Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nature ; 615(7953): 697-704, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36890230

RESUMO

Neoantigens are peptides derived from non-synonymous mutations presented by human leukocyte antigens (HLAs), which are recognized by antitumour T cells1-14. The large HLA allele diversity and limiting clinical samples have restricted the study of the landscape of neoantigen-targeted T cell responses in patients over their treatment course. Here we applied recently developed technologies15-17 to capture neoantigen-specific T cells from blood and tumours from patients with metastatic melanoma with or without response to anti-programmed death receptor 1 (PD-1) immunotherapy. We generated personalized libraries of neoantigen-HLA capture reagents to single-cell isolate the T cells and clone their T cell receptors (neoTCRs). Multiple T cells with different neoTCR sequences (T cell clonotypes) recognized a limited number of mutations in samples from seven patients with long-lasting clinical responses. These neoTCR clonotypes were recurrently detected over time in the blood and tumour. Samples from four patients with no response to anti-PD-1 also demonstrated neoantigen-specific T cell responses in the blood and tumour to a restricted number of mutations with lower TCR polyclonality and were not recurrently detected in sequential samples. Reconstitution of the neoTCRs in donor T cells using non-viral CRISPR-Cas9 gene editing demonstrated specific recognition and cytotoxicity to patient-matched melanoma cell lines. Thus, effective anti-PD-1 immunotherapy is associated with the presence of polyclonal CD8+ T cells in the tumour and blood specific for a limited number of immunodominant mutations, which are recurrently recognized over time.


Assuntos
Antígenos de Neoplasias , Linfócitos T CD8-Positivos , Inibidores de Checkpoint Imunológico , Imunoterapia , Melanoma , Humanos , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/imunologia , Melanoma/patologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Antígenos HLA/imunologia , Metástase Neoplásica , Medicina de Precisão , Edição de Genes , Sistemas CRISPR-Cas , Mutação
2.
Nature ; 615(7953): 687-696, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36356599

RESUMO

T cell receptors (TCRs) enable T cells to specifically recognize mutations in cancer cells1-3. Here we developed a clinical-grade approach based on CRISPR-Cas9 non-viral precision genome-editing to simultaneously knockout the two endogenous TCR genes TRAC (which encodes TCRα) and TRBC (which encodes TCRß). We also inserted into the TRAC locus two chains of a neoantigen-specific TCR (neoTCR) isolated from circulating T cells of patients. The neoTCRs were isolated using a personalized library of soluble predicted neoantigen-HLA capture reagents. Sixteen patients with different refractory solid cancers received up to three distinct neoTCR transgenic cell products. Each product expressed a patient-specific neoTCR and was administered in a cell-dose-escalation, first-in-human phase I clinical trial ( NCT03970382 ). One patient had grade 1 cytokine release syndrome and one patient had grade 3 encephalitis. All participants had the expected side effects from the lymphodepleting chemotherapy. Five patients had stable disease and the other eleven had disease progression as the best response on the therapy. neoTCR transgenic T cells were detected in tumour biopsy samples after infusion at frequencies higher than the native TCRs before infusion. This study demonstrates the feasibility of isolating and cloning multiple TCRs that recognize mutational neoantigens. Moreover, simultaneous knockout of the endogenous TCR and knock-in of neoTCRs using single-step, non-viral precision genome-editing are achieved. The manufacture of neoTCR engineered T cells at clinical grade, the safety of infusing up to three gene-edited neoTCR T cell products and the ability of the transgenic T cells to traffic to the tumours of patients are also demonstrated.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Edição de Genes , Neoplasias , Medicina de Precisão , Receptores de Antígenos de Linfócitos T , Linfócitos T , Transgenes , Humanos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Biópsia , Terapia Baseada em Transplante de Células e Tecidos/efeitos adversos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Síndrome da Liberação de Citocina/complicações , Progressão da Doença , Encefalite/complicações , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Genes Codificadores da Cadeia alfa de Receptores de Linfócitos T , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Mutação , Neoplasias/complicações , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/terapia , Segurança do Paciente , Medicina de Precisão/efeitos adversos , Medicina de Precisão/métodos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transgenes/genética , Antígenos HLA/imunologia , Sistemas CRISPR-Cas
3.
Cell Rep ; 28(10): 2728-2738.e7, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31484081

RESUMO

Neoantigen-specific T cells are increasingly viewed as important immunotherapy effectors, but physically isolating these rare cell populations is challenging. Here, we describe a sensitive method for the enumeration and isolation of neoantigen-specific CD8+ T cells from small samples of patient tumor or blood. The method relies on magnetic nanoparticles that present neoantigen-loaded major histocompatibility complex (MHC) tetramers at high avidity by barcoded DNA linkers. The magnetic particles provide a convenient handle to isolate the desired cell populations, and the barcoded DNA enables multiplexed analysis. The method exhibits superior recovery of antigen-specific T cell populations relative to literature approaches. We applied the method to profile neoantigen-specific T cell populations in the tumor and blood of patients with metastatic melanoma over the course of anti-PD1 checkpoint inhibitor therapy. We show that the method has value for monitoring clinical responses to cancer immunotherapy and might help guide the development of personalized mutational neoantigen-specific T cell therapies and cancer vaccines.


Assuntos
Antígenos de Neoplasias/sangue , Melanoma/sangue , Melanoma/imunologia , Linfócitos T/imunologia , Biópsia , Células HEK293 , Humanos , Imunoterapia , Células Jurkat , Cinética , Linfócitos do Interstício Tumoral/imunologia , Nanopartículas de Magnetita/química , Complexo Principal de Histocompatibilidade , Melanoma/patologia , Melanoma/secundário , Ácidos Nucleicos/metabolismo , Receptor de Morte Celular Programada 1/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Reprodutibilidade dos Testes , Tomografia Computadorizada por Raios X
4.
Nat Methods ; 16(2): 191-198, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30700902

RESUMO

CD8+ T cells recognize and eliminate tumors in an antigen-specific manner. Despite progress in characterizing the antitumor T cell repertoire and function, the identification of target antigens remains a challenge. Here we describe the use of chimeric receptors called signaling and antigen-presenting bifunctional receptors (SABRs) in a cell-based platform for T cell receptor (TCR) antigen discovery. SABRs present an extracellular complex comprising a peptide and major histocompatibility complex (MHC), and induce intracellular signaling via a TCR-like signal after binding with a cognate TCR. We devised a strategy for antigen discovery using SABR libraries to screen thousands of antigenic epitopes. We validated this platform by identifying the targets recognized by public TCRs of known specificities. Moreover, we extended this approach for personalized neoantigen discovery.


Assuntos
Apresentação de Antígeno , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Células Apresentadoras de Antígenos/citologia , Antígenos/química , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Linfócitos T CD8-Positivos/citologia , Clonagem Molecular , Técnicas de Cocultura , Epitopos/química , Reações Falso-Positivas , Biblioteca Gênica , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Imunoterapia/métodos , Células Jurkat , Células K562 , Lectinas Tipo C/metabolismo , Complexo Principal de Histocompatibilidade , Oligonucleotídeos/genética , Peptídeos/química
5.
Nat Methods ; 16(2): 183-190, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30700903

RESUMO

T cell receptor (TCR) ligand discovery is essential for understanding and manipulating immune responses to tumors. We developed a cell-based selection platform for TCR ligand discovery that exploits a membrane transfer phenomenon called trogocytosis. We discovered that T cell membrane proteins are transferred specifically to target cells that present cognate peptide-major histocompatibility complex (MHC) molecules. Co-incubation of T cells expressing an orphan TCR with target cells collectively presenting a library of peptide-MHCs led to specific labeling of cognate target cells, enabling isolation of these target cells and sequencing of the cognate TCR ligand. We validated this method for two clinically employed TCRs and further used the platform to identify the cognate neoepitope for a subject-derived neoantigen-specific TCR. Thus, target cell trogocytosis is a robust tool for TCR ligand discovery that will be useful for studying basic tumor immunology and identifying new targets for immunotherapy.


Assuntos
Antígenos/química , Técnicas Genéticas , Receptores de Antígenos de Linfócitos T/química , Linfócitos T/citologia , Imunidade Adaptativa , Animais , Biotinilação , DNA/análise , Epitopos/química , Biblioteca Gênica , Células HEK293 , Humanos , Imunoterapia , Células Jurkat , Células K562 , Ligantes , Camundongos , Peptídeos/química , Fagocitose , Linfócitos T/imunologia
6.
Proc Natl Acad Sci U S A ; 115(45): E10702-E10711, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30348802

RESUMO

Tumor-specific T cell receptor (TCR) gene transfer enables specific and potent immune targeting of tumor antigens. Due to the prevalence of the HLA-A2 MHC class I supertype in most human populations, the majority of TCR gene therapy trials targeting public antigens have employed HLA-A2-restricted TCRs, limiting this approach to those patients expressing this allele. For these patients, TCR gene therapy trials have resulted in both tantalizing successes and lethal adverse events, underscoring the need for careful selection of antigenic targets. Broad and safe application of public antigen-targeted TCR gene therapies will require (i) selecting public antigens that are highly tumor-specific and (ii) targeting multiple epitopes derived from these antigens by obtaining an assortment of TCRs restricted by multiple common MHC alleles. The canonical cancer-testis antigen, NY-ESO-1, is not expressed in normal tissues but is aberrantly expressed across a broad array of cancer types. It has also been targeted with A2-restricted TCR gene therapy without adverse events or notable side effects. To enable the targeting of NY-ESO-1 in a broader array of HLA haplotypes, we isolated TCRs specific for NY-ESO-1 epitopes presented by four MHC molecules: HLA-A2, -B07, -B18, and -C03. Using these TCRs, we pilot an approach to extend TCR gene therapies targeting NY-ESO-1 to patient populations beyond those expressing HLA-A2.


Assuntos
Proteínas de Homeodomínio/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Receptores de Antígenos de Linfócitos T/isolamento & purificação , Receptores de Antígenos de Linfócitos T/metabolismo , Animais , Clonagem Molecular , Humanos
7.
PLoS One ; 13(1): e0191634, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29360859

RESUMO

For adoptive cell transfer (ACT) immunotherapy of tumor-reactive T cells, an effective therapeutic outcome depends upon cell dose, cell expansion in vivo through a minimally differentiated phenotype, long term persistence, and strong cytolytic effector function. An incomplete understanding of the biological coupling between T cell expansion, differentiation, and response to stimulation hinders the co-optimization of these factors. We report on a biophysical investigation of how the short-term kinetics of T cell functional activation, through molecular stimulation and cell-cell interactions, competes with phenotype differentiation. T cells receive molecular stimulation for a few minutes to a few hours in bulk culture. Following this priming period, the cells are then analyzed at the transcriptional level, or isolated as single cells, with continuing molecular stimulation, within microchambers for analysis via 11-plex secreted protein assays. We resolve a rapid feedback mechanism, promoted by T cell-T cell contact interactions, which strongly amplifies T cell functional performance while yielding only minimal phenotype differentiation. When tested in mouse models of ACT, optimally primed T cells lead to complete tumor eradication. A similar kinetic process is identified in CD8+ and CD4+ T cells collected from a patient with metastatic melanoma.


Assuntos
Transferência Adotiva , Imunofenotipagem , Neoplasias/terapia , Linfócitos T/imunologia , Animais , Feminino , Citometria de Fluxo , Xenoenxertos , Humanos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
8.
Cell ; 172(3): 549-563.e16, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29275860

RESUMO

The immune system can mount T cell responses against tumors; however, the antigen specificities of tumor-infiltrating lymphocytes (TILs) are not well understood. We used yeast-display libraries of peptide-human leukocyte antigen (pHLA) to screen for antigens of "orphan" T cell receptors (TCRs) expressed on TILs from human colorectal adenocarcinoma. Four TIL-derived TCRs exhibited strong selection for peptides presented in a highly diverse pHLA-A∗02:01 library. Three of the TIL TCRs were specific for non-mutated self-antigens, two of which were present in separate patient tumors, and shared specificity for a non-mutated self-antigen derived from U2AF2. These results show that the exposed recognition surface of MHC-bound peptides accessible to the TCR contains sufficient structural information to enable the reconstruction of sequences of peptide targets for pathogenic TCRs of unknown specificity. This finding underscores the surprising specificity of TCRs for their cognate antigens and enables the facile indentification of tumor antigens through unbiased screening.


Assuntos
Adenocarcinoma/imunologia , Antígenos de Neoplasias/imunologia , Neoplasias Colorretais/imunologia , Linfócitos do Interstício Tumoral/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Idoso , Animais , Antígenos de Neoplasias/química , Linhagem Celular Tumoral , Células Cultivadas , Células HEK293 , Antígenos HLA-A/química , Antígenos HLA-A/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Biblioteca de Peptídeos , Células Sf9 , Spodoptera
9.
Curr Opin Biotechnol ; 48: 142-152, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28494274

RESUMO

Immunotherapies are yielding effective treatments for several previously untreatable cancers. Until recently, vaccines and adoptive cell therapies have been designed to target public tumor antigens common to multiple patients rather than private antigens specific to a single patient. Due to the difficulty of identifying public antigens that are expressed exclusively on tumor cells, these studies have yielded both clinical successes and serious immune-related adverse events. Multiple avenues of research now underscore the centrality of tumor-specific mutated private antigens to endogenous anti-tumor immunity. Immunotherapies that target these neoantigens may enable safer and more durable tumor regression, but personalized targeting presents a number of challenges. Foremost among these is to develop processes that accelerate advancement from neoantigen discovery to use of these neoantigens as vaccines or as targets for adoptive cell therapies. Exome sequencing has facilitated discovery of neoantigens for melanoma and other highly mutated cancers. New technologies - possibly proceeding from T cell receptor repertoire sequencing - are needed to identify antigens for cancers with low mutational burden and few neoantigens. In this review, we discuss progress toward personalizing T cell-mediated immunotherapy for cancer as well as challenges going forward.


Assuntos
Antígenos de Neoplasias/imunologia , Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T/imunologia , Humanos
10.
Nat Methods ; 14(5): 521-530, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28369043

RESUMO

Studies of human T cell development require robust model systems that recapitulate the full span of thymopoiesis, from hematopoietic stem and progenitor cells (HSPCs) through to mature T cells. Existing in vitro models induce T cell commitment from human HSPCs; however, differentiation into mature CD3+TCR-αß+ single-positive CD8+ or CD4+ cells is limited. We describe here a serum-free, artificial thymic organoid (ATO) system that supports efficient and reproducible in vitro differentiation and positive selection of conventional human T cells from all sources of HSPCs. ATO-derived T cells exhibited mature naive phenotypes, a diverse T cell receptor (TCR) repertoire and TCR-dependent function. ATOs initiated with TCR-engineered HSPCs produced T cells with antigen-specific cytotoxicity and near-complete lack of endogenous TCR Vß expression, consistent with allelic exclusion of Vß-encoding loci. ATOs provide a robust tool for studying human T cell differentiation and for the future development of stem-cell-based engineered T cell therapies.


Assuntos
Órgãos Artificiais , Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Organoides/citologia , Linfócitos T/citologia , Timo/citologia , Biotecnologia/métodos , Células-Tronco Hematopoéticas/imunologia , Humanos , Organoides/imunologia , Células-Tronco/citologia , Células-Tronco/imunologia , Linfócitos T/imunologia , Timo/imunologia
11.
Biotechniques ; 62(3): 123-130, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28298179

RESUMO

Peptide-major histocompatibility complex (pMHC) multimers enable the detection, characterization, and isolation of antigen-specific T-cell subsets at the single-cell level via flow cytometry and fluorescence microscopy. These labeling reagents exploit a multivalent scaffold to increase the avidity of individually weak T-cell receptor (TCR)-pMHC interactions. Dextramers are an improvement over the original streptavidin-based tetramer technology because they are more multivalent, improving sensitivity for rare, low-avidity T cells, including self/tumor-reactive clones. However, commercial pMHC dextramers are expensive, and in-house production is very involved for a typical biology research laboratory. Here, we present a simple, inexpensive protocol for preparing pMHC dextramers by doping in biotinylated dextran during conventional tetramer preparation. We use these pMHC dextramers to identify patient-derived, tumor-reactive T cells. We apply the same dextran doping technique to prepare TCR dextramers and use these novel reagents to yield new insight into MHC I-mediated antigen presentation.


Assuntos
Biotecnologia/métodos , Dextranos/metabolismo , Antígenos de Histocompatibilidade/metabolismo , Peptídeos/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Biotina , Dextranos/química , Citometria de Fluxo , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Células HEK293 , Antígenos de Histocompatibilidade/química , Antígenos de Histocompatibilidade/genética , Humanos , Células Jurkat , Células K562 , Peptídeos/química , Peptídeos/genética , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Estreptavidina , Transdução Genética
12.
Elife ; 52016 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-27823582

RESUMO

T cells engineered to express a tumor-specific αß T cell receptor (TCR) mediate anti-tumor immunity. However, mispairing of the therapeutic αß chains with endogenous αß chains reduces therapeutic TCR surface expression and generates self-reactive TCRs. We report a general strategy to prevent TCR mispairing: swapping constant domains between the α and ß chains of a therapeutic TCR. When paired, domain-swapped (ds)TCRs assemble with CD3, express on the cell surface, and mediate antigen-specific T cell responses. By contrast, dsTCR chains mispaired with endogenous chains cannot properly assemble with CD3 or signal, preventing autoimmunity. We validate this approach in cell-based assays and in a mouse model of TCR gene transfer-induced graft-versus-host disease. We also validate a related approach whereby replacement of αß TCR domains with corresponding γδ TCR domains yields a functional TCR that does not mispair. This work enables the design of safer TCR gene therapies for cancer immunotherapy.


Assuntos
Genes Codificadores dos Receptores de Linfócitos T , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Animais , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro , Camundongos , Domínios Proteicos , Recombinação Genética
13.
Chem Biol ; 16(8): 868-81, 2009 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-19716477

RESUMO

New tools are needed for managing celiac sprue, a lifelong immune disease of the small intestine. Ongoing drug trials are also prompting a search for noninvasive biomarkers of gluten-induced intestinal change. We have synthesized and characterized noninflammatory gluten peptide analogs in which key Gln residues are replaced by Asn or His. Like their proinflammatory counterparts, these biomarkers are resistant to gastrointestinal proteases, susceptible to glutenases, and permeable across enterocyte barriers. Unlike gluten peptides, however, they are not appreciably recognized by transglutaminase, HLA-DQ2, or disease-specific T cells. In vitro and animal studies show that the biomarkers can detect intestinal permeability changes as well as glutenase-catalyzed gastric detoxification of gluten. Accordingly, controlled clinical studies are warranted to evaluate the use of these peptides as probes for abnormal intestinal permeability in celiac patients and for glutenase efficacy in clinical trials and practice.


Assuntos
Doença Celíaca/metabolismo , Glutens/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Biomarcadores/metabolismo , Doença Celíaca/imunologia , Glutens/química , Glutens/farmacologia , Humanos , Marcação por Isótopo , Masculino , Dados de Sequência Molecular , Peptídeos/química , Ratos , Ratos Wistar , Linfócitos T/imunologia , Transglutaminases/imunologia , Transglutaminases/metabolismo
14.
PLoS One ; 4(7): e6313, 2009 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-19621078

RESUMO

BACKGROUND AND AIMS: Celiac sprue is a life-long disease characterized by an intestinal inflammatory response to dietary gluten. A gluten-free diet is an effective treatment for most patients, but accidental ingestion of gluten is common, leading to incomplete recovery or relapse. Food-grade proteases capable of detoxifying moderate quantities of dietary gluten could mitigate this problem. METHODS: We evaluated the gluten detoxification properties of two food-grade enzymes, aspergillopepsin (ASP) from Aspergillus niger and dipeptidyl peptidase IV (DPPIV) from Aspergillus oryzae. The ability of each enzyme to hydrolyze gluten was tested against synthetic gluten peptides, a recombinant gluten protein, and simulated gastric digests of whole gluten and whole-wheat bread. Reaction products were analyzed by mass spectrometry, HPLC, ELISA with a monoclonal antibody that recognizes an immunodominant gluten epitope, and a T cell proliferation assay. RESULTS: ASP markedly enhanced gluten digestion relative to pepsin, and cleaved recombinant alpha2-gliadin at multiple sites in a non-specific manner. When used alone, neither ASP nor DPPIV efficiently cleaved synthetic immunotoxic gluten peptides. This lack of specificity for gluten was especially evident in the presence of casein, a competing dietary protein. However, supplementation of ASP with DPPIV enabled detoxification of moderate amounts of gluten in the presence of excess casein and in whole-wheat bread. ASP was also effective at enhancing the gluten-detoxifying efficacy of cysteine endoprotease EP-B2 under simulated gastric conditions. CONCLUSIONS: Clinical studies are warranted to evaluate whether a fixed dose ratio combination of ASP and DPPIV can provide near-term relief for celiac patients suffering from inadvertent gluten exposure. Due to its markedly greater hydrolytic activity against gluten than endogenous pepsin, food-grade ASP may also augment the activity of therapeutically relevant doses of glutenases such as EP-B2 and certain prolyl endopeptidases.


Assuntos
Dipeptidil Peptidase 4/metabolismo , Alimentos , Glutens/metabolismo , Sequência de Aminoácidos , Cromatografia Líquida de Alta Pressão , Ensaio de Imunoadsorção Enzimática , Glutens/química , Hidrólise , Espectrometria de Massas , Dados de Sequência Molecular
15.
J Pharmacol Exp Ther ; 329(2): 657-68, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19218531

RESUMO

Celiac sprue is a T-cell-mediated enteropathy elicited in genetically susceptible individuals by dietary gluten proteins. To initiate and propagate inflammation, proteolytically resistant gluten peptides must be translocated across the small intestinal epithelium and presented to DQ2-restricted T cells, but the effectors enabling this translocation under normal and inflammatory conditions are not well understood. We demonstrate that a fluorescently labeled antigenic 33-mer gluten peptide is translocated intact across a T84 cultured epithelial cell monolayer and that preincubation of the monolayer with media from gluten-stimulated, celiac patient-derived intestinal T cells enhances the apical-to-basolateral flux of this peptide in a dose-dependent, saturable manner. The permeability-enhancing activity of activated T-cell media is inhibited by blocking antibodies against either interferon-gamma or its receptor and is recapitulated using recombinant interferon-gamma. At saturating levels of interferon-gamma, activated T-cell media does not further increase transepithelial peptide flux, indicating the primacy of interferon-gamma as an effector of increased epithelial permeability during inflammation. Reducing the assay temperature to 4 degrees C reverses the effect of interferon-gamma but does not reduce basal peptide flux occurring in the absence of interferon-gamma, suggesting active transcellular transport of intact peptides is increased during inflammation. A panel of disease-relevant gluten peptides exhibited an inverse correlation between size and transepithelial flux but no apparent sequence constraints. Anti-interferon-gamma therapy may mitigate the vicious cycle of gluten-induced interferon-gamma secretion and interferon-gamma-mediated enhancement of gluten peptide flux but is unlikely to prevent translocation of gluten peptides in the absence of inflammatory conditions.


Assuntos
Doença Celíaca/imunologia , Glutens/farmacologia , Interferon gama/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Linfócitos T/efeitos dos fármacos , Transporte Biológico , Western Blotting , Doença Celíaca/metabolismo , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Glutens/farmacocinética , Humanos , Interferon gama/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Fragmentos de Peptídeos/farmacocinética , Permeabilidade , Linfócitos T/imunologia , Linfócitos T/metabolismo
16.
PLoS Pathog ; 4(2): e34, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18425213

RESUMO

Pathogens are exogenous agents capable of causing disease in susceptible organisms. In celiac sprue, a disease triggered by partially hydrolyzed gluten peptides in the small intestine, the offending immunotoxins cannot replicate, but otherwise have many hallmarks of classical pathogens. First, dietary gluten and its peptide metabolites are ubiquitous components of the modern diet, yet only a small, genetically susceptible fraction of the human population contracts celiac sprue. Second, immunotoxic gluten peptides have certain unusual structural features that allow them to survive the harsh proteolytic conditions of the gastrointestinal tract and thereby interact extensively with the mucosal lining of the small intestine. Third, they invade across epithelial barriers intact to access the underlying gut-associated lymphoid tissue. Fourth, they possess recognition sequences for selective modification by an endogenous enzyme, transglutaminase 2, allowing for in situ activation to a more immunotoxic form via host subversion. Fifth, they precipitate a T cell-mediated immune reaction comprising both innate and adaptive responses that causes chronic inflammation of the small intestine. Sixth, complete elimination of immunotoxic gluten peptides from the celiac diet results in remission, whereas reintroduction of gluten in the diet causes relapse. Therefore, in analogy with antibiotics, orally administered proteases that reduce the host's exposure to the immunotoxin by accelerating gluten peptide destruction have considerable therapeutic potential. Last but not least, notwithstanding the power of in vitro methods to reconstitute the essence of the immune response to gluten in a celiac patient, animal models for the disease, while elusive, are likely to yield fundamentally new systems-level insights.


Assuntos
Doença Celíaca/imunologia , Glutens/imunologia , Imunidade Inata , Imunotoxinas/imunologia , Adaptação Fisiológica , Animais , Sequência de Bases , Doença Celíaca/genética , Doença Celíaca/terapia , Modelos Animais de Doenças , Predisposição Genética para Doença , Glutens/metabolismo , Humanos , Imunidade Inata/fisiologia , Imunotoxinas/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Dados de Sequência Molecular , Peptídeos/efeitos adversos , Peptídeos/imunologia , Peptídeos/metabolismo
17.
J Pharmacol Exp Ther ; 318(3): 1178-86, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16757540

RESUMO

Celiac Sprue is a multifactorial disease characterized by an intestinal inflammatory response to ingested gluten. Proteolytically resistant gluten peptides from wheat, rye, and barley persist in the intestinal lumen and elicit an immune response in genetically susceptible individuals. Here, we demonstrate the in vivo ability of a gluten-digesting protease ("glutenase") to accelerate the breakdown of a gluten-rich solid meal. The proenzyme form of endoprotease B, isoform 2 from Hordeum vulgare (EP-B2), was orally administered to adult rats with a solid meal containing 1 g of gluten. Gluten digestion in the stomach and small intestine was monitored as a function of enzyme dose and time by high-performance liquid chromatography and mass spectrometry. In the absence of supplementary EP-B2, gluten was solubilized and proteolyzed to a limited extent in the stomach and was hydrolyzed and assimilated mostly in the small intestine. In contrast, EP-B2 was remarkably effective at digesting gluten in the rat stomach in a dose- and time-dependent fashion. At a 1:25 EP-B2/gluten dose, the gastric concentration of the highly immunogenic 33-mer gliadin peptide was reduced by more than 50-fold within 90 min with no overt signs of toxicity. Evaluation of EP-B2 as an adjunct to diet control is therefore warranted in celiac patients.


Assuntos
Cisteína Endopeptidases/farmacologia , Digestão , Glutens/metabolismo , Hordeum/enzimologia , Sequência de Aminoácidos , Animais , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Trato Gastrointestinal/metabolismo , Masculino , Dados de Sequência Molecular , Ratos , Ratos Wistar , Reprodutibilidade dos Testes
18.
Chem Biol ; 13(6): 637-47, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16793521

RESUMO

We describe the heterologous expression in Escherichia coli of the proenzyme precursor to EP-B2, a cysteine endoprotease from germinating barley seeds. High yields (50 mg/l) of recombinant proEP-B2 were obtained from E. coli inclusion bodies in shake flask cultures following purification and refolding. The zymogen was rapidly autoactivated to its mature form under acidic conditions at a rate independent of proEP-B2 concentration, suggesting a cis mechanism of autoactivation. Mature EP-B2 was stable and active over a wide pH range and efficiently hydrolyzed a recombinant wheat gluten protein, alpha2-gliadin, at sequences with known immunotoxicity in celiac sprue patients. The X-ray crystal structure of mature EP-B2 bound to leupeptin was solved to 2.2 A resolution and provided atomic insights into the observed subsite specificity of the endoprotease. Our findings suggest that orally administered proEP-B2 may be especially well suited for treatment of celiac sprue.


Assuntos
Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Hordeum/enzimologia , Dobramento de Proteína , Sequência de Aminoácidos , Cristalografia por Raios X , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/isolamento & purificação , Ativação Enzimática , Escherichia coli , Expressão Gênica , Glutens/metabolismo , Hordeum/genética , Concentração de Íons de Hidrogênio , Cinética , Leupeptinas/química , Leupeptinas/metabolismo , Dados de Sequência Molecular , Pepsina A/metabolismo , Precursores de Proteínas/química , Precursores de Proteínas/genética , Precursores de Proteínas/isolamento & purificação , Precursores de Proteínas/metabolismo , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato , Tripsina/metabolismo
19.
Chem Biol ; 13(6): 649-58, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16793522

RESUMO

Celiac sprue (also known as celiac disease) is an inheritable, gluten-induced enteropathy of the upper small intestine with an estimated prevalence of 0.5%-1% in most parts of the world. The ubiquitous nature of food gluten, coupled with inadequate labeling regulations in most countries, constantly poses a threat of disease exacerbation and relapse for patients. Here, we demonstrate that a two-enzyme cocktail comprised of a glutamine-specific cysteine protease (EP-B2) that functions under gastric conditions and a PEP, which acts in concert with pancreatic proteases under duodenal conditions, is a particularly potent candidate for celiac sprue therapy. At a gluten:EP-B2:PEP weight ratio of 75:3:1, grocery store gluten is fully detoxified within 10 min of simulated duodenal conditions, as judged by chromatographic analysis, biopsy-derived T cell proliferation assays, and a commercial antigluten antibody test.


Assuntos
Doença Celíaca/enzimologia , Doença Celíaca/terapia , Cisteína Endopeptidases/metabolismo , Sequência de Aminoácidos , Doença Celíaca/imunologia , Doença Celíaca/patologia , Linhagem Celular , Proliferação de Células , Separação Celular , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/farmacologia , Epitopos/imunologia , Flavobacterium/enzimologia , Flavobacterium/genética , Glutens/química , Glutens/imunologia , Glutens/metabolismo , Humanos , Inativação Metabólica , Cinética , Dados de Sequência Molecular , Myxococcus xanthus/enzimologia , Myxococcus xanthus/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA