematologic malignancies.


Assuntos
Anticorpos Biespecíficos , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/terapia , Recidiva Local de Neoplasia , Imunoterapia/métodos , Anticorpos Monoclonais/uso terapêutico
6.
J Biochem ; 174(6): 533-548, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37725528

RESUMO

Sterile alpha and Toll/interleukin receptor motif-containing protein 1 (SARM1) is a NAD+ hydrolase that plays a key role in axonal degeneration and neuronal cell death. We reported that c-Jun N-terminal kinase (JNK) activates SARM1 through phosphorylation at Ser-548. The importance of SARM1 phosphorylation in the pathological process of Parkinson's disease (PD) has not been determined. We thus conducted the present study by using rotenone (an inducer of PD-like pathology) and neurons derived from induced pluripotent stem cells (iPSCs) from healthy donors and a patient with familial PD PARK2 (FPD2). The results showed that compared to the healthy neurons, FPD2 neurons were more vulnerable to rotenone-induced stress and had higher levels of SARM1 phosphorylation. Similar cellular events were obtained when we used PARK2-knockdown neurons derived from healthy donor iPSCs. These events in both types of PD-model neurons were suppressed in neurons treated with JNK inhibitors, Ca2+-signal inhibitors, or by a SARM1-knockdown procedure. The degenerative events were enhanced in neurons overexpressing wild-type SARM1 and conversely suppressed in neurons overexpressing the SARM1-S548A mutant. We also detected elevated SARM1 phosphorylation in the midbrain of PD-model mice. The results indicate that phosphorylated SARM1 plays an important role in the pathological process of rotenone-induced neurodegeneration.


Assuntos
Doença de Parkinson , Rotenona , Humanos , Animais , Camundongos , Rotenona/farmacologia , Rotenona/metabolismo , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Morte Celular , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/metabolismo
8.
Cell Signal ; 108: 110717, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37187216

RESUMO

Nicotinamide adenine dinucleotide (NAD)+-biosynthetic and consuming enzymes are involved in various intracellular events through the regulation of NAD+ metabolism. Recently, it has become clear that alterations in the expression of NAD+-biosynthetic and consuming enzymes contribute to the axonal stability of neurons. We explored soluble bioactive factor(s) that alter the expression of NAD+-metabolizing enzymes and found that cytokine interferon (IFN)-γ increased the expression of nicotinamide nucleotide adenylyltransferase 2 (NMNAT2), an NAD+-biosynthetic enzyme. IFN-γ activated signal transducers and activators of transcription 1 and 3 (STAT1/3) followed by c-Jun N-terminal kinase (JNK) suppression. As a result, STAT1/3 increased the expression of NMNAT2 at both mRNA and protein levels in a dose- and time-dependent manner and, at the same time, suppressed activation of sterile alpha and Toll/interleukin receptor motif-containing 1 (SARM1), an NAD+-consuming enzyme, and increased intracellular NAD+ levels. We examined the protective effect of STAT1/3 signaling against vincristine-mediated cell injury as a model of chemotherapy-induced peripheral neuropathy (CIPN), in which axonal degeneration is involved in disease progression. We found that IFN-γ-mediated STAT1/3 activation inhibited vincristine-induced downregulation of NMNAT2 and upregulation of SARM1 phosphorylation, resulting in modest suppression of subsequent neurite degradation and cell death. These results indicate that STAT1/3 signaling induces NMNAT2 expression while simultaneously suppressing SARM1 phosphorylation, and that both these actions contribute to suppression of axonal degeneration and cell death.


Assuntos
Axônios , NAD , NAD/metabolismo , Vincristina/metabolismo , Axônios/metabolismo , Neurônios/metabolismo , Morte Celular , Proteínas do Domínio Armadillo/metabolismo
9.
Rinsho Ketsueki ; 63(9): 1298-1309, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-36198556

RESUMO

Based on the progress of gene-modification technologies, bispecific antibodies that possess antigen-binding sites with two different specificities have been developed. After the success of blinatumomab for treating refractory B-cell leukemia, series of clinical trials using bispecific antibodies for relapsed and refractory hematological malignancies are being conducted. Several bispecific antibodies target an antigen expressed by tumor cells and the CD3 molecule where binding of bispecific antibodies can generate artificial immunological synapses between tumor cells and human T cells. Therefore, fine tuning of binding affinity and/or structural conformation concomitant with bispecific antibodies may be required to induce antitumor effects and regulate immune-related adverse events, such as cytokine release syndrome. In the future, combination therapy of conventional chemotherapy and/or allogeneic stem-cell transplantation with bispecific antibody therapy will be necessary. Furthermore, molecular target therapy with bispecific antibody therapy is expected to pave the way for next-generation target therapy, resulting in the development of a further effective and safe treatment strategy for hematological malignancies.


Assuntos
Anticorpos Biespecíficos , Neoplasias Hematológicas , Anticorpos Biespecíficos/uso terapêutico , Antígenos CD19 , Complexo CD3/metabolismo , Terapia Combinada , Neoplasias Hematológicas/tratamento farmacológico , Humanos
10.
J Pers Med ; 12(6)2022 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-35743673

RESUMO

Immune checkpoint inhibitor (ICI) therapy increases the risk of immune-related adverse events (irAEs). In particular, combination checkpoint blockade (CCB) targeting inhibitory CTLA-4 and PD-1 receptors could lead to irAEs at a higher rate than ICI monotherapy. Management of irAEs is important while using ICIs. However, there are no reliable biomarkers for predicting irAEs. The aim of this study was to elucidate early B cell changes after CCB therapy in patients with renal cell carcinoma (RCC) and verify whether B cells can be a predictor of irAEs. This prospective cohort study was conducted with 23 Japanese patients with metastatic RCC. An increase in the proportion of CD21lo B cells and CD21lo memory B cells was confirmed following CCB therapy. Although there were no differences in clinical outcomes between irAE and no-irAE groups, the proportion of CD21lo B cells at baseline was lower in the irAE group, with a significant increase after the first cycle of CCB therapy. Further analysis revealed a moderate correlation between irAEs and CD21lo B cell levels at baseline (area under the curve: 0.83, cut-off: 3.13%, sensitivity: 92.3, specificity: 70.0). This finding indicates that patients with low baseline CD21lo B cell levels warrant closer monitoring for irAEs. The clinical registration number by the Certified Review Board of Ehime University is No. 1902011.

12.
Commun Biol ; 4(1): 273, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33654176

RESUMO

Cancer immunotherapy using T cells redirected with chimeric antigen receptor (CAR) has shown a lot of promise. We have established a single-chain antibody (scFv) generation system in which scFv library-expressing CAR-T cells can be screened appropriately based on their antitumor functions. A variable region library containing the variable and J regions of the human immunoglobulin light or heavy chain was fused with the variable region of a heavy or light chain encoded by an existing tumor-specific antibody to generate a new scFv library. Then, scFv library-expressing CAR-T cells were generated and stimulated with target cells to concentrate the antigen-specific population. Using this system, target-specific recognition of CAR-T cells appeared to be finely tuned by selecting a new variable region. Importantly, we have demonstrated that the newly optimized scFv-expressing CAR-T cells had better proliferation capacity and durable phenotypes, enabling superior reactivity against advanced tumors in vivo in comparison with the original CAR-T cells. Therefore, the optimization of an scFv is needed to maximize the in vivo antitumor functions of CAR-T cells. This system may allow us to adjust an immunological synapse formed by an scFv expressed by CAR-T cells and a target antigen, representing an ideal form of CAR-T-cell immunotherapy.


Assuntos
Cadeias Pesadas de Imunoglobulinas/metabolismo , Cadeias Leves de Imunoglobulina/metabolismo , Região Variável de Imunoglobulina/metabolismo , Imunoterapia Adotiva , Linfoma/terapia , Receptores de Antígenos Quiméricos/metabolismo , Anticorpos de Cadeia Única/metabolismo , Linfócitos T/transplante , Animais , Feminino , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Leves de Imunoglobulina/genética , Região Variável de Imunoglobulina/genética , Sinapses Imunológicas , Células Jurkat , Células K562 , Linfoma/genética , Linfoma/imunologia , Linfoma/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores de Antígenos Quiméricos/genética , Anticorpos de Cadeia Única/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Sci Rep ; 9(1): 13293, 2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31527633

RESUMO

The development of chimeric antigen receptor (CAR) and bispecific T-cell engager (BiTE) has led to the successful application of cancer immunotherapy. The potential reactivity mediated by CAR- and BiTE-redirected T cells needs to be assessed to facilitate the application of these treatment options to a broader range of patients. Here, we have generated CAR and BiTE possessing the same single chain fragment variable (scFv) specific for the HLA-A2/NY-ESO-1157-165 complex (A2/NY-ESO-1157). Using HLA-A2+NY-ESO-1+ myeloma cells and peptides presented by HLA-A2 molecules as a model, both sets of redirected T cells recognized and killed HLA-A2+NY-ESO-1+ myeloma cells in an A2/NY-ESO-1157-specific manner in vitro. Moreover, CAR- and BiTE-activated T cells showed similar functional avidity, as assessed by cytokine production and killing activity, both displaying antitumor reactivity against HLA-A2+NY-ESO-1+ myeloma cells in vivo. Interestingly, cross-reactivity for homologous peptides presented by HLA-A*02:01 and NY-ESO-1157 peptide presented by HLA-A2 alleles was not identical between CAR- and BiTE-redirected T cells, probably due to structural differences of modified antibodies. These results have demonstrated that both antitumor CAR- and BiTE-activated T cells have comparable potential to recognize tumors, while paying attention to unknown off-target reactivity that would differ for each antibody-based modality even if the same scFv was employed.


Assuntos
Antígenos de Neoplasias/imunologia , Antígeno HLA-A2/imunologia , Imunoterapia Adotiva/métodos , Proteínas de Membrana/imunologia , Mieloma Múltiplo/terapia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T Citotóxicos/transplante , Linhagem Celular Tumoral , Humanos , Região Variável de Imunoglobulina/imunologia , Ativação Linfocitária/imunologia , Mieloma Múltiplo/imunologia , Linfócitos T Citotóxicos/imunologia
15.
Methods Mol Biol ; 2048: 27-39, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31396926

RESUMO

Cancer immunotherapy has been developed and established as a new treatment modality. Recently, adoptive transfer therapy using T cells redirected with antigen-specific antitumor receptors, such as T-cell receptor (TCR) and chimeric antigen receptor (CAR), has demonstrated clinical benefits even in patients with refractory malignancies. To advance this treatment modality, both generation of gene-modified T cells and evaluation of their reactivity with high quality in vitro are required. To achieve this, it is important to establish the ways (1) to generate optimal viral particle for T-cell transduction, (2) to transduce antitumor receptors into T cells and expand redirected T cells efficiently, and (3) to assess the functionality of antigen-specific gene-modified T cells precisely. Here, we summarize established protocols to generate and analyze antitumor receptor-transduced T cells. These procedures help to further assess characteristics of gene-modified T cells, resulting in promotion of translational research for cancer immunotherapy.


Assuntos
Citometria de Fluxo/métodos , Proteínas do Tecido Nervoso/genética , Receptores de Antígenos Quiméricos/genética , Receptores de Fator de Crescimento Neural/genética , Linfócitos T/transplante , Transdução Genética/métodos , Linhagem Celular Tumoral , Separação Celular/instrumentação , Separação Celular/métodos , Ensaio de Imunoadsorção Enzimática/instrumentação , Ensaio de Imunoadsorção Enzimática/métodos , Citometria de Fluxo/instrumentação , Técnica Direta de Fluorescência para Anticorpo/instrumentação , Técnica Direta de Fluorescência para Anticorpo/métodos , Edição de Genes/métodos , Vetores Genéticos/genética , Humanos , Imunoterapia Adotiva/métodos , Ativação Linfocitária , Neoplasias/imunologia , Neoplasias/terapia , Proteínas do Tecido Nervoso/imunologia , Proteínas do Tecido Nervoso/metabolismo , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Receptores de Fator de Crescimento Neural/imunologia , Receptores de Fator de Crescimento Neural/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Retroviridae/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo
16.
Rinsho Ketsueki ; 60(7): 824-833, 2019.
Artigo em Japonês | MEDLINE | ID: mdl-31391373

RESUMO

With the emergence of cancer immunotherapy, T cells have played important roles in inducing antitumor responses. Many types of antitumor receptors, which possess tumor-binding sites and T-cell activation sites, have been developed. For example, genetically engineered T-cell receptor, chimeric antigen receptor, and bispecific antibody can help us to educate and activate T cells specific for certain tumors. To generate optimal antitumor receptors, (1) selection/distribution of tumor antigens, (2) affinity/specificity and cross-reactivity of antitumor receptors, and (3) T-cell activation signals delivered from antitumor receptors should be considered. Accordingly, we explain how antitumor receptors recognize target antigens and summarize the mechanisms for on-target/off-target reactivity induced by T cells redirected with antitumor receptors. Furthermore, we discuss how antitumor receptors can be optimized for the development of next-generation cancer immunotherapy.


Assuntos
Neoplasias Hematológicas/terapia , Imunoterapia Adotiva , Linfócitos T , Anticorpos Biespecíficos , Humanos , Receptores de Antígenos de Linfócitos T
17.
Sci Rep ; 8(1): 4804, 2018 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-29555965

RESUMO

While the principles of classical antigen presentation via MHC class II are well-established, the mechanisms for the many routes of cross-presentation by which endogenous antigens become associated with class II molecules are not fully understood. We have recently demonstrated that the single amino acid polymorphism HLA-DPß84Gly (DP84Gly) is critical to abrogate class II invariant chain associated peptide (CLIP) region-mediated binding of invariant chain (Ii) to DP, allowing endoplasmic reticulum (ER)-resident endogenous antigens to constitutively associate with DP84Gly such as DP4. In this study, we demonstrate that both the CLIP and N-terminal non-CLIP Ii regions cooperatively generate an Ii conformation that cannot associate with DP84Gly via the CLIP region. We also demonstrate the ability of DP4 to efficiently process and present antigens encoded in place of CLIP in a chimeric Ii, regardless of wild type Ii and HLA-DM expression. These data highlight the complex interplay between DP polymorphisms and the multiple Ii regions that cooperatively regulate this association, ultimately controlling the presentation of endogenous antigens on DP molecules. These results may also offer a mechanistic explanation for recent studies identifying the differential effects between DP84Gly and DP84Asp as clinically relevant in human disease.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos de Diferenciação de Linfócitos B/metabolismo , Antígenos HLA-DP/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Fragmentos de Peptídeos/metabolismo , Linfócitos T/imunologia , Antígenos de Diferenciação de Linfócitos B/genética , Antígenos de Diferenciação de Linfócitos B/imunologia , Antígenos HLA-D/genética , Antígenos HLA-D/imunologia , Antígenos HLA-D/metabolismo , Antígenos HLA-DP/genética , Antígenos HLA-DP/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Ativação Linfocitária , Fragmentos de Peptídeos/imunologia
18.
Nat Commun ; 8: 15244, 2017 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-28489076

RESUMO

Classical antigen processing leads to the presentation of antigenic peptides derived from endogenous and exogenous sources for MHC class I and class II molecules, respectively. Here we show that, unlike other class II molecules, prevalent HLA-DP molecules with ß-chains encoding Gly84 (DP84Gly) constitutively present endogenous peptides. DP84Gly does not bind invariant chain (Ii) via the class II-associated invariant chain peptide (CLIP) region, nor does it present CLIP. However, Ii does facilitate the transport of DP84Gly from the endoplasmic reticulum (ER) to the endosomal/lysosomal pathway by transiently binding DP84Gly via a non-CLIP region(s) in a pH-sensitive manner. Accordingly, like class I, DP84Gly constitutively presents endogenous peptides processed by the proteasome and transported to the ER by the transporter associated with antigen processing (TAP). Therefore, DP84Gly, found only in common chimpanzees and humans, uniquely uses both class I and II antigen-processing pathways to present peptides derived from intracellular and extracellular sources.


Assuntos
Apresentação de Antígeno/imunologia , Cadeias beta de HLA-DP/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Peptídeos/imunologia , Animais , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Células HEK293 , Cadeias beta de HLA-DP/genética , Cadeias beta de HLA-DP/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Células K562 , Lisossomos/imunologia , Lisossomos/metabolismo , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Peptídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Transporte Proteico/imunologia
19.
JCI Insight ; 2(2): e89580, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-28138559

RESUMO

Adoptive cell therapy is a potentially curative therapeutic approach for patients with cancer. In this treatment modality, antitumor T cells are exponentially expanded in vitro prior to infusion. Importantly, the results of recent clinical trials suggest that the quality of expanded T cells critically affects their therapeutic efficacy. Although anti-CD3 mAb-based stimulation is widely used to expand T cells in vitro, a protocol to generate T cell grafts for optimal adoptive therapy has yet to be established. In this study, we investigated the differences between T cell stimulation mediated by anti-CD3/CD28 mAb-coated beads and cell-based artificial antigen-presenting cells (aAPCs) expressing CD3/CD28 counter-receptors. We found that transient stimulation with cell-based aAPCs, but not prolonged stimulation with beads, resulted in the superior expansion of CD8+ T cells. Transiently stimulated CD8+ T cells maintained a stem cell-like memory phenotype and were capable of secreting multiple cytokines significantly more efficiently than chronically stimulated T cells. Importantly, the chimeric antigen receptor-engineered antitumor CD8+ T cells expanded via transient stimulation demonstrated superior persistence and antitumor responses in adoptive immunotherapy mouse models. These results suggest that restrained stimulation is critical for generating T cell grafts for optimal adoptive immunotherapy for cancer.


Assuntos
Imunização/métodos , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Linfócitos T/transplante , Animais , Células Apresentadoras de Antígenos , Antígenos CD28/imunologia , Complexo CD3/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/imunologia , Doença Enxerto-Hospedeiro , Humanos , Memória Imunológica/imunologia , Células K562 , Ativação Linfocitária/imunologia , Masculino , Camundongos , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Arthritis Rheumatol ; 69(5): 1054-1066, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28029757

RESUMO

OBJECTIVE: To identify risk alleles relevant to the causal and biologic mechanisms of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). METHODS: A genome-wide association study and subsequent replication study were conducted in a total cohort of 1,986 cases of AAV (patients with granulomatosis with polyangiitis [Wegener's] [GPA] or microscopic polyangiitis [MPA]) and 4,723 healthy controls. Meta-analysis of these data sets and functional annotation of identified risk loci were performed, and candidate disease variants with unknown functional effects were investigated for their impact on gene expression and/or protein function. RESULTS: Among the genome-wide significant associations identified, the largest effect on risk of AAV came from the single-nucleotide polymorphism variants rs141530233 and rs1042169 at the HLA-DPB1 locus (odds ratio [OR] 2.99 and OR 2.82, respectively) which, together with a third variant, rs386699872, constitute a triallelic risk haplotype associated with reduced expression of the HLA-DPB1 gene and HLA-DP protein in B cells and monocytes and with increased frequency of complementary proteinase 3 (PR3)-reactive T cells relative to that in carriers of the protective haplotype. Significant associations were also observed at the SERPINA1 and PTPN22 loci, the peak signals arising from functionally relevant missense variants, and at PRTN3, in which the top-scoring variant correlated with increased PRTN3 expression in neutrophils. Effects of individual loci on AAV risk differed between patients with GPA and those with MPA or between patients with PR3-ANCAs and those with myeloperoxidase-ANCAs, but the collective population attributable fraction for these variants was substantive, at 77%. CONCLUSION: This study reveals the association of susceptibility to GPA and MPA with functional gene variants that explain much of the genetic etiology of AAV, could influence and possibly be predictors of the clinical presentation, and appear to alter immune cell proteins and responses likely to be key factors in the pathogenesis of AAV.


Assuntos
Granulomatose com Poliangiite/genética , Cadeias beta de HLA-DP/genética , Poliangiite Microscópica/genética , Mieloblastina/genética , Proteína Tirosina Fosfatase não Receptora Tipo 22/genética , Linfócitos T/metabolismo , alfa 1-Antitripsina/genética , Adulto , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/genética , Autoantígenos/imunologia , Linfócitos B/metabolismo , Estudos de Casos e Controles , Feminino , Expressão Gênica , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Antígenos HLA-DP/metabolismo , Cadeias beta de HLA-DP/metabolismo , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , Mieloblastina/imunologia , Neutrófilos/metabolismo , Razão de Chances , Peroxidase/imunologia , Polimorfismo de Nucleotídeo Único , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA