Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38683966

RESUMO

Relapse is the leading cause of death after allogeneic hematopoietic stem cell transplantation (HCT) for leukemia. T cells engineered by gene transfer to express T cell receptors (TCR; TCR-T) specific for hematopoietic-restricted minor histocompatibility (H) antigens may provide a potent selective anti-leukemic effect post-HCT. We conducted a phase I clinical trial employing a novel TCR-T product targeting the minor H antigen HA-1 to treat or consolidate treatment of persistent or recurrent leukemia and myeloid neoplasms. The primary objective was to evaluate the feasibility and safety of administration of HA-1 TCR-T post-HCT. CD8+ and CD4+ T cells expressing the HA-1 TCR and a CD8-co-receptor were successfully manufactured from HA-1 disparate HCT donors. One or more infusions of HA-1 TCR-T following lymphodepleting chemotherapy were administered to nine HCT recipients who had developed disease recurrence post-HCT. TCR-T cells expanded and persisted in vivo after adoptive transfer. No dose-limiting toxicities occurred. Although the study was not designed to assess efficacy, four patients achieved or maintained complete remissions following lymphodepletion and HA-1 TCR-T, with one ongoing at >2 years. Single-cell RNA sequencing of relapsing/progressive leukemia after TCR-T therapy identified upregulated molecules associated with T cell dysfunction or cancer cell survival. HA-1 TCR-T therapy appears feasible and safe and shows preliminary signals of efficacy. This clinical trial is registered at clinicaltrials.gov as NCT03326921.

2.
J Immunol ; 200(8): 2748-2756, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29540580

RESUMO

The sensing of viral nucleic acids within the cytosol is essential for the induction of innate immune responses following infection. However, this sensing occurs within cells that have already been infected. The death of infected cells can be beneficial to the host by eliminating the virus's replicative niche and facilitating the release of inflammatory mediators. In this study, we show that sensing of intracellular DNA or RNA by cGAS-STING or RIG-I-MAVS, respectively, leads to activation of RIPK3 and necroptosis in bone marrow-derived macrophages. Notably, this requires signaling through both type I IFN and TNF receptors, revealing synergy between these pathways to induce cell death. Furthermore, we show that hyperactivation of STING in mice leads to a shock-like phenotype, the mortality of which requires activation of the necroptotic pathway and IFN and TNF cosignaling, demonstrating that necroptosis is one outcome of STING signaling in vivo.


Assuntos
Morte Celular/fisiologia , Interferon Tipo I/metabolismo , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , DNA Viral/imunologia , Imunidade Inata/imunologia , Interferon Tipo I/imunologia , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , RNA Viral/imunologia , Fator de Necrose Tumoral alfa/imunologia , Viroses/imunologia
3.
Immunol Cell Biol ; 95(2): 131-136, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27909314

RESUMO

Necroptosis is a lytic form of programmed cell death that involves the swelling and rupture of dying cells. Although several necroptosis-inducing stimuli have been defined, in most cells this pathway is kept in check by the action of the pro-apoptotic protease caspase-8 and the IAP ubiquitin ligases. How and when necroptosis is triggered under physiological conditions therefore remains a persistent question. Because necroptosis likely arose as a defensive mechanism against viral infection, exploration of this question requires a consideration of host-pathogen interactions, and how the sensing of infection could sensitize cells to necroptosis. Here, we will discuss the role of necroptosis in the response to viral infection, consider why the necroptotic pathway has been favored during evolution, and describe emerging evidence for death-independent functions of key necroptotic signaling components.


Assuntos
Apoptose , Interações Hospedeiro-Patógeno , Animais , Evolução Biológica , Humanos , Inflamação/patologia , Modelos Biológicos , Necrose
4.
Nucleic Acids Res ; 42(1): e4, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24121685

RESUMO

The creation of a DNA break at a specific locus by a designer endonuclease can be harnessed to edit a genome. However, DNA breaks may engage one of several competing repair pathways that lead to distinct types of genomic alterations. Therefore, understanding the contribution of different repair pathways following the introduction of a targeted DNA break is essential to further advance the safety and efficiency of nuclease-induced genome modification. To gain insight into the role of different DNA repair pathways in resolving nuclease-induced DNA breaks into genome editing outcomes, we previously developed a fluorescent-based reporter system, designated the Traffic Light Reporter, which provides a readout of gene targeting and gene disruption downstream of a targeted DNA double-strand break. Here we describe two related but novel reporters that extend this technology: one that allows monitoring of the transcriptional activity at the reporter locus, and thus can be applied to interrogate break resolution at active and repressed loci; and a second that reads out single-strand annealing in addition to gene targeting and gene disruption. Application of these reporters to assess repair pathway usage in several common gene editing contexts confirms the importance that chromatin status and initiation of end resection have on the resolution of nuclease-induced breaks.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Endodesoxirribonucleases , Genes Reporter , Citometria de Fluxo , Fluorescência , Inativação Gênica , Genes , Loci Gênicos , Genoma , Genômica/métodos , Células HEK293 , Humanos , Proteínas Luminescentes/genética , Transcrição Gênica
5.
Nat Methods ; 9(10): 973-5, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22941364

RESUMO

Targeted DNA double-strand breaks introduced by rare-cleaving designer endonucleases can be harnessed for gene disruption applications by engaging mutagenic nonhomologous end-joining DNA repair pathways. However, endonuclease-mediated DNA breaks are often subject to precise repair, which limits the efficiency of targeted genome editing. To address this issue, we coupled designer endonucleases to DNA end-processing enzymes to drive mutagenic break resolution, achieving up to 25-fold enhancements in gene disruption rates.


Assuntos
Quebras de DNA de Cadeia Dupla , Endonucleases/fisiologia , Animais , Reparo do DNA por Junção de Extremidades , Reparo do DNA , Exodesoxirribonucleases/fisiologia , Células HEK293 , Humanos , Camundongos , Fosfoproteínas/fisiologia , Receptores CCR5/fisiologia
6.
Nucleic Acids Res ; 39(21): e143, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21911364

RESUMO

A key challenge for the academic and biopharmaceutical communities is the rapid and scalable production of recombinant proteins for supporting downstream applications ranging from therapeutic trials to structural genomics efforts. Here, we describe a novel system for the production of recombinant mammalian proteins, including immune receptors, cytokines and antibodies, in a human cell line culture system, often requiring <3 weeks to achieve stable, high-level expression: Daedalus. The inclusion of minimized ubiquitous chromatin opening elements in the transduction vectors is key for preventing genomic silencing and maintaining the stability of decigram levels of expression. This system can bypass the tedious and time-consuming steps of conventional protein production methods by employing the secretion pathway of serum-free adapted human suspension cell lines, such as 293 Freestyle. Using optimized lentiviral vectors, yields of 20-100 mg/l of correctly folded and post-translationally modified, endotoxin-free protein of up to ~70 kDa in size, can be achieved in conventional, small-scale (100 ml) culture. At these yields, most proteins can be purified using a single size-exclusion chromatography step, immediately appropriate for use in structural, biophysical or therapeutic applications.


Assuntos
Lentivirus/genética , Proteínas Recombinantes/biossíntese , Proteínas de Fase Aguda/biossíntese , Proteínas de Fase Aguda/química , Proteínas de Fase Aguda/genética , Animais , Linhagem Celular , Cristalografia , Citocinas/genética , Citocinas/metabolismo , Técnicas Genéticas , Vetores Genéticos , Humanos , Lipocalina-2 , Lipocalinas/biossíntese , Lipocalinas/química , Lipocalinas/genética , Camundongos , Proteínas Oncogênicas/biossíntese , Proteínas Oncogênicas/química , Proteínas Oncogênicas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Transdução Genética
7.
J Clin Invest ; 130(10): 5127-5141, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32831296

RESUMO

Proteins created from recurrent fusion genes like CBFB-MYH11 are prevalent in acute myeloid leukemia (AML), often necessary for leukemogenesis, persistent throughout the disease course, and highly leukemia specific, making them attractive neoantigen targets for immunotherapy. A nonameric peptide derived from a prevalent CBFB-MYH11 fusion protein was found to be immunogenic in HLA-B*40:01+ donors. High-avidity CD8+ T cell clones isolated from healthy donors killed CBFB-MYH11+ HLA-B*40:01+ AML cell lines and primary human AML samples in vitro. CBFB-MYH11-specific T cells also controlled CBFB-MYH11+ HLA-B*40:01+ AML in vivo in a patient-derived murine xenograft model. High-avidity CBFB-MYH11 epitope-specific T cell receptors (TCRs) transduced into CD8+ T cells conferred antileukemic activity in vitro. Our data indicate that the CBFB-MYH11 fusion neoantigen is naturally presented on AML blasts and enables T cell recognition and killing of AML. We provide proof of principle for immunologically targeting AML-initiating fusions and demonstrate that targeting neoantigens has clinical relevance even in low-mutational frequency cancers like fusion-driven AML. This work also represents a first critical step toward the development of TCR T cell immunotherapy targeting fusion gene-driven AML.


Assuntos
Leucemia Mieloide Aguda , Animais , Carcinogênese , Subunidade beta de Fator de Ligação ao Core/genética , Humanos , Leucócitos , Camundongos , Mutação , Cadeias Pesadas de Miosina/genética , Proteínas de Fusão Oncogênica/genética
8.
Cancer J ; 25(3): 179-190, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31135525

RESUMO

Adoptive immunotherapy with engineered T cells is at the forefront of cancer treatment. T cells can be engineered to express T-cell receptors (TCRs) specific for tumor-associated antigens (TAAs) derived from intracellular or cell surface proteins. T cells engineered with TCRs (TCR-T) allow for targeting diverse types of TAAs, including proteins overexpressed in malignant cells, those with lineage-restricted expression, cancer-testis antigens, and neoantigens created from abnormal, malignancy-restricted proteins. Minor histocompatibility antigens can also serve as TAAs for TCR-T to treat relapsed hematologic malignancies after allogeneic hematopoietic cell transplantation. Moreover, TCR constructs can be modified to improve safety and enhance function and persistence of TCR-T. Transgenic T-cell receptor therapies targeting 3 different TAAs are in early-phase clinical trials for treatment of hematologic malignancies. Preclinical studies of TCR-T specific for many other TAAs are underway and offer great promise as safe and effective therapies for a wide range of cancers.


Assuntos
Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/terapia , Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/imunologia , Suscetibilidade a Doenças , Humanos , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética , Resultado do Tratamento
9.
Mol Cell Biol ; 33(6): 1149-63, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23297344

RESUMO

Antigen receptors activate pathways that control cell survival, proliferation, and differentiation. Two important targets of antigen receptors, NF-κB and Jun N-terminal kinase (JNK), are activated downstream of CARMA1, a scaffolding protein that nucleates a complex including BCL10, MALT1, and other IκB kinase (IKK)-signalosome components. Somatic mutations that constitutively activate CARMA1 occur frequently in diffuse large B cell lymphoma (DLBCL) and mediate essential survival signals. Mechanisms that downregulate this pathway might thus yield important therapeutic targets. Stimulation of antigen receptors induces not only BCL10 activation but also its degradation downstream of CARMA1, thereby ultimately limiting signals to its downstream targets. Here, using lymphocyte cell models, we identify a kinase-independent requirement for TAK1 and its adaptor, TAB1, in antigen receptor-induced BCL10 degradation. We show that TAK1 acts as an adaptor for E3 ubiquitin ligases that target BCL10 for degradation. Functionally, TAK1 overexpression restrains CARMA1-dependent activation of NF-κB by reducing BCL10 levels. TAK1 also promotes counterselection of NF-κB-addicted DLBCL lines by a dual mechanism involving kinase-independent degradation of BCL10 and kinase-dependent activation of JNK. Thus, by directly promoting BCL10 degradation, TAK1 counterbalances NF-κB and JNK signals essential for the activation and survival of lymphocytes and CARMA1-addicted lymphoma types.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , MAP Quinase Quinase Quinases/metabolismo , NF-kappa B/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linfócitos B/metabolismo , Linhagem Celular , Galinhas , Células HEK293 , Humanos , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase C/metabolismo , Proteína Quinase C beta , Proteólise , Receptores de Antígenos/metabolismo , Transdução de Sinais , Linfócitos T/metabolismo , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa