Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Biol Aujourdhui ; 211(3): 229-231, 2017.
Artigo em Francês | MEDLINE | ID: mdl-29412133

RESUMO

Antigen receptors, which form the base of the adaptive immune system, are created stochastically by a DNA editing process called V(D)J recombination. As high-throughput sequencing enables to study the repertoire of these receptors, it is now possible to learn the probabilistic laws of this random process, and to use them to analyse receptors of interest, generate synthetic repertoires to create controls, or aid the identification of receptors that are specific to diseases, with possible applications for medical diagnostics. This article describes how these tasks can be performed using the IGoR software, which can learn statistical models from data, annotate existing sequences, or generate new synthetic ones with the same laws as the recombination process.


Assuntos
Biologia Computacional , Engenharia de Proteínas/métodos , Receptores de Antígenos/biossíntese , Receptores de Antígenos/química , Treinamento por Simulação/métodos , Software , Animais , Biologia Computacional/educação , Biologia Computacional/métodos , Simulação por Computador , Bases de Dados de Proteínas , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Aprendizagem , Biologia Sintética/educação , Biologia Sintética/métodos , Recombinação V(D)J/fisiologia
2.
EMBO Mol Med ; 8(7): 702-11, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27189167

RESUMO

Chimeric antigen receptor (CAR) T-cell therapy is a new successful treatment for refractory B-cell leukemia. Successful therapeutic outcome depends on long-term expression of CAR transgene in T cells, which is achieved by delivering transgene using integrating gamma retrovirus (RV) or lentivirus (LV). However, uncontrolled RV/LV integration in host cell genomes has the potential risk of causing insertional mutagenesis. Herein, we describe a novel episomal long-term cell engineering method using non-integrating lentiviral (NILV) vector containing a scaffold/matrix attachment region (S/MAR) element, for either expression of transgenes or silencing of target genes. The insertional events of this vector into the genome of host cells are below detection level. CD19 CAR T cells engineered with a NILV-S/MAR vector have similar levels of CAR expression as T cells engineered with an integrating LV vector, even after numerous rounds of cell division. NILV-S/MAR-engineered CD19 CAR T cells exhibited similar cytotoxic capacity upon CD19(+) target cell recognition as LV-engineered T cells and are as effective in controlling tumor growth in vivo We propose that NILV-S/MAR vectors are superior to current options as they enable long-term transgene expression without the risk of insertional mutagenesis and genotoxicity.


Assuntos
Transferência Adotiva/métodos , Engenharia Celular/métodos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Neoplasias/terapia , Plasmídeos , Linfócitos T/imunologia , Transgenes , Animais , Antígenos CD19/imunologia , Células Cultivadas , Modelos Animais de Doenças , Expressão Gênica , Vetores Genéticos , Instabilidade Genômica , Humanos , Lentivirus/genética , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores de Antígenos/biossíntese , Receptores de Antígenos/genética , Resultado do Tratamento
3.
Methods Mol Biol ; 1393: 75-86, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27033217

RESUMO

The adoptive transfer of T lymphocytes expressing chimeric antigen receptors (CARs) has emerged as a promising treatment for various lymphoid and solid malignancies. Patients treated with CAR-T cells have achieved dramatic responses and in some cases, complete tumor eradication. Given that CARs combine the specificity of a monoclonal antibody with the internal signaling domains of T cells, there is flexibility for choice of target antigen and strength of T-cell activation. This targeting mechanism also relinquishes the need for antigen processing and presentation via the major histocompatibility complex (MHC), making CARs a very attractive therapeutic option for the majority of patients. This review describes current methodological strategies used to generate CAR molecules, how to insert these molecules in T lymphocytes and how to evaluate the functionality of these CAR-T cells using various in vitro and in vivo experiments.


Assuntos
Transferência Adotiva , Neoplasias/terapia , Receptores de Antígenos/biossíntese , Linfócitos T/imunologia , Animais , Técnicas de Cocultura , Células HEK293 , Humanos , Células K562 , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Receptores de Antígenos/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética
4.
J Mol Med (Berl) ; 94(1): 83-93, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26300042

RESUMO

UNLABELLED: Natural killer (NK) cells play an important role in tumor immunotherapy with their unique capability of killing transformed cells without the need for prior sensitization and without major histocompatibility complex (MHC)/peptide restriction. However, tumor cells can escape NK cell cytotoxicity by various tumor immune escape mechanisms. To overcome these escape mechanisms, NK cells can be modified to express chimeric antigen receptors (CARs), enhancing their tumor-specific cytotoxicity. To determine the most efficacious method to modify human NK cells, we compared different retroviral vector systems, retroviral pseudotypes, and transduction protocols. Using optimized transduction conditions, the highest transduction levels (up to 60%) were achieved with alpharetroviral vectors. Alpharetroviral-modified primary human NK cells exhibited no alteration in receptor expression and had similar degranulation activity as untransduced NK cells, thus demonstrating that alpharetroviral modification did not negatively affect NK cell cytotoxicity. Transduction of NK cells with an alpharetroviral vector containing a CD19 CAR expression cassette selectively enhanced NK cell cytotoxicity towards CD19-expressing leukemia cells, achieving nearly complete elimination of leukemia cells after 48 h. Taken together, alpharetroviral vectors are promising tools for NK cell-mediated cancer immunotherapy applications. KEY MESSAGES: Efficient modification of human NK cells using alpharetroviral vectors. Anti-CD19-CAR-NK cells exhibited improved cytotoxicity towards CD19(+) leukemia cells. Alpharetroviral vectors are promising tools for immunotherapy applications using NK cells.


Assuntos
Alpharetrovirus/genética , Antígenos CD19/genética , Citotoxicidade Imunológica/genética , Vetores Genéticos/genética , Células Matadoras Naturais/imunologia , Leucemia/terapia , Receptores de Antígenos/genética , Linhagem Celular Tumoral , Citotoxicidade Imunológica/imunologia , Terapia Genética/métodos , Proteínas de Fluorescência Verde/genética , Humanos , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/citologia , Leucemia/imunologia , Receptores de Antígenos/biossíntese , Receptores de Antígenos/imunologia , Transdução Genética/métodos , Evasão Tumoral/imunologia
5.
Cancer Immunol Immunother ; 64(5): 621-34, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25711293

RESUMO

The disialoganglioside GD2 is a well-established target antigen for passive immunotherapy in neuroblastoma (NB). Despite the recent success of passive immunotherapy with the anti-GD2 antibody ch14.18 and cytokines, treatment of high-risk NB remains challenging. We expanded the approach of GD2-specific, antibody-based immunotherapy to an application of a GD2-specific natural killer (NK) cell line, NK-92-scFv(ch14.18)-zeta. NK-92-scFv(ch14.18)-zeta is genetically engineered to express a GD2-specific chimeric antigen receptor generated from ch14.18. Here, we show that chimeric receptor expression enables NK-92-scFv(ch14.18)-zeta to effectively lyse GD2(+) NB cells also including partially or multidrug-resistant lines. Our data suggest that recognition of GD2 by the chimeric receptor is the primary mechanism involved in NK-92-scFv(ch14.18)-zeta-mediated lysis and is independent of activating NK cell receptor/ligand interactions. Furthermore, we demonstrate that NK-92-scFv(ch14.18)-zeta is able to mediate a significant anti-tumor response in vivo in a drug-resistant GD2(+) NB xenograft mouse model. NK-92-scFv(ch14.18)-zeta is an NB-specific NK cell line that has potential for future clinical development due to its high stability and activity toward GD2(+) NB cell lines.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Gangliosídeos/imunologia , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/imunologia , Neuroblastoma/terapia , Animais , Anticorpos Anti-Idiotípicos/imunologia , Linhagem Celular , Citotoxicidade Imunológica/imunologia , Feminino , Gangliosídeos/genética , Engenharia Genética , Humanos , Camundongos , Camundongos Endogâmicos NOD , Transplante de Neoplasias , Neuroblastoma/imunologia , Receptores de Antígenos/biossíntese , Receptores de Antígenos/imunologia , Anticorpos de Cadeia Única/imunologia
6.
Dev Comp Immunol ; 46(2): 267-78, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24844613

RESUMO

Nonspecific cytotoxic cells (NCCs) are an important cytotoxic cell population in the innate teleost immune system. The receptor designated "NCC receptor protein 1" (NCCRP-1) has been reported to be involved in the recognition and activation of NCCs. In this study, the full-length cDNA of Epinephelus coioides NCCRP-1 (ecnccrp-1) was cloned. The open reading frame (ORF) of ecnccrp-1 is 699 bp, encoding a 232 amino acid protein that includes proline-rich motifs at the N-terminus and is related to the F-box associated family. Although a bioinformatics analysis showed that EcNCCRP-1 had no signal peptide or transmembrane helices, a polyclonal antibody directed against recombinant EcNCCRP-1 efficiently labeled a membrane protein in the head kidney, detected with Western blot analysis, which indicated that the protein localized to the cell surface. RT-PCR showed that the constitutive expression of ecnccrp-1 was higher in the lymphoid organs, such as the trunk kidney, spleen, head kidney, and thymus, and lower in brain, heart, fat, liver, muscle, and skin. After infection with Cryptocaryon irritans, the transcription of ecnccrp-1 was analyzed at the infected sites (skin and gills) and in the systemic immune organs (head kidney and spleen). At the infected sites, especially the skin, ecnccrp-1 expression was upregulated at 6h post infection, reaching peak expression on day 3 post the primary infection. However, the expression patterns differed in the systemic immune organs. In the spleen, ecnccrp-1 was gradually increased in the early infection period and decreased sharply on day 3 post the primary infection, whereas in the head kidney, the transcription of ecnccrp-1 was depressed during almost the whole course of infection. An immunohistochemical analysis showed that EcNCCRP-1(+) cells accumulated at the sites of infection with C. irritans. These results suggested that NCCs were involved in the process of C. irritans infection in E. coioides.


Assuntos
Infecções por Cilióforos/veterinária , Doenças dos Peixes/imunologia , Proteínas de Peixes/genética , Perciformes/genética , Receptores de Antígenos/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Cilióforos/fisiologia , Infecções por Cilióforos/imunologia , Infecções por Cilióforos/metabolismo , Clonagem Molecular , Sequência Conservada , Doenças dos Peixes/metabolismo , Doenças dos Peixes/parasitologia , Proteínas de Peixes/biossíntese , Expressão Gênica/imunologia , Interações Hospedeiro-Parasita , Dados de Sequência Molecular , Especificidade de Órgãos , Perciformes/imunologia , Perciformes/parasitologia , Filogenia , Receptores de Antígenos/biossíntese
7.
Virology ; 446(1-2): 268-75, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24074590

RESUMO

The current antiretroviral therapy (ART) can effectively reduce plasma HIV loads to undetectable levels, but cannot eliminate latently infected resting memory CD4 T cells that persist for the lifetime of infected patients. Therefore, designing new therapeutic approaches to eliminate these latently infected cells or the cells that produce HIV upon reactivation from latency is a priority in the ART era in order to progress to a cure of HIV. Here, we show that "designer" T cells expressing chimeric antigen receptor (CAR), CD4-CD28-CD3ζ, can target and kill HIV Env-expressing cells. Further, they secrete effector cytokines upon contact with HIV Env+ target cells that can reactivate latent HIV in a cell line model, thereby exposing those cells to recognition and killing by anti-HIV CAR+ T cells. Taken to the limit, this process could form the basis for an eventual functional or sterilizing cure for HIV in patients.


Assuntos
Antígenos CD28/biossíntese , Complexo CD3/biossíntese , Antígenos CD4/biossíntese , HIV/imunologia , Receptores de Antígenos/biossíntese , Linfócitos T/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Antígenos CD28/genética , Complexo CD3/genética , Antígenos CD4/genética , Linhagem Celular , Citotoxicidade Imunológica , Infecções por HIV/terapia , Humanos , Imunoterapia/métodos , Receptores de Antígenos/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Produtos do Gene env do Vírus da Imunodeficiência Humana/biossíntese
8.
Eur J Immunol ; 43(8): 2033-42, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23636825

RESUMO

In contrast to thymic epithelial cells, which induce the positive selection of conventional CD8(+) T cells, hematopoietic cells (HCs) select innate CD8(+) T cells whose Ag specificity is not fully understood. Here we show that CD8(+) T cells expressing an H-Y Ag-specific Tg TCR were able to develop in mice in which only HCs expressed MHC class I, when HCs also expressed the H-Y Ag. These HC-selected self-specific CD8(+) T cells resemble innate CD8(+) T cells in WT mice in terms of the expression of memory markers and effector functions, but are phenotypically distinct from the thymus-independent CD8(+) T-cell population. The peripheral maintenance of H-Y-specific CD8(+) T cells required presentation of the self-Ag and IL-15 on HCs. HC-selected CD8(+) T cells in mice lacking the Tg TCR also showed these features. Furthermore, by using MHC class I tetramers with a male Ag peptide, we found that self-Ag-specific CD8(+) T cells in TCR non-Tg mice could develop via HC-induced positive selection, supporting results obtained from H-Y TCR Tg mice. These findings indicate the presence of self-specific CD8(+) T cells that are positively selected by HCs in the peripheral T-cell repertoire.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Antígeno H-Y/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos/biossíntese , Transferência Adotiva , Animais , Autoantígenos/imunologia , Autoantígenos/metabolismo , Diferenciação Celular , Feminino , Antígeno H-Y/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Interleucina-15/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/metabolismo
9.
Methods Mol Biol ; 969: 127-38, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23296932

RESUMO

Genetic modification for enhancing cellular function has been continuously pursued for fighting diseases. Messenger RNA (mRNA) transfection is found to be a promising solution in modifying hematopoietic and immune cells for therapeutic purpose. We have developed a flow electroporation-based system for large volume electroporation of cells with various molecules, including mRNA. This allows robust and scalable mRNA transfection of primary cells of different origin. Here we describe transfection of chimeric antigen receptor (CAR) mRNA into NK cells to modulate the ability of NK cells to target tumor cells. High levels of CAR expression in NK cells can be maintained for 3-7 days post transfection. CD19-specific CAR mRNA transfected NK cells demonstrate targeted lysis of CD19-expressing tumor cells OP-1, primary B-CLL tumor cells, and autologous CD19+ B cells in in vitro assays with enhanced potency: >80% lysis at effector-target ratio of 1:1. This allows current good manufacturing practices (cGMP) and regulatory compliant manufacture of CAR mRNA transfected NK cells for clinical delivery.


Assuntos
Eletroporação/métodos , Células Matadoras Naturais , RNA Mensageiro/química , Transfecção/métodos , Animais , Antígenos CD19/biossíntese , Antígenos CD19/genética , Antígenos CD19/imunologia , Humanos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/imunologia , RNA Mensageiro/metabolismo , Receptores de Antígenos/biossíntese , Receptores de Antígenos/genética , Receptores de Antígenos/imunologia
10.
Methods Mol Biol ; 969: 187-201, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23296935

RESUMO

Redirecting T cells with a chimeric antigen receptor (CAR) of predefined specificity showed remarkable efficacy in the adoptive therapy trials of malignant diseases. The CAR consists of a single chain fragment of variable region (scFv) antibody targeting domain covalently linked to the CD3ζ signalling domain of the T cell receptor complex to mediate T cell activation upon antigen engagement. By using an antibody-derived targeting domain a CAR can potentially redirect T cells towards any target expressed on the cell surface as long as a binding domain is available. Antibody-mediated targeting moreover circumvents MHC restriction of the targeted antigen, thereby broadening the potential of applicability of adoptive T cell therapy. While T cells were so far genetically modified by viral transduction, transient modification with a CAR by RNA transfection gained increasing interest during the last years. This chapter focuses on methods to modify human T cells from peripheral blood with a CAR by electroporation of in vitro transcribed RNA and to test modified T cells for function for use in adoptive immunotherapy.


Assuntos
Transferência Adotiva , Engenharia Celular/métodos , Receptores de Antígenos , Proteínas Recombinantes de Fusão , Linfócitos T , Transfecção/métodos , Animais , Complexo CD3/biossíntese , Complexo CD3/genética , Complexo CD3/imunologia , Expressão Gênica , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Receptores de Antígenos/biossíntese , Receptores de Antígenos/genética , Receptores de Antígenos/imunologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
11.
Cancer Immunol Immunother ; 61(9): 1451-61, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22310931

RESUMO

Natural killer (NK) cells hold promise for adoptive cancer immunotherapy but are dependent on cytokines such as interleukin (IL)-2 for growth and cytotoxicity. Here, we investigated the consequences of ectopic expression of IL-15 in human NK cells. IL-2 and IL-15 belong to the common γ chain family of cytokines and have overlapping activities. Transduction of clinically applicable NK-92 cells with lentiviral vectors encoding human IL-15 resulted in predominantly intracellular expression of the cytokine, and STAT5 activation, proliferation and cytotoxicity of the producer cells in the absence of IL-2. Growth of non-transduced bystander cells was not supported, allowing rapid enrichment of gene-modified cells solely by IL-2 withdrawal. This was also the case upon transduction of NK-92 and NKL cells with a bicistronic lentiviral vector encoding IL-15 and a chimeric antigen receptor (CAR) targeting the pancarcinoma antigen EpCAM. Effector cells co-expressing CAR and IL-15 continued to proliferate in the absence of exogenous cytokines and displayed high and selective cell-killing activity against EpCAM-expressing breast carcinoma cells that were resistant to the natural cytotoxicity of unmodified NK cells. This strategy facilitates rapid isolation and continuous expansion of retargeted NK cells and may extend their potential clinical utility.


Assuntos
Citocinas/biossíntese , Imunoterapia/métodos , Interleucina-15/biossíntese , Células Matadoras Naturais/imunologia , Receptores de Antígenos/imunologia , Animais , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/imunologia , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , DNA Complementar/genética , Molécula de Adesão da Célula Epitelial , Humanos , Interleucina-15/genética , Interleucina-15/imunologia , Interleucina-2/imunologia , Interleucina-2/farmacologia , Células K562 , Células Matadoras Naturais/metabolismo , Leucemia Eritroblástica Aguda/imunologia , Leucemia Eritroblástica Aguda/terapia , Melanoma/imunologia , Melanoma/terapia , Camundongos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Antígenos/biossíntese , Receptores de Antígenos/genética
12.
Expert Opin Biol Ther ; 11(12): 1551-4, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21995504

RESUMO

Adoptive cell transfer (ACT) therapy involves transfer of therapeutic lymphocytes to patients mostly for the treatment of cancer and viral infections. One modality to generate therapeutic lymphocytes is to genetically engineer them to express a chimeric antigen receptor (CAR) capable of recognizing the desired target. Current ACT approaches employ the patient's own (syngeneic) lymphocytes, which is both economically and technically challenging. Using foreign (allogeneic) lymphocytes in ACT is problematic because of the severe immunological reaction that occurs between genetically mismatched individuals. However, recently our group has developed a protocol, which allows for safe and effective ACT therapy in a murine model of metastatic disease using allogeneic T cells redirected with a human EGFR2/neuregulin (Her2/neu)-specific CAR. Mild preconditioning of the recipient delayed the rejection of the allogeneic donor T cells such that they had enough time to destroy the tumor, but not enough to cause significant damage to the host. By modulating lymphocyte migration using FTY720, we were actually able to exploit the allogeneic anti-host reaction in order to augment therapeutic benefit while concurrently improving the safety of the treatment. Therefore, we suggest that CAR-based allogeneic ACT therapy could be universally used as a safe and potent 'off-the-shelf' treatment for cancer.


Assuntos
Terapia Genética , Imunoterapia Adotiva , Neoplasias/terapia , Receptores de Antígenos/biossíntese , Linfócitos T/imunologia , Animais , Terapia Genética/efeitos adversos , Humanos , Imunoterapia Adotiva/efeitos adversos , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Receptores de Antígenos/genética , Resultado do Tratamento
13.
Cancer Res ; 71(8): 2871-81, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21487038

RESUMO

Human T cells genetically modified to express chimeric antigen receptors (CAR) specific to the B cell tumor antigen CD19 can successfully eradicate systemic human CD19(+) tumors in immunocompromised SCID (severe combined immunodeficient)-Beige mice. However, in the clinical setting, CD4(+) CD25(hi) T regulatory cells (Treg) present within the tumor microenvironment may be potent suppressors of tumor-targeted effector T cells. In order to assess the impact of Tregs on CAR-modified T cells in the SCID-Beige xenotransplant model, we isolated, genetically targeted and expanded natural T regulatory cells (nTreg). In vitro nTregs modified to express CD19-targeted CARs efficiently inhibited the proliferation of activated human T cells, as well as the capacity of CD19-targeted 19-28z(+) effector T cells to lyse CD19(+) Raji tumor cells. Intravenous infusion of CD19-targeted nTregs into SCID-Beige mice with systemic Raji tumors traffic to sites of tumor and recapitulate a clinically relevant hostile tumor microenvironment. Antitumor efficacy of subsequently infused 19-28z(+) effector T cells was fully abrogated as assessed by long-term survival of treated mice. Optimal suppression by genetically targeted nTregs was dependent on nTreg to effector T-cell ratios and in vivo nTreg activation. Prior infusion of cyclophosphamide in the setting of this nTreg-mediated hostile microenvironment was able to restore the antitumor activity of subsequently infused 19-28z(+) effector T cells through the eradication of tumor-targeted nTregs. These findings have significant implications for the design of future clinical trials utilizing CAR-based adoptive T-cell therapies of cancer.


Assuntos
Antígenos CD19/imunologia , Linfoma de Burkitt/terapia , Linfócitos T CD8-Positivos/imunologia , Imunoterapia Adotiva/métodos , Receptores de Antígenos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linfoma de Burkitt/tratamento farmacológico , Linfoma de Burkitt/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Ciclofosfamida/farmacologia , Citotoxicidade Imunológica , Humanos , Ativação Linfocitária , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos SCID , Células NIH 3T3 , Receptores de Antígenos/biossíntese , Receptores de Antígenos/genética , Retroviridae/genética , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Virol ; 85(9): 4085-94, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21345961

RESUMO

Viral infections often gain access to the body of their host by exploiting areas of natural vulnerability, such as the semipermeable surfaces of mucosal tissues which are adapted for adsorption of nutrients and other diffusible molecules. Once the microbes have crossed the epithelial barrier, they can disperse to other tissues where eradication may not be possible. The best opportunity for successful immune intervention is immediately after infection while the pathogen is confined to a localized area of the body. Cytotoxic T lymphocytes (CTL) which reside at the site where the infection begins can make an important contribution to immunity by reducing early dissemination of the infection. Because the lungs provide easy access points for many pathogens to enter the body, they require protection from many complementary mechanisms, including pathogen-specific cytotoxic T cells. In this study we show that an enduring response to pathogen-derived peptide antigens facilitates sustained surveillance of the lungs by pathogen-specific CTL during the recovery from influenza virus infection. Our studies show that these processed peptide antigens reinforce expression of two homing receptors (CD69 and CD103) which help recently activated virus-specific CTL colonize the lungs during a mild inflammatory response. We suggest that this requirement for prolonged antigen presentation to reinforce local CTL responses in the lungs explains why protective cellular immunity quickly declines following influenza virus infection and other viral infections that enter the body via mucosal tissues.


Assuntos
Antígenos Virais/imunologia , Pulmão/imunologia , Pulmão/virologia , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/imunologia , Receptores de Antígenos/biossíntese , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos CD/biossíntese , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/biossíntese , Antígenos de Diferenciação de Linfócitos T/imunologia , Expressão Gênica , Cadeias alfa de Integrinas/biossíntese , Cadeias alfa de Integrinas/imunologia , Lectinas Tipo C/biossíntese , Lectinas Tipo C/imunologia , Camundongos , Orthomyxoviridae/patogenicidade , Infecções por Orthomyxoviridae/virologia , Receptores de Antígenos/imunologia
15.
J Comp Physiol B ; 176(7): 599-605, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16676190

RESUMO

The teleost non-specific cytotoxic cells (NCC) are evolutionary precursors of the mammalian natural killer (NK) cells and an important element of innate immunity. The non-specific cytotoxic cell receptor protein (NCCRP1) is a characteristic cell surface protein with main functions in target cell recognition and cytotoxicity with sequence information available for many species of fish. We have isolated a cDNA encoding the Axolotl homologue of fish NCCRP1 out of limb regeneration blastema and analysed its expression by RT-PCR. Sequence analysis revealed a high degree of homology with teleost NCCRP1 on nucleotide and deduced amino acid levels. NCCRP1 contains a conserved C-terminal F-box-associated domain (FBA) and proline-rich motifs (PRM) characteristic for this protein family. NCCRP1 is expressed in multiple tissues with high levels in limb regeneration blastema. The present work describes for the first time the cloning of the NCCRP1 gene in a tetrapod vertebrate providing a valuable link between fish and higher vertebrates. Our findings suggest the existence of NCC in axolotl and a role of the innate immune system in the processes of limb regeneration.


Assuntos
Ambystoma mexicanum/fisiologia , Carpo Animal/fisiologia , Receptores de Antígenos/biossíntese , Regeneração/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Complementar/isolamento & purificação , Imunidade Inata , Dados de Sequência Molecular , Filogenia , Reação em Cadeia da Polimerase , Receptores de Antígenos/análise , Receptores de Antígenos/genética , Alinhamento de Sequência
16.
J Immunol ; 174(5): 2885-91, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15728499

RESUMO

Jawed vertebrates are equipped with TCR and BCR with the capacity to rearrange their V domains. By contrast, jawless vertebrates, represented by hagfish and lampreys, apparently lack such receptors. We describe in this study a family of hagfish genes carrying a single V-type domain resembling those of TCR/BCR. This multigene family, which we call agnathan paired receptors resembling Ag receptors (APAR), is expressed in leukocytes and predicted to encode a group of membrane glycoproteins with organizations characteristic of paired Ig-like receptors, consisting of activating and inhibitory forms. APAR has a J region in its V-type domain, and its V and J regions are encoded in a single exon. Thus, APAR is a member of the emerging families of diversified, innate immune-type receptors with TCR/BCR-like V-type domains and has many of the features expected for a primordial TCR/BCR-like receptor. The extracellular domain of APAR may be descended from a V-type domain postulated to have acquired recombination signal sequences in a jawed vertebrate lineage.


Assuntos
Feiticeiras (Peixe)/imunologia , Região Variável de Imunoglobulina/química , Leucócitos/imunologia , Leucócitos/metabolismo , Glicoproteínas de Membrana/biossíntese , Receptores de Antígenos/biossíntese , Homologia de Sequência de Aminoácidos , Sequência de Aminoácidos , Animais , Éxons , Dosagem de Genes , Genoma , Feiticeiras (Peixe)/genética , Região de Junção de Imunoglobulinas/química , Região de Junção de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Íntrons , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , Família Multigênica/imunologia , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Estrutura Terciária de Proteína/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/metabolismo , Receptores de Antígenos/química , Receptores de Antígenos/genética , Receptores de Antígenos de Linfócitos B/química , Receptores de Antígenos de Linfócitos T/química
17.
Biol Reprod ; 72(2): 460-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15469995

RESUMO

We have examined the passage of maternal cells into the fetus during the gestation and postpartum in mice. Using enhanced green fluorescent protein (EGFP)-transgenic females, we showed that maternal cells frequently gain access to the fetus, mostly in syngeneic pregnancies, but also in allogeneic and outbred crosses. EGFP-transgenic cells, including B, T, and natural killer cells, can persist until adulthood, primarily in bone marrow and thymus. We then asked whether maternal cells, bearing antigens not inherited by the fetus, influence the development of fetal and neonatal B lymphocytes. We have used the B cell receptor 3-83 mu/delta transgenic mouse model, whose B cells recognize the major histocompatibility complex class I molecules H-2Kk and H-2Kb, with a high or moderate affinity, respectively. The fate of transgenic B cells in animals exposed to noninherited H-2Kk or H-2Kb maternal antigens (NIMA) during gestation and lactation was compared with those of nonexposed controls. In H-2Kk-exposed fetuses, NIMA-specific transgenic B cells are partially deleted during late gestation. Nondeleted cells have downmodulated their B cell receptor. In contrast, in NIMA H-2Kb-exposed neonates, transgenic B cells present an activated phenotype, including proliferation, upregulation of surface CD69, and preferential localization in the T cell zone of splenic follicles. This state of activation is still clearly detectable up to 3 wk of age. Thus, we show that fetal and neonatal B cell development is affected by maternal cells bearing antigens noninherited by the fetus and that this phenomenon is highly dependent on the affinity of the B cell receptor for the NIMA.


Assuntos
Linfócitos B/fisiologia , Genes MHC Classe I/fisiologia , Antígenos H-2/fisiologia , Troca Materno-Fetal/fisiologia , Animais , Animais Recém-Nascidos , Linfócitos B/enzimologia , Linfócitos B/metabolismo , Bromodesoxiuridina , Caspase 3 , Caspases/metabolismo , Feminino , Citometria de Fluxo , Imunofluorescência , Idade Gestacional , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Gravidez , Receptores de Antígenos/biossíntese , Receptores de Antígenos/genética , Linfócitos T/enzimologia , Linfócitos T/fisiologia
18.
Novartis Found Symp ; 259: 146-58; discussion 158-69, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15171252

RESUMO

Changes in chromatin structure play a key role in the regulation of the mammalian genome, governing diverse processes including transcription, replication and recombination. In the earliest stages of antigen receptor assembly, D and J segments of the immunoglobulin heavy chain (IgH) and T cell receptor (TCR) beta loci are recombined in B and T cells respectively, while the V segments are not. Distinct distribution patterns of various histone modifications and the nucleosome-remodelling factor Brg1 are found at recombinationally 'active' (DJ) and 'inactive' (V) regions, offering a means independent of transcription or DNAse I hypersensitivity to define chromatin domains at these loci. Within some inactive loci marked by H3-K9 dimethylation, two distinct levels of methylation are found in a non-random, gene-segment specific pattern. Brg1 is not localized to specific sequences, as it is with transcriptional initiation, but rather associates with the entire active locus in a pattern that mirrors acetylation of histone H3. Distinct 'hotspots' of histone H3 dimethylated at lysine 4 are localized at the ends of the active DJ domains of both the IgH and TCRbeta loci, suggesting they may serve as important marks for locus accessibility. The specific patterns of modification imply that the regulation of V(D)J recombination involves recruitment of specific methyltransferases in a localized manner.


Assuntos
Cromatina/fisiologia , Regulação da Expressão Gênica/fisiologia , Receptores de Antígenos/biossíntese , Acetilação , Animais , Linfócitos B/metabolismo , Rearranjo Gênico , Histonas/metabolismo , Humanos , Mamíferos/genética , Mamíferos/fisiologia , Metilação , Receptores de Antígenos/genética , Linfócitos T/metabolismo
19.
J Immunol ; 172(10): 6482-9, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15128841

RESUMO

Sanglifehrin A (SFA) is a recently developed immunosuppressant that belongs to the family of immunophilin-binding ligands. SFA is a cyclophilin A-binding immunosuppressive drug with a novel, but unidentified, mechanism of action. Several reports exist about the effect of SFA on T cells, but its effect on the initiators of the immune response, i.e., dendritic cells (DCs), is relatively unknown. Therefore, we examined the effect of SFA on the differentiation and function of human monocyte-derived DCs. Unlike the well-known cyclophilin A-binding immunosuppressant cyclosporin A, which did not affect DC phenotype, differentiation of DCs in the presence of SFA resulted in CD14-CD1a DCs with normal DC morphology, viability, and a proper capacity to activate allogeneic T cells. However, DCs generated in the presence of SFA demonstrated reduced macropinocytosis and lectin-mediated endocytosis, which was in line with a decreased expression of C-type lectins, including mannose receptor, C1qRP, DC-ASGPR, and especially, DC-SIGN. In contrast, FcalphaRI (CD89) and FcgammaRII (CD32) were increased by SFA. The explicit effect of SFA on the expression of Ag uptake receptors and Ag capture by DCs makes SFA unique among immunophilin-binding immunosuppressive drugs.


Assuntos
Ciclofilinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Endocitose/efeitos dos fármacos , Endocitose/imunologia , Fluoresceína-5-Isotiocianato/análogos & derivados , Imunossupressores/farmacologia , Lactonas/farmacologia , Receptores de Antígenos/metabolismo , Compostos de Espiro/farmacologia , Antígenos CD/biossíntese , Diferenciação Celular/imunologia , Células Cultivadas , Ciclosporina/metabolismo , Ciclosporina/farmacologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Dextranos/antagonistas & inibidores , Dextranos/metabolismo , Fluoresceína-5-Isotiocianato/metabolismo , Humanos , Imunofenotipagem , Lactonas/metabolismo , Lectinas Tipo C/antagonistas & inibidores , Lectinas Tipo C/biossíntese , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Teste de Cultura Mista de Linfócitos , Manose/antagonistas & inibidores , Manose/metabolismo , Receptores de Antígenos/biossíntese , Receptores Fc/biossíntese , Receptores de IgG/biossíntese , Albumina Sérica/antagonistas & inibidores , Albumina Sérica/metabolismo , Compostos de Espiro/metabolismo , Linfócitos T/imunologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA