Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Regen Ther ; 27: 104-111, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38545443

RESUMO

Introduction: Genetically modified human induced pluripotent stem cell (iPSC)-based regenerative medicine has substantial potential in the treatment of refractory human diseases. Thus, preclinical studies on the safety and efficacy of these products are essential. Non-human primate (NHP) models such as the rhesus macaque are highly similar to humans in terms of size, lifespan, and immune system, rendering them superior models. However, effective gene transduction in rhesus macaque iPSCs (Rh-iPSCs) remains challenging. In this study, we investigated the effective gene transduction into Rh-iPSCs and its effect on differentiation efficiency. Methods: We established a gene transduction method using the piggyBac transposon vector system. Gene transduced Rh-iPSCs were analyzed for undifferentiated markers. We did teratoma assay to check pluripotency. Gene transduced Rh-iPSCs were differentiated into hematopoietic stem and progenitor cells (HSPCs) and T-cell lineage cells. Additionally, gene transduced Rh-iPSCs were compared the differentiation efficiency with parental Rh-iPSCs. Results: We could establish a gene transduction method using the piggyBac transposon vector system, demonstrating high efficiency and stable transgene expression in Rh-iPSCs. These Rh-iPSCs maintained long-term gene expression while expressing undifferentiated markers. Teratoma assay indicated that these Rh-iPSCs had pluripotency. These Rh-iPSCs could differentiate into HPSCs and T cells that express transgenes. These Rh-iPSCs can differentiate into hematopoietic stem cells and T cells that express transgenes. No significant differences in efficiency of differentiation were observed between parental Rh-iPSCs and these Rh-iPSCs. Conclusions: These results indicate that the piggyBac transposon vector is an excellent gene transfer tool for rhesus macaque iPSCs and could contribute to the advancement of preclinical studies using rhesus macaque iPSCs.

2.
Nat Biomed Eng ; 7(1): 24-37, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36509913

RESUMO

The effectiveness of chimaeric antigen receptor (CAR) T-cell immunotherapies against solid tumours relies on the accumulation, proliferation and persistency of T cells at the tumour site. Here we show that the proliferation of CD8αß cytotoxic CAR T cells in solid tumours can be enhanced by deriving and expanding them from a single human induced-pluripotent-stem-cell clone bearing a CAR selected for efficient differentiation. We also show that the proliferation and persistency of the effector cells in the tumours can be further enhanced by genetically knocking out diacylglycerol kinase, which inhibits antigen-receptor signalling, and by transducing the cells with genes encoding for membrane-bound interleukin-15 (IL-15) and its receptor subunit IL-15Rα. In multiple tumour-bearing animal models, the engineered hiPSC-derived CAR T cells led to therapeutic outcomes similar to those of primary CD8 T cells bearing the same CAR. The optimization of effector CAR T cells derived from pluripotent stem cells may aid the development of long-lasting antigen-specific T-cell immunotherapies for the treatment of solid tumours.


Assuntos
Células-Tronco Pluripotentes Induzidas , Neoplasias , Animais , Humanos , Receptores de Antígenos de Linfócitos T/genética , Células-Tronco Pluripotentes Induzidas/patologia , Linfócitos T CD8-Positivos , Neoplasias/terapia , Proliferação de Células
3.
Commun Biol ; 4(1): 694, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099861

RESUMO

Tumor-infiltrating lymphocytes (TIL), which include tumor-specific T lymphocytes with frequency, are used for adoptive cell transfer therapy (ACT) in clinical practice. The optimization of TIL preparation has been investigated to reduce the senescence and increase the abundance of TIL, as both the quality and quantity of the transferred cells have great influence on the outcome of TIL-based ACT (TIL-ACT). Considering the effects of cell reprogramming on senescence, we expected that the anti-tumor effect could be enhanced by TIL regeneration. To confirm this hypothesis, we established tumor-specific TIL-derived iPS cells (TIL-iPSC) with human colorectal cancer specimens. T cells differentiated from TIL-iPSC (TIL-iPS-T) retained not only intrinsic T cell functions and tumor specificity, but also exhibited improved proliferation capacity and additional killing activity. Moreover, less differentiated profiles and prolonged persistency were seen in TIL-iPS-T compared with primary cells. Our findings imply that iPSC technology has great potential for TIL-ACT.


Assuntos
Neoplasias Colorretais/terapia , Células-Tronco Pluripotentes Induzidas/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos T/imunologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Neoplasias Colorretais/imunologia , Feminino , Humanos , Imunoterapia , Células-Tronco Pluripotentes Induzidas/citologia , Ativação Linfocitária , Linfócitos do Interstício Tumoral/citologia , Camundongos Endogâmicos NOD , Camundongos SCID , Linfócitos T/citologia , Linfócitos T/transplante
4.
Nat Biomed Eng ; 5(5): 429-440, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34002062

RESUMO

Avoiding the immune rejection of transplanted T cells is central to the success of allogeneic cancer immunotherapies. One solution to protecting T-cell grafts from immune rejection involves the deletion of allogeneic factors and of factors that activate cytotoxic immune cells. Here we report the generation of hypoimmunogenic cancer-antigen-specific T cells derived from induced pluripotent stem cells (iPSCs) lacking ß2-microglobulin, the class-II major histocompatibility complex (MHC) transactivator and the natural killer (NK) cell-ligand poliovirus receptor CD155, and expressing single-chain MHC class-I antigen E. In mouse models of CD20-expressing leukaemia or lymphoma, differentiated T cells expressing a CD20 chimeric antigen receptor largely escaped recognition by NKG2A+ and DNAM-1+ NK cells and by CD8 and CD4 T cells in the allogeneic recipients while maintaining anti-tumour potency. Hypoimmunogenic iPSC-derived T cells may contribute to the creation of off-the-shelf T cell immunotherapies.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Leucemia/terapia , Linfoma/terapia , Receptores Virais/genética , Linfócitos T/transplante , Microglobulina beta-2/genética , Animais , Antígenos de Diferenciação de Linfócitos T/metabolismo , Diferenciação Celular , Linhagem Celular , Técnicas de Inativação de Genes , Engenharia Genética , Humanos , Células-Tronco Pluripotentes Induzidas/imunologia , Leucemia/imunologia , Linfoma/imunologia , Masculino , Camundongos , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Mol Ther Methods Clin Dev ; 21: 262-273, 2021 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-33869654

RESUMO

Because of their close biological similarity to humans, non-human primate (NHP) models are very useful for the development of induced pluripotent stem cell (iPSC)-based cell and regenerative organ transplantation therapies. However, knowledge on the establishment, differentiation, and genetic modification of NHP-iPSCs, especially rhesus macaque iPSCs, is limited. We succeeded in establishing iPSCs from the peripheral blood of rhesus macaques (Rh-iPSCs) by combining the Yamanaka reprograming factors and two inhibitors (GSK-3 inhibitor [CHIR 99021] and MEK1/2 inhibitor [PD0325901]) and differentiated the cells into functional macrophages through hematopoietic progenitor cells. To confirm feasibility of the Rh-iPSC-derived macrophages as a platform for bioassays to model diseases, we knocked out TRIM5 gene in Rh-iPSCs by CRISPR-Cas9, which is a species-specific HIV resistance factor. TRIM5 knockout (KO) iPSCs had the same differentiation potential to macrophages as did Rh-iPSCs, but the differentiated macrophages showed a gain of sensitivity to HIV infection in vitro. Our reprogramming, gene editing, and differentiation protocols used to obtain Rh-iPSC-derived macrophages can be applied to other gene mutations, expanding the number of NHP gene therapy models.

6.
Nat Commun ; 12(1): 430, 2021 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-33462228

RESUMO

Clinical successes demonstrated by chimeric antigen receptor T-cell immunotherapy have facilitated further development of T-cell immunotherapy against wide variety of diseases. One approach is the development of "off-the-shelf" T-cell sources. Technologies to generate T-cells from pluripotent stem cells (PSCs) may offer platforms to produce "off-the-shelf" and synthetic allogeneic T-cells. However, low differentiation efficiency and poor scalability of current methods may compromise their utilities. Here we show improved differentiation efficiency of T-cells from induced PSCs (iPSCs) derived from an antigen-specific cytotoxic T-cell clone, or from T-cell receptor (TCR)-transduced iPSCs, as starting materials. We additionally describe feeder-free differentiation culture systems that span from iPSC maintenance to T-cell proliferation phases, enabling large-scale regenerated T-cell production. Moreover, simultaneous addition of SDF1α and a p38 inhibitor during T-cell differentiation enhances T-cell commitment. The regenerated T-cells show TCR-dependent functions in vitro and are capable of in vivo anti-tumor activity. This system provides a platform to generate a large number of regenerated T-cells for clinical application and investigate human T-cell differentiation and biology.


Assuntos
Técnicas de Cultura de Células/métodos , Imunoterapia Adotiva/métodos , Células-Tronco Pluripotentes Induzidas/fisiologia , Neoplasias/terapia , Linfócitos T Citotóxicos/transplante , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Quimiocina CXCL12/metabolismo , Meios de Cultura/metabolismo , Meios de Cultura/farmacologia , Feminino , Humanos , Imidazóis/farmacologia , Camundongos , Neoplasias/imunologia , Piridinas/farmacologia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T Citotóxicos/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cancer Sci ; 111(5): 1478-1490, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32133731

RESUMO

The use of allogeneic, pluripotent stem-cell-derived immune cells for cancer immunotherapy has been the subject of recent clinical trials. In Japan, investigator-initiated clinical trials will soon begin for ovarian cancer treatment using human leukocyte antigen (HLA)-homozygous-induced pluripotent stem cell (iPSC)-derived anti-glypican-3 (GPC3) chimeric antigen receptor (CAR)-expressing natural killer/innate lymphoid cells (NK/ILC). Using pluripotent stem cells as the source for allogeneic immune cells facilitates stringent quality control of the final product, in terms of efficacy, safety and producibility. In this paper, we describe our methods for the stable, feeder-free production of CAR-expressing NK/ILC cells from CAR-transduced iPSC with clinically relevant scale and materials. The average number of cells that could be differentiated from 1.8-3.6 × 106 iPSC within 7 weeks was 1.8-4.0 × 109 . These cells showed stable CD45/CD7/CAR expression, effector functions of cytotoxicity and interferon gamma (IFN-γ) production against GPC3-expressing tumor cells. When the CAR-NK/ILC cells were injected into a GPC3-positive, ovarian-tumor-bearing, immunodeficient mouse model, we observed a significant therapeutic effect that prolonged the survival of the animals. When the cells were injected into immunodeficient mice during non-clinical safety tests, no acute systemic toxicity or tumorigenicity of the final product or residual iPSC was observed. In addition, our test results for the CAR-NK/ILC cells generated with clinical manufacturing standards are encouraging, and these methods should accelerate the development of allogeneic pluripotent stem cell-based immune cell cancer therapies.


Assuntos
Glipicanas/imunologia , Células-Tronco Pluripotentes Induzidas/imunologia , Células Matadoras Naturais/imunologia , Linfócitos/imunologia , Receptores de Antígenos Quiméricos/imunologia , Animais , Diferenciação Celular , Sobrevivência Celular , Citotoxicidade Imunológica , Modelos Animais de Doenças , Feminino , Glipicanas/genética , Glipicanas/metabolismo , Humanos , Imunidade Inata , Imunoterapia Adotiva , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Interferon gama/imunologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/transplante , Transfusão de Linfócitos , Linfócitos/citologia , Camundongos , Camundongos SCID , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo
8.
Methods Mol Biol ; 2048: 59-70, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31396929

RESUMO

In this chapter, we describe a protocol for hematopoietic differentiation of human pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) derived from non-T cells, followed by the differentiation of the T-cell lineage. Derivation of T cells from PSCs involves three steps: induction of PSCs to hematopoietic progenitor cells (HPCs), differentiation of HPCs into progenitor T cells, and maturation of progenitor T cells into mature T cells (CD8 single-positive (SP) or CD4 SP).


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Meios de Cultura/metabolismo , Animais , Técnicas de Cultura de Células/instrumentação , Linhagem Celular , Separação Celular/instrumentação , Separação Celular/métodos , Técnicas de Cocultura/instrumentação , Técnicas de Cocultura/métodos , Fibroblastos , Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Técnica Direta de Fluorescência para Anticorpo/instrumentação , Técnica Direta de Fluorescência para Anticorpo/métodos , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Mesenquimais , Camundongos , Proteínas Recombinantes/metabolismo , Linfócitos T/fisiologia
9.
Methods Mol Biol ; 2048: 93-106, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31396934

RESUMO

In this chapter, we describe a protocol for hematopoietic differentiation of nonhuman primate (NHP)-induced pluripotent stem cells (iPSCs) derived from T cells and generation of T cells. Derivation of T cells from PSCs involves three steps: induction of PSCs to hematopoietic progenitor cells (HPCs), differentiation of HPCs into progenitor T cells, and maturation of progenitor T cells into mature T cells, in particular CD8 single-positive (SP) T cells.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Diferenciação Celular , Separação Celular/métodos , Citometria de Fluxo/métodos , Células-Tronco Pluripotentes Induzidas/fisiologia , Animais , Linhagem Celular , Separação Celular/instrumentação , Técnicas de Cocultura/instrumentação , Técnicas de Cocultura/métodos , Meios de Cultura/metabolismo , Citocinas/metabolismo , Fibroblastos , Citometria de Fluxo/instrumentação , Técnica Direta de Fluorescência para Anticorpo/instrumentação , Técnica Direta de Fluorescência para Anticorpo/métodos , Células-Tronco Hematopoéticas , Humanos , Células-Tronco Mesenquimais , Camundongos , Primatas
10.
Cancer Sci ; 110(1): 16-22, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30485606

RESUMO

Recent outstanding clinical results produced by engineered T cells, including chimeric antigen receptors, have already facilitated further research that broadens their applicability. One such direction is to explore new T cell sources for allogeneic "off-the-shelf" adoptive immunotherapy. Human pluripotent stem cells could serve as an alternative cell source for this purpose due to their unique features of infinite propagation ability and pluripotency. Here, we describe the current state of engineered T cell transfer with the focus on cell manufacturing processes and the potentials and challenges of induced pluripotent stem cell-derived T cells as a starting material to construct off-the-shelf T-cell banks.


Assuntos
Engenharia Celular/métodos , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Linfócitos T/transplante , Diferenciação Celular/imunologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/imunologia , Neoplasias/imunologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transplante Homólogo
11.
Cell Stem Cell ; 23(6): 850-858.e4, 2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30449714

RESUMO

Limited T cell availability and proliferative exhaustion present major barriers to successful T cell-based immunotherapies and may potentially be overcome through the use of "rejuvenated" induced pluripotent stem cells derived from antigen-specific T cells (T-iPSCs). However, strict antigen specificity is essential for safe and efficient T cell immunotherapy. Here, we report that CD8αß T cells from human T-iPSCs lose their antigen specificity through additional rearrangement of the T cell receptor (TCR) α chain gene during the CD4/CD8 double positive stage of in vitro differentiation. CRISPR knockout of a recombinase gene in the T-iPSCs prevented this additional TCR rearrangement. Moreover, when CD8αß T cells were differentiated from monocyte-derived iPSCs that were transduced with an antigen-specific TCR, they showed monoclonal expression of the transduced TCR. TCR-stabilized, regenerated CD8αß T cells effectively inhibit tumor growth in xenograft cancer models. These approaches could contribute to safe and effective regenerative T cell immunotherapies.


Assuntos
Antígenos CD8/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunoterapia , Células-Tronco Pluripotentes Induzidas/imunologia , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias/imunologia , Células Tumorais Cultivadas
12.
Mol Ther Nucleic Acids ; 12: 793-804, 2018 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-30141412

RESUMO

Highly active antiretroviral therapy (HAART) has markedly prolonged the prognosis of HIV-1 patients. However, lifelong dependency on HAART is a continuing challenge, and an effective therapeutic is much desired. Recently, introduction of short hairpin RNA (shRNA) targeting the HIV-1 promoter was found to suppress HIV-1 replication via transcriptional gene silencing (TGS). The technology is expected to be applied with hemato-lymphopoietic cell transplantation of HIV patients to suppress HIV transcription in transplanted hemato-lymphopoietic cells. Combination of the TGS technology with new cell transplantation strategy with induced pluripotent stem cell (iPSC)-derived hemato-lymphopoietic cells might contribute to new gene therapy in the HIV field. In this study, we evaluated iPSC-derived macrophage functions and feasibility of TGS technology in macrophages. Human iPSCs were transduced with shRNAs targeting the HIV-1 promoter region (shPromA) by using a lentiviral vector. The shPromA-transfected iPSCs were successfully differentiated into functional macrophages, and they exhibited strong protection against HIV-1 replication with alteration in the histone structure of the HIV-1 promoter region to induce heterochromatin formation. These results indicated that iPS-derived macrophage is a useful tool to investigate HIV infection and protection, and that the TGS technology targeting the HIV promoter is a potential candidate of new gene therapy.

13.
Stem Cell Reports ; 10(6): 1935-1946, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29805109

RESUMO

CD4+ T helper (Th) cell activation is essential for inducing cytotoxic T lymphocyte (CTL) responses against malignancy. We reprogrammed a Th clone specific for chronic myelogenous leukemia (CML)-derived b3a2 peptide to pluripotency and re-differentiated the cells into original TCR-expressing T-lineage cells (iPS-T cells) with gene expression patterns resembling those of group 1 innate lymphoid cells. CD4 gene transduction into iPS-T cells enhanced b3a2 peptide-specific responses via b3a2 peptide-specific TCR. iPS-T cells upregulated CD40 ligand (CD40L) expression in response to interleukin-2 and interleukin-15. In the presence of Wilms tumor 1 (WT1) peptide, antigen-specific dendritic cells (DCs) conditioned by CD4-modified CD40Lhigh iPS-T cells stimulated WT1-specific CTL priming, which eliminated WT1 peptide-expressing CML cells in vitro and in vivo. Thus, CD4 modification of CD40Lhigh iPS-T cells generates innate lymphoid helper-like cells inducing bcr-abl-specific TCR signaling that mediates effectiveanti-leukemic CTL responses via DC maturation, showing potential for adjuvant immunotherapy against leukemia.


Assuntos
Expressão Gênica , Imunidade Inata , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Biomarcadores , Ligante de CD40/metabolismo , Diferenciação Celular , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Imunofenotipagem , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/imunologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/imunologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/mortalidade , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Proteínas WT1/imunologia
14.
Stem Cell Reports ; 6(2): 213-27, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26862702

RESUMO

Vα24 invariant natural killer T (iNKT) cells are a subset of T lymphocytes implicated in the regulation of broad immune responses. They recognize lipid antigens presented by CD1d on antigen-presenting cells and induce both innate and adaptive immune responses, which enhance effective immunity against cancer. Conversely, reduced iNKT cell numbers and function have been observed in many patients with cancer. To recover these numbers, we reprogrammed human iNKT cells to pluripotency and then re-differentiated them into regenerated iNKT cells in vitro through an IL-7/IL-15-based optimized cytokine combination. The re-differentiated iNKT cells showed proliferation and IFN-γ production in response to α-galactosylceramide, induced dendritic cell maturation and downstream activation of both cytotoxic T lymphocytes and NK cells, and exhibited NKG2D- and DNAM-1-mediated NK cell-like cytotoxicity against cancer cell lines. The immunological features of re-differentiated iNKT cells and their unlimited availability from induced pluripotent stem cells offer a potentially effective immunotherapy against cancer.


Assuntos
Adjuvantes Imunológicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/citologia , Células Matadoras Naturais/citologia , Receptores de Antígenos de Linfócitos T/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo
15.
Semin Immunol ; 28(1): 35-44, 2016 02.
Artigo em Inglês | MEDLINE | ID: mdl-26589493

RESUMO

Induced pluripotent stem cells (iPSCs) describe somatic cells that have been reprogrammed to the pluripotent state from which they can then be differentiated into any cell type of the body. This ability has tremendous implications on a wide number of medical sciences and applications, including cancer treatments. In many cancer patients, tumor infiltrating lymphocytes (TILs) have reached an exhausted state and are unable to exert effector function despite detecting and localizing at the tumor. Although the isolation, ex vivo expansion and transplantation of TILs is effective in a significant group of patients, too many patients do not respond positively to this treatment, in part because the expanded TIL population does not include a sufficient number of cells with the naïve or memory phenotype. Cell reprogramming using iPSC technologies aims to overcome this problem by returning TILs to the pluripotent state from which they can be differentiated into a heterogeneous population of T cells that are best suited to combat the tumor.


Assuntos
Vacinas Anticâncer/imunologia , Reprogramação Celular , Imunoterapia Adotiva/métodos , Células-Tronco Pluripotentes Induzidas/fisiologia , Linfócitos do Interstício Tumoral/fisiologia , Neoplasias/terapia , Linfócitos T/fisiologia , Diferenciação Celular , Senescência Celular , Humanos , Memória Imunológica , Ativação Linfocitária , Neoplasias/imunologia , Linfócitos T/transplante
16.
Blood ; 125(2): 370-82, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25349175

RESUMO

Although overexpression of T-bet, a master transcription factor in type-1 helper T lymphocytes, has been reported in several hematologic and immune diseases, its role in their pathogenesis is not fully understood. In the present study, we used transgenic model mice (T-bet(tg/wt) and T-bet(tg/tg)) to investigate the effects of T-bet overexpression selectively in T lymphocytes on the development of hematologic and immune diseases. The results showed that T-bet overexpression in T cells spontaneously induced maturation arrest in the mononuclear phagocyte lineage, as well as spontaneous dermatitis and pulmonary alveolar proteinosis (PAP)-like disease in T-bet(tg/wt) and T-bet(tg/tg) mice, respectively. T-bet(tg/tg) alveoli with the PAP phenotype showed remarkable reorganization of alveolar mononuclear phagocyte subpopulations and impaired function, in addition to augmented T-cell infiltration. In addition, PAP development in T-bet(tg/tg) mice was found to be associated with increased migration of myeloid cells from the bone marrow into the peripheral blood. These findings reveal an unexpected link between T-bet overexpression in T lymphocytes and the development of PAP caused by reorganization of mononuclear phagocytes in the lung, and provide new insight into the molecular pathogenesis of secondary PAP accompanied by hematologic disorders.


Assuntos
Hematopoese/imunologia , Macrófagos/imunologia , Células Mieloides/imunologia , Proteinose Alveolar Pulmonar/imunologia , Proteínas com Domínio T/biossíntese , Animais , Citometria de Fluxo , Imuno-Histoquímica , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas com Domínio T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia
17.
Cell Stem Cell ; 12(1): 114-26, 2013 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-23290140

RESUMO

Adoptive immunotherapy with functional T cells is potentially an effective therapeutic strategy for combating many types of cancer and viral infection. However, exhaustion of antigen-specific T cells represents a major challenge to this type of approach. In an effort to overcome this problem, we reprogrammed clonally expanded antigen-specific CD8(+) T cells from an HIV-1-infected patient to pluripotency. The T cell-derived induced pluripotent stem cells were then redifferentiated into CD8(+) T cells that had a high proliferative capacity and elongated telomeres. These "rejuvenated" cells possessed antigen-specific killing activity and exhibited T cell receptor gene-rearrangement patterns identical to those of the original T cell clone from the patient. We also found that this method can be effective for generating specific T cells for other pathology-associated antigens. Thus, this type of approach may have broad applications in the field of adoptive immunotherapy.


Assuntos
Diferenciação Celular/fisiologia , Linfócitos T/citologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Linfócitos T/metabolismo
18.
Artigo em Japonês | MEDLINE | ID: mdl-21381400

RESUMO

Johne disease is ruminant chronic granulomatous enteritis caused by Mycobacterium avium subsp. paratuberculosis (MAP). The domestic animals infected with this pathogen present severe weight loss due to chronic diarrhea and a reduction in lactation yield. These result in enormous economic loss since the affected animals are subsequently subject to artificial selections and disinfection of the environment are absolutely necessary. Furthermore, MAP has been suspected to have pathological relationship to Crohn's disease, human chronic granulomatous enteritis. The bacterium grows slower on solid culture and its colony becomes visible after two months of culture. In Japan, there has been almost no investigation on pasteurization temperature of commercial milk using MAP. It comes from the fact that the growth rate of MAP is very slow and that MAP is a related species to Mycobacterium tuberculosis, which pasteurization condition has been well defined. The studies on the pasteurization conditions of commercial milk have been mainly targeted to reduce the risk of infection to Coxiella and Mycobacterium tuberculosis. However, there has been a concern about the possibility that MAP is remained in pasteurized milk because MAPs form an aggregate and the bacterium at its center may not receive enough heat to get pasteurized. From these reasons, the present study aims to investigate validity of the current pasteurization conditions of commercial milk by implementing experimental pasteurization at various pasteurization temperatures using milk experimentally infected with MAP, and to clarify if MAP is eliminated at these temperatures in order to achieve smooth enforcement of the current ministry order. We conducted plant pasteurization experiment at four pasteurization conditions (high temperature, short time (HTST); 82, 77, 72 degrees C for 15 seconds and low temperature, long time (LTLT); 63 degrees C for 30 minutes) using two MAP strains, ATCC19698 and OKY-20. In conclusion, there appeared no colony of the two MAP strains formed from the milk pasteurized at the four pasteurization conditions examined.


Assuntos
Laticínios , Manipulação de Alimentos , Microbiologia de Alimentos , Leite/microbiologia , Mycobacterium avium subsp. paratuberculosis , Esterilização , Temperatura , Animais , Indústria de Processamento de Alimentos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA