Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Nucleic Acids Res ; 52(1): 141-153, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-37985205

RESUMO

Genetic modification of specific genes is emerging as a useful tool to enhance the functions of antitumor T cells in adoptive immunotherapy. Current advances in CRISPR/Cas9 technology enable gene knockout during in vitro preparation of infused T-cell products through transient transfection of a Cas9-guide RNA (gRNA) ribonucleoprotein complex. However, selecting optimal gRNAs remains a major challenge for efficient gene ablation. Although multiple in silico tools to predict the targeting efficiency have been developed, their performance has not been validated in cultured human T cells. Here, we explored a strategy to select optimal gRNAs using our pooled data on CRISPR/Cas9-mediated gene knockout in human T cells. The currently available prediction tools alone were insufficient to accurately predict the indel percentage in T cells. We used data on the epigenetic profiles of cultured T cells obtained from transposase-accessible chromatin with high-throughput sequencing (ATAC-seq). Combining the epigenetic information with sequence-based prediction tools significantly improved the gene-editing efficiency. We further demonstrate that epigenetically closed regions can be targeted by designing two gRNAs in adjacent regions. Finally, we demonstrate that the gene-editing efficiency of unstimulated T cells can be enhanced through pretreatment with IL-7. These findings enable more efficient gene editing in human T cells.


Assuntos
Sistemas CRISPR-Cas , Técnicas de Inativação de Genes , Linfócitos T , Humanos , Sistemas CRISPR-Cas/genética , Edição de Genes , Linfócitos T/metabolismo
2.
Int Immunol ; 36(2): 49-56, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-37591521

RESUMO

Adoptive immunotherapy using chimeric antigen-receptor (CAR)-engineered T cells can induce robust antitumor responses against hematologic malignancies. However, its efficacy is not durable in the majority of the patients, warranting further improvement of T-cell functions. Cytokine signaling is one of the key cascades regulating T-cell survival and effector functions. In addition to cytokines that use the common γ chain as a receptor subunit, multiple cytokines regulate T-cell functions directly or indirectly. Modulating cytokine signaling in CAR-T cells by genetic engineering is one promising strategy to augment their therapeutic efficacy. These strategies include ectopic expression of cytokines, cytokine receptors, and synthetic molecules that mimic endogenous cytokine signaling. Alternatively, autocrine IL-2 signaling can be augmented through reprogramming of CAR-T cell properties through transcriptional and epigenetic modification. On the other hand, cytokine production by CAR-T cells triggers systemic inflammatory responses, which mainly manifest as adverse events such as cytokine-release syndrome (CRS) and neurotoxicity. In addition to inhibiting direct inflammatory mediators such as IL-6 and IL-1 released from activated macrophages, suppression of T-cell-derived cytokines associated with the priming of macrophages can be accomplished through genetic modification of CAR-T cells. In this review, I will outline recently developed synthetic biology approaches to exploit cytokine signaling to enhance CAR-T cell functions. I will also discuss therapeutic target molecules to prevent or alleviate CAR-T cell-related toxicities.


Assuntos
Imunoterapia Adotiva , Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos de Linfócitos T/genética , Citocinas/metabolismo , Terapia Baseada em Transplante de Células e Tecidos
3.
Int Immunol ; 36(7): 353-364, 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38517027

RESUMO

The efficient generation of chimeric antigen receptor (CAR) T cells is highly influenced by the quality of apheresed T cells. Healthy donor-derived T cells usually proliferate better than patients-derived T cells and are precious resources to generate off-the-shelf CAR-T cells. However, relatively little is known about the determinants that affect the efficient generation of CAR-T cells from healthy donor-derived peripheral blood mononuclear cells (PBMCs) compared with those from the patients' own PBMCs. We here examined the efficiency of CAR-T cell generation from multiple healthy donor samples and analyzed its association with the phenotypic features of the starting peripheral blood T cells. We found that CD62L expression levels within CD8+ T cells were significantly correlated with CAR-T cell expansion. Moreover, high CD62L expression within naïve T cells was associated with the efficient expansion of T cells with a stem cell-like memory phenotype, an indicator of high-quality infusion products. Intriguingly, genetic disruption of CD62L significantly impaired CAR-T cell proliferation and cytokine production upon antigen stimulation. Conversely, ectopic expression of a shedding-resistant CD62L mutant augmented CAR-T cell effector functions compared to unmodified CAR-T cells, resulting in improved antitumor activity in vivo. Collectively, we identified the surface expression of CD62L as a concise indicator of potent T-cell proliferation. CD62L expression is also associated with the functional properties of CAR-T cells. These findings are potentially applicable to selecting optimal donors to massively generate CAR-T cell products.


Assuntos
Imunoterapia Adotiva , Selectina L , Receptores de Antígenos Quiméricos , Selectina L/metabolismo , Selectina L/imunologia , Humanos , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Animais , Camundongos , Imunoterapia Adotiva/métodos , Proliferação de Células
4.
Blood ; 139(14): 2156-2172, 2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-34861037

RESUMO

Adoptive cancer immunotherapy can induce objective clinical efficacy in patients with advanced cancer; however, a sustained response is achieved in a minority of cases. The persistence of infused T cells is an essential determinant of a durable therapeutic response. Antitumor T cells undergo a genome-wide remodeling of the epigenetic architecture upon repeated antigen encounters, which inevitably induces progressive T-cell differentiation and the loss of longevity. In this study, we identified PR domain zinc finger protein 1 (PRDM1) ie, Blimp-1, as a key epigenetic gene associated with terminal T-cell differentiation. The genetic knockout of PRDM1 by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) supported the maintenance of an early memory phenotype and polyfunctional cytokine secretion in repeatedly stimulated chimeric antigen receptor (CAR)-engineered T cells. PRDM1 disruption promoted the expansion of less differentiated memory CAR-T cells in vivo, which enhanced T-cell persistence and improved therapeutic efficacy in multiple tumor models. Mechanistically, PRDM1-ablated T cells displayed enhanced chromatin accessibility of the genes that regulate memory formation, thereby leading to the acquisition of gene expression profiles representative of early memory T cells. PRDM1 knockout also facilitated maintaining an early memory phenotype and cytokine polyfunctionality in T-cell receptor-engineered T cells as well as tumor-infiltrating lymphocytes. In other words, targeting PRDM1 enabled the generation of superior antitumor T cells, which is potentially applicable to a wide range of adoptive cancer immunotherapies.


Assuntos
Imunoterapia Adotiva , Neoplasias , Citocinas , Técnicas de Inativação de Genes , Humanos , Ativação Linfocitária , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética
5.
Blood ; 138(24): 2555-2569, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34587247

RESUMO

Neutrophils play an essential role in innate immune responses to bacterial and fungal infections, and loss of neutrophil function can increase the risk of acquiring lethal infections in clinical settings. Here, we show that engineered neutrophil-primed progenitors derived from human induced pluripotent stem cells can produce functional neutrophil-like cells at a clinically applicable scale that can act rapidly in vivo against lethal bacterial infections. Using 5 different mouse models, we systematically demonstrated that these neutrophil-like cells migrate to sites of inflammation and infection and increase survival against bacterial infection. In addition, we found that these human neutrophil-like cells can recruit murine immune cells. This system potentially provides a straight-forward solution for patients with neutrophil deficiency: an off-the-shelf neutrophil transfusion. This platform should facilitate the administration of human neutrophils for a broad spectrum of physiological and pathological conditions.


Assuntos
Infecções Bacterianas/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Neutrófilos/transplante , Animais , Infecções Bacterianas/imunologia , Técnicas de Cultura de Células , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Humanos , Imunidade Inata , Células-Tronco Pluripotentes Induzidas/imunologia , Inflamação/imunologia , Inflamação/terapia , Camundongos Endogâmicos BALB C , Neutrófilos/citologia , Neutrófilos/imunologia
6.
Int Immunol ; 34(11): 547-553, 2022 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-35561668

RESUMO

Our understanding of mechanisms underlying T-cell exhaustion has been refined by analysis of exhausted T cells at the molecular level. The development and functions of exhausted T cells are regulated by a number of transcription factors, epigenetic factors and metabolic enzymes. In addition, recent work to dissect exhausted T cells at the single-cell level has enabled us to discover a precursor exhausted T-cell subset equipped with long-term survival capacity. Starting from the analysis of mouse models, the existence of precursor exhausted T cells has also been documented in human T cells in the context of chronic virus infections or tumors. Clinical data suggest that evaluating the quality of exhausted T cells on the basis of their differentiation status may be helpful to predict the therapeutic response to inhibition of programmed death 1 (PD1). Moreover, beyond immune-checkpoint blockade, novel therapeutic approaches to re-invigorate exhausted T cells have been explored based on molecular insights into T-cell exhaustion. Here I will discuss key molecular profiles associated with the development, maintenance and differentiation of exhausted T cells and how these findings can be applicable in the field of cancer immunotherapy.


Assuntos
Linfócitos T CD8-Positivos , Receptor de Morte Celular Programada 1 , Animais , Linfócitos T CD8-Positivos/patologia , Humanos , Inibidores de Checkpoint Imunológico , Camundongos , Subpopulações de Linfócitos T/metabolismo , Fatores de Transcrição
7.
Cancer Sci ; 113(11): 3664-3671, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36000807

RESUMO

Recent advancements in cancer immunotherapy, such as chimeric antigen receptor (CAR)-engineered T cell therapy and immune checkpoint therapy, have significantly improved the clinical outcomes of patients with several types of cancer. To broaden its applicability further and induce durable therapeutic efficacy, it is imperative to understand how antitumor T cells elicit cytotoxic functions, survive as memory T cells, or are impaired in their effector functions (exhausted) at the molecular level. T cell properties are regulated by their gene expression profiles, which are further controlled by epigenetic architectures, such as DNA methylation and histone modifications. Multiple studies have elucidated specific epigenetic genes associated with T-cell phenotypic changes. Conversely, exogenous modification of these key epigenetic factors can significantly alter T cell functions by extensively altering the transcription network, which can be applied in cancer immunotherapy by improving T cell persistence or augmenting effector functions. As CAR-T cell therapy involves a genetic engineering step during the preparation of the infusion products, it would be a feasible strategy to additionally modulate specific epigenetic genes in CAR-T cells to improve their quality. Here, we review recent studies investigating how individual epigenetic factors play a crucial role in T-cell biology. We further discuss future directions to integrate these findings for optimal cancer immunotherapy.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Humanos , Imunoterapia Adotiva/efeitos adversos , Neoplasias/genética , Neoplasias/terapia , Terapia Baseada em Transplante de Células e Tecidos , Epigênese Genética , Receptores de Antígenos de Linfócitos T
8.
Rinsho Ketsueki ; 63(9): 1290-1297, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-36198555

RESUMO

Chimeric antigen receptor (CAR)-engineered T-cell therapy against B-cell malignancies and multiple myeloma was recently introduced for clinical use. However, the efficacy of CAR-T cell therapy is not durable in most patients, warranting the development of CAR-T cells with additional genetic modification or engineering of synthetic molecules to enhance their functions. This review will provide an overview of the molecular mechanisms underlying the functional alteration of T cells, especially transcriptional networks associated with memory formation and T cell exhaustion. In addition, methods to rationally improve CAR-T cell functions based on these mechanistic insights will be discussed.


Assuntos
Mieloma Múltiplo , Receptores de Antígenos Quiméricos , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Imunoterapia Adotiva , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética , Linfócitos T
9.
Rinsho Ketsueki ; 63(7): 782-789, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-35922948

RESUMO

The efficacy of adoptive immunotherapy using CD19-targeting chimeric antigen receptor (CAR)-engineered T cells against B-cell malignancies has already been established in the clinic. However, high economic costs and heterogeneous quality of CAR-T cells derived from individual patients hinder further expansion of their applicability to various cancer types, including solid tumors. Mass CAR-T cell production from healthy donors is a promising approach to overcome these problems, given that allogeneic immunity elicited against donor CAR-T cells by the recipient's immune system is controlled. CAR-T cells genetically ablated with T-cell receptor and human leukocyte antigen molecules, referred to as universal CAR-T cells, may enable the use of allogeneic T cells for off-the-shelf adoptive cancer immunotherapy. However, several concerns, such as poor persistence of infused CAR-T cells and chromosomal abnormalities due to genome editing, remain to be addressed. Thus, recent clinical trials on universal CAR-T cells are summarized and future perspectives to overcome current challenges are discussed in this review.


Assuntos
Neoplasias , Receptores de Antígenos de Linfócitos T , Antígenos CD19 , Humanos , Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T
10.
Gan To Kagaku Ryoho ; 49(6): 609-614, 2022 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-35799382

RESUMO

T cell exhaustion is induced in the context of chronic virus infection and tumor microenvironment, in which cytotoxic T cells are repeatedly exposed to the target antigen and deprived of their effector functions. Multiple studies have already shown the significant impact of immune checkpoint molecules such as PD1 on functional properties of exhausted T cells. In addition to these signals, exhausted T cells possess distinct transcriptional and epigenetic profiles compared with conventional effector and memory T cells. Importantly, most of these features are not affected by immune checkpoint blockade, suggesting that genetic and epigenetic remodeling of T cells is an underlying molecular mechanism essential for T cell exhaustion. Moreover, it has now been evident that exhausted T cells are a heterogeneous cell population composed of distinct T cell subsets, and these functional differences profoundly affect therapeutic efficacy of cancer immunotherapy. In this review, I will discuss recent studies investigating molecular mechanisms of T cell exhaustion, including novel key molecules essentially associated with T cell exhaustion. These findings are potentially applicable to reinvigorate effector functions of exhausted T cells.


Assuntos
Inibidores de Checkpoint Imunológico , Imunoterapia , Antígenos , Humanos , Contagem de Linfócitos , Microambiente Tumoral
11.
Cancer Sci ; 112(10): 4112-4126, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34363719

RESUMO

Evi1 is a transcription factor essential for the development as well as progression of acute myeloid leukemia (AML) and high Evi1 AML is associated with extremely poor clinical outcome. Since targeting metabolic vulnerability is the emerging therapeutic strategy of cancer, we herein investigated a novel therapeutic target of Evi1 by analyzing transcriptomic, epigenetic, and metabolomic profiling of mouse high Evi1 leukemia cells. We revealed that Evi1 overexpression and Evi1-driven leukemic transformation upregulate transcription of gluconeogenesis enzyme Fbp1 and other pentose phosphate enzymes with interaction between Evi1 and the enhancer region of these genes. Metabolome analysis using Evi1-overexpressing leukemia cells uncovered pentose phosphate pathway upregulation by Evi1 overexpression. Suppression of Fbp1 as well as pentose phosphate pathway enzymes by shRNA-mediated knockdown selectively decreased Evi1-driven leukemogenesis in vitro. Moreover, pharmacological or shRNA-mediated Fbp1 inhibition in secondarily transplanted Evi1-overexpressing leukemia mouse significantly decreased leukemia cell burden. Collectively, targeting FBP1 is a promising therapeutic strategy of high Evi1 AML.


Assuntos
Frutose-Bifosfatase/metabolismo , Leucemia Mieloide Aguda/etiologia , Leucemia Mieloide Aguda/metabolismo , Proteína do Locus do Complexo MDS1 e EVI1/metabolismo , Via de Pentose Fosfato , Animais , Modelos Animais de Doenças , Progressão da Doença , Elementos Facilitadores Genéticos , Epigênese Genética , Frutose-Bifosfatase/antagonistas & inibidores , Frutose-Bifosfatase/genética , Perfilação da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/patologia , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Via de Pentose Fosfato/genética , RNA Interferente Pequeno , Ensaio Tumoral de Célula-Tronco , Regulação para Cima
12.
J Immunol ; 200(2): 500-511, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29237773

RESUMO

In humans, a substantial portion of T cells recognize lipids presented by the monomorphic CD1 proteins. Recent studies have revealed the molecular basis of mycobacterial lipid recognition by CD1c-restricted T cells. Subsets of CD1c-restricted T cells recognize self-lipids in addition to foreign lipids, which may have implications in human diseases involving autoimmunity and malignancy. However, the molecular identity of these self-reactive T cells remains largely elusive. In this study, using a novel CD1c+ artificial APC (aAPC)-based system, we isolated human CD1c-restricted autoreactive T cells and characterized them at the molecular level. By using the human cell line K562, which is deficient in MHC class I/II and CD1 expression, we generated an aAPC expressing CD1c as the sole Ag-presenting molecule. When stimulated with this CD1c+ aAPC presenting endogenous lipids, a subpopulation of primary CD4+ T cells from multiple donors was consistently activated, as measured by CD154 upregulation and cytokine production in a CD1c-specific manner. These activated CD4+ T cells preferentially expressed TRBV4-1+ TCRs. Clonotypic analyses of the reconstituted TRBV4-1+ TCR genes confirmed CD1c-restricted autoreactivity of this repertoire, and the strength of CD1c reactivity was influenced by the diversity of CDR3ß sequences. Finally, alanine scanning of CDR1 and CDR2 sequences of TRBV4-1 revealed two unique residues, Arg30 and Tyr51, as critical in conferring CD1c-restricted autoreactivity, thus elucidating the molecular basis of the observed V gene bias. These data provide new insights into the molecular identity of human autoreactive CD1c-restricted T cells.


Assuntos
Antígenos CD1/imunologia , Antígenos CD1/metabolismo , Autoimunidade , Expressão Gênica , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Arginina/genética , Biomarcadores , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular , Evolução Clonal/genética , Evolução Clonal/imunologia , Códon , Regiões Determinantes de Complementaridade/genética , Humanos , Imunofenotipagem , Ativação Linfocitária , Fenótipo , Tirosina/genética
13.
J Autoimmun ; 102: 114-125, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31078377

RESUMO

Recent work has delineated key differences in the antigen processing and presentation mechanisms underlying HLA-DP alleles encoding glycine at position 84 of the DPß chain (DP84GGPM87). These DPs are unable to associate with the class II-associated Ii peptide (CLIP) region of the invariant chain (Ii) chaperone early in the endocytic pathway, leading to continuous presentation of endogenous antigens. However, little is known about the chaperone support involved in the loading of these endogenous antigens onto DP molecules. Here, we demonstrate the proteasome and TAP dependency of this pathway and reveal the ability of HLA class I to compete with DP84GGPM87 for the presentation of endogenous antigens, suggesting that shared subcellular machinery may exist between the two classes of HLA. We identify physical interactions of prototypical class I-associated chaperones with numerous DP alleles, including TAP2, tapasin, ERp57, calnexin, and calreticulin, using a conventional immunoprecipitation and immunoblot approach and confirm the existence of these interactions in vivo through the use of the BioID2 proximal biotinylation system in human cells. Based on immunological assays, we then demonstrate the ability of each of these chaperones to facilitate the presentation of endogenously derived, but not exogenously derived, antigens on DP molecules. Considering previous genetic and clinical studies linking DP84GGPM87 to disease frequency and severity in autoimmune disease, viral infections, and cancer, we suggest that the above chaperones may form the molecular basis of these observable clinical differences through facilitating the presentation of endogenously derived antigens to CD4+ T cells.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos HLA-DP/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Chaperonas Moleculares/imunologia , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/imunologia , Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Calnexina/genética , Calnexina/imunologia , Calreticulina/genética , Calreticulina/imunologia , Linhagem Celular , Células HEK293 , Humanos , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/imunologia , Chaperonas Moleculares/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/imunologia
14.
J Autoimmun ; 97: 10-21, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30318155

RESUMO

Forkhead box transcription factor 3 (FOXP3) plays a pivotal role in the suppressive function of regulatory T cells. In addition to mRNA levels, FOXP3 activity can also be controlled by posttranslational mechanisms, which have not been studied in a comprehensive manner. Through extensive screening using selective inhibitors, we demonstrate that the inhibition of type I protein arginine methytransferases (PRMTs) attenuates the suppressive functions of regulatory T cells. FOXP3 undergoes methylation on arginine residues at positions 48 and 51 by interacting with protein arginine methyltransferase 1 (PRMT1). The inhibition of arginine methylation confers gene expression profiles representing type I helper T cells to FOXP3+ T cells, which results in attenuated suppressive activity. A methylation-defective mutant of FOXP3 displays less potent activity to suppress xenogeneic graft-versus-host disease in vivo. These results elucidate an important role of arginine methylation to enhance FOXP3 functions and are potentially applicable to modulate regulatory T cell functions.


Assuntos
Arginina/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Linfócitos T Reguladores/metabolismo , Animais , Biomarcadores , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Imunofluorescência , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/metabolismo , Doença Enxerto-Hospedeiro/mortalidade , Humanos , Estimativa de Kaplan-Meier , Masculino , Metilação , Camundongos , Mutação , Processamento de Proteína Pós-Traducional , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia
15.
J Immunol ; 198(3): 1056-1065, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28003379

RESUMO

The human invariant NK (iNK) TCR is largely composed of the invariant TCR Vα24-Jα18 chain and semivariant TCR Vß11 chains with variable CDR3ß sequences. The direct role of CDR3ß in Ag recognition has been studied extensively. Although it was noted that CDR3ß can interact with CDR3α, how this interaction might indirectly influence Ag recognition is not fully elucidated. We observed that the third position of Vß11 CDR3 can encode an Arg or Ser residue as a result of somatic rearrangement. Clonotypic analysis of the two iNK TCR types with a single amino acid substitution revealed that the staining intensity by anti-Vα24 Abs depends on whether Ser or Arg is encoded. When stained with an anti-Vα24-Jα18 Ab, human primary invariant NKT cells could be divided into Vα24 low- and high-intensity subsets, and Arg-encoding TCR Vß11 chains were more frequently isolated from the Vα24 low-intensity subpopulation compared with the Vα24 high-intensity subpopulation. The Arg/Ser substitution also influenced Ag recognition as determined by CD1d multimer staining and CD1d-restricted functional responses. Importantly, in silico modeling validated that this Ser-to-Arg mutation could alter the structure of the CDR3ß loop, as well as the CDR3α loop. Collectively, these results indicate that the Arg/Ser encoded at the third CDR3ß residue can effectively modulate the overall structure of, and Ag recognition by, human iNK TCRs.


Assuntos
Antígenos/imunologia , Células T Matadoras Naturais/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Antígenos CD1d/imunologia , Regiões Determinantes de Complementaridade/química , Humanos , Simulação de Dinâmica Molecular
16.
Blood ; 124(24): 3587-96, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25298035

RESUMO

Ecotropic viral integration site 1 (Evi1) is a transcription factor that is highly expressed in hematopoietic stem cells and is crucial for their self-renewal capacity. Aberrant expression of Evi1 is observed in 5% to 10% of de novo acute myeloid leukemia (AML) patients and predicts poor prognosis, reflecting multiple leukemogenic properties of Evi1. Here, we show that thrombopoietin (THPO) signaling is implicated in growth and survival of Evi1-expressing cells using a mouse model of Evi1 leukemia. We first identified that the expression of megakaryocytic surface molecules such as ITGA2B (CD41) and the THPO receptor, MPL, positively correlates with EVI1 expression in AML patients. In agreement with this finding, a subpopulation of bone marrow and spleen cells derived from Evi1 leukemia mice expressed both CD41 and Mpl. CD41(+) Evi1 leukemia cells induced secondary leukemia more efficiently than CD41(-) cells in a serial bone marrow transplantation assay. Importantly, the CD41(+) cells predominantly expressing Mpl effectively proliferated and survived on OP9 stromal cells in the presence of THPO via upregulating BCL-xL expression, suggesting an essential role of the THPO/MPL/BCL-xL cascade in enhancing the progression of Evi1 leukemia. These observations provide a novel aspect of the diverse functions of Evi1 in leukemogenesis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Leucemia/metabolismo , Neoplasias Experimentais/metabolismo , Proteínas Oncogênicas/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Receptores de Trombopoetina/metabolismo , Transdução de Sinais , Trombopoetina/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sobrevivência Celular , Proteínas de Ligação a DNA/genética , Regulação Leucêmica da Expressão Gênica/genética , Leucemia/genética , Leucemia/patologia , Proteína do Locus do Complexo MDS1 e EVI1 , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Proteínas Oncogênicas/genética , Glicoproteína IIb da Membrana de Plaquetas/genética , Proto-Oncogenes/genética , Receptores de Trombopoetina/genética , Trombopoetina/genética , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Proteína bcl-X/biossíntese , Proteína bcl-X/genética
17.
Blood ; 124(19): 2996-3006, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25217696

RESUMO

Genetic instability is strongly involved in cancer development and progression, and elucidating the mechanism could lead to novel therapeutics for preventing carcinogenesis. Philadelphia-negative myeloproliferative neoplasms (MPNs) are clonal myeloid disorders with a high prevalence of JAK2V617F mutation, and transformation to acute myeloid leukemia through accumulation of additional mutations is a major complication in MPNs. Here, we showed that JAK2V617F(+) cells conferred paracrine DNA damage to neighboring normal cells as well as to themselves through increased reactive oxygen species (ROS). We screened candidate factors responsible for the effect and found that lipocalin-2 (Lcn2) is overexpressed in JAK2V617F(+) cells and that short hairpin RNA-mediated knockdown of Lcn2 significantly alleviated the paracrine DNA damage. Normal hematopoietic cells showed elevated ROS levels through increased intracellular iron levels when treated with lipocalin-2, which led to p53 pathway activation, increased apoptosis, and decreased cellular proliferation. In contrast, JAK2V617F(+) cells did not suffer from lipocalin-2-induced growth suppression resulting from attenuated p53 pathway activation, which conferred a relative growth advantage to JAK2V617F(+) clones. In summary, we demonstrated that JAK2V617F-harboring cells cause paracrine DNA damage accumulation through secretion of lipocalin-2, which gives proliferative advantage to themselves and an increased risk for leukemic transformation to both JAK2V617F(+) and JAK2V617F(-) clones.


Assuntos
Proteínas de Fase Aguda/metabolismo , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Lipocalinas/metabolismo , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Proteínas Oncogênicas/metabolismo , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Dano ao DNA/fisiologia , Feminino , Hematopoese/fisiologia , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Janus Quinase 2/antagonistas & inibidores , Lipocalina-2 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitrilas , Estresse Oxidativo/genética , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/genética , Pirazóis/farmacologia , Pirimidinas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
18.
Rinsho Ketsueki ; 56(4): 384-91, 2015 Apr.
Artigo em Japonês | MEDLINE | ID: mdl-25971268

RESUMO

Acute myeloid leukemia (AML) is a hematologic malignancy which originates from leukemia initiating cells (LICs). Since LICs are considered to be resistant to standard chemotherapy, the identification of common mechanisms involved in LIC maintenance and progression is crucial to establishing broadly effective therapeutic agents for AML. The NF-κB pathway is one of the most promising targets, considering that its constitutive activation has been reported in different types of AML. In myeloid leukemia mouse models, we found that NF-κB activity is maintained specifically in LICs through autocrine TNF-α secretion, forming an NF-κB/TNF-α positive feedback loop. LICs had increased levels of active proteasome machinery, which promoted the degradation of IκBα and further supported NF-κB activity. Genetic ablation of TNF-α or NF-κB markedly suppressed leukemia progression in vivo. These findings demonstrate that NF-κB/TNF-α signaling in LICs contributes to leukemia progression and provide a widely applicable approach for targeting LICs.


Assuntos
Proteínas I-kappa B/metabolismo , Leucemia Mieloide Aguda/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Inibidor de NF-kappaB alfa , NF-kappa B/genética , Transdução de Sinais/fisiologia
19.
Cancer Sci ; 105(5): 512-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24612037

RESUMO

Leukemia stem cells (LSC) are resistant to conventional chemotherapy and persistent LSC after chemotherapy are supposed to be a major cause of relapse. However, information on genetic or epigenetic regulation of stem cell properties is still limited and LSC-targeted drugs have scarcely been identified. Epigenetic regulators are associated with many cellular processes including maintenance of stem cells. Of note are polycomb group proteins, because they potentially control stemness, and can be pharmacologically targeted by a selective inhibitor (DZNep). Therefore, we investigated the therapeutic potential of EZH2 inhibition in mixed lineage leukemia (MLL) fusion leukemia. Intriguingly, EZH2 inhibition by DZNep or shRNA not only suppressed MLL fusion leukemia proliferation but also reduced leukemia initiating cells (LIC) frequency. Expression analysis suggested that p16 upregulation was responsible for LICs reduction. Knockdown of p16 canceled the survival advantage of mice treated with DZNep. Chromatin immunoprecipitation assays demonstrated that EZH2 was highly enriched around the transcription-start-site of p16, together with H3K27 methylation marks in MLL/ENL and Hoxa9/Meis1 transduced cells but not in E2A/HLF transduced cells. Although high expression of Hoxa9 in MLL fusion leukemia is supposed to be responsible for the recruitment of EZH2, our data also suggest that there may be some other mechanisms independent of Hoxa9 activation to suppress p16 expression, because expression levels of Hoxa9 and p16 were not inversely related between MLL/ENL and Hoxa9/Meis1 transduced cells. In summary, our findings show that EZH2 is a potential therapeutic target of MLL fusion leukemia stem cells.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Leucemia/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Complexo Repressor Polycomb 2/antagonistas & inibidores , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição de Zíper de Leucina Básica/genética , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Leucemia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína Meis1 , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Ativação Transcricional , Transplante Heterólogo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA