Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cytokine ; 85: 101-8, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27309676

RESUMO

In October 2013, the International Life Sciences Institute - Health and Environmental Sciences Institute Immunotoxicology Technical Committee (ILSI-HESI ITC) held a one-day workshop entitled, "Workshop on Cytokine Release: State-of-the-Science, Current Challenges and Future Directions". The workshop brought together scientists from pharmaceutical, academic, health authority, and contract research organizations to discuss novel approaches and current challenges for the use of in vitro cytokine release assays (CRAs) for the identification of cytokine release syndrome (CRS) potential of novel monoclonal antibody (mAb) therapeutics. Topics presented encompassed a regulatory perspective on cytokine release and assessment, case studies regarding the translatability of preclinical cytokine data to the clinic, and the latest state of the science of CRAs, including comparisons between mAb therapeutics within one platform and across several assay platforms, a novel physiological assay platform, and assay optimization approaches such as determination of FcR expression profiles and use of statistical tests. The data and approaches presented confirmed that multiple CRA platforms are in use for identification of CRS potential and that the choice of a particular CRA platform is highly dependent on the availability of resources for individual laboratories (e.g. positive and negative controls, number of human blood donors), the assay through-put required, and the mechanism-of-action of the therapeutic candidate to be tested. Workshop participants agreed that more data on the predictive performance of CRA platforms is needed, and current efforts to compare in vitro assay results with clinical cytokine assessments were discussed. In summary, many laboratories continue to focus research efforts on the improvement of the translatability of current CRA platforms as well explore novel approaches which may lead to more accurate, and potentially patient-specific, CRS prediction in the future.


Assuntos
Citocinas/sangue , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Bioensaio/métodos , Humanos , Doenças do Sistema Imunitário/sangue , Doenças do Sistema Imunitário/tratamento farmacológico
2.
J Immunotoxicol ; 13(2): 226-34, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26001195

RESUMO

An important component of safety assessment of new pharmaceuticals is evaluation of their potential to increase the risk of developing cancer in humans. The traditional 2-year rodent bioassay often is not feasible or scientifically applicable for evaluation of biotherapeutics. Additionally, it has poor predictive value for non-genotoxic immunosuppressive compounds. Thus, there is a need for alternative testing strategies. A novel 3-stage tumor model in syngeneic C3H/HeN mice was evaluated here to study the effects of immunosuppressive drugs on tumor promotion and progression in vivo. The model employed a skin squamous cell carcinoma cell line (SCC VII) due to the increased prevalence of squamous cell carcinoma (SCC) in humans associated with immunosuppression after transplants. Local invasion, colonization and tumor progression were evaluated. The validation set of immunosuppressive drugs included: Cyclosporin (CSA), cyclophosphamide (CTX), azathioprine, etanercept, abatacept and prednisone. Local invasion was evaluated by histological assessment as well as fluorescence trafficking from Qdot(®)-labeled tumor cells from the site of inoculation to the draining lymph node. Colonization was evaluated by lung colony counts following intravenous inoculation. Tumor progression was assessed by morphometric analysis of lesion area, angiogenesis and growth fraction of established metastatic neoplasia. Immunosuppressive drugs in the validation set yielded mixed results, including decreased progression. The methods and results described herein using an in vivo syngeneic mouse tumor model can provide insight about the assessment of immunosuppressive drugs in carcinogenicity risk assessment.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas , Neoplasias Experimentais , Neoplasias Cutâneas , Animais , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/patologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Camundongos , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia
3.
J Immunotoxicol ; 12(4): 330-41, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25412621

RESUMO

Murine gammaherpesvirus-68 (MHV-68), a natural pathogen of mice, is being evaluated as a model of Epstein Barr Virus (EBV) infection for use in investigation of the effects of immunomodulatory therapy on herpesvirus pathogenesis in humans. Immunosuppressive agents are used for treatment of a variety of autoimmune diseases as well as for prevention of tissue rejection after organ transplantation and can result in recrudescence of latent herpesvirus infections. Prior to examination of MHV-68 as a suitable model for EBV, better characterization of the MHV-68 model was desirable. Characterization of the MHV-68 model involved development of assays for detecting virus and for demonstration of safety when present in murine colonies. Limited information is available in the literature regarding MHV-68 transmission, although recent reports indicate the virus is not horizontally spread in research facilities. To further determine transmission potential, immunocompetent and immunodeficient mice were infected with MHV-68 and co-habitated with naïve animals. Molecular pathology assays were developed to characterize the MHV-68 model and to determine viral transmission. Horizontal transmission of virus was not observed from infected animals to naïve cagemates after fluorescence microscopy assays and quantitative PCR (qPCR). Serologic analysis complemented these studies and was used as a method of monitoring infection amongst murine colonies. Overall, these findings demonstrate that MHV-68 infection can be controlled and monitored in murine research facilities, and the potential for unintentional infection is low.


Assuntos
Infecções por Herpesviridae/transmissão , Modelos Biológicos , Rhadinovirus/patogenicidade , Infecções Tumorais por Vírus/transmissão , Animais , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Rhadinovirus/imunologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/patologia
4.
J Immunotoxicol ; 12(1): 1-15, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24512328

RESUMO

Immunosuppressive agents are used for treatment of a variety of autoimmune diseases including rheumatoid arthritis (RA), systemic lupus erythematosis (SLE), and psoriasis, as well as for prevention of tissue rejection after organ transplantation. Recrudescence of herpesvirus infections, and increased risk of carcinogenesis from herpesvirus-associated tumors are related with immunosuppressive therapy in humans. Post-transplant lymphoproliferative disorder (PTLD), a condition characterized by development of Epstein Barr Virus (EBV)-associated B-lymphocyte lymphoma, and Kaposi's Sarcoma (KS), a dermal tumor associated with Kaposi Sarcoma-associated virus (KSHV), may develop in solid organ transplant patients. KS also occurs in immunosuppressed Acquired Immunodeficiency (AIDS) patients. Kaposi Sarcoma-associated virus (KSHV) is a herpes virus genetically related to EBV. Murine gammaherpes-virus-68 (MHV-68) is proposed as a mouse model of gammaherpesvirus infection and recrudescence and may potentially have relevance for herpesvirus-associated neoplasia. The pathogenesis of MHV-68 infection in mice mimics EBV/KSHV infection in humans with acute lytic viral replication followed by dissemination and establishment of persistent latency. MHV-68-infected mice may develop lymphoproliferative disease that is accelerated by disruption of the immune system. This manuscript first presents an overview of gammaherpesvirus pathogenesis and immunology as well as factors involved in viral recrudescence. A description of different types of immunodeficiency then follows, with particular focus on viral association with lymphomagenesis after immunosuppression. Finally, this review discusses different gammaherpesvirus animal models and describes a proposed MHV-68 model to further examine the interplay of immunomodulatory agents and gammaherpesvirus-associated neoplasia.


Assuntos
Infecções por Vírus Epstein-Barr/imunologia , Rejeição de Enxerto/prevenção & controle , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 8/imunologia , Imunossupressores/administração & dosagem , Transtornos Linfoproliferativos/imunologia , Transplante de Órgãos , Rhadinovirus/imunologia , Sarcoma de Kaposi/imunologia , Animais , Carcinogênese , Modelos Animais de Doenças , Rejeição de Enxerto/etiologia , Humanos , Imunomodulação , Imunossupressores/efeitos adversos , Recidiva , Ativação Viral/efeitos dos fármacos
5.
Int Immunopharmacol ; 22(2): 465-79, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25107440

RESUMO

The possible onset of Cytokine Release Syndrome (CRS) is an important consideration in the development of monoclonal antibody (mAb) therapeutics. In this study, several machine learning approaches are used to analyze CRS data. The analyzed data come from a human blood in vitro assay which was used to assess the potential of mAb-based therapeutics to produce cytokine release similar to that induced by Anti-CD28 superagonistic (Anti-CD28 SA) mAbs. The data contain 7 mAbs and two negative controls, a total of 423 samples coming from 44 donors. Three (3) machine learning approaches were applied in combination to observations obtained from that assay, namely (i) Hierarchical Cluster Analysis (HCA); (ii) Principal Component Analysis (PCA) followed by K-means clustering; and (iii) Decision Tree Classification (DTC). All three approaches were able to identify the treatment that caused the most severe cytokine response. HCA was able to provide information about the expected number of clusters in the data. PCA coupled with K-means clustering allowed classification of treatments sample by sample, and visualizing clusters of treatments. DTC models showed the relative importance of various cytokines such as IFN-γ, TNF-α and IL-10 to CRS. The use of these approaches in tandem provides better selection of parameters for one method based on outcomes from another, and an overall improved analysis of the data through complementary approaches. Moreover, the DTC analysis showed in addition that IL-17 may be correlated with CRS reactions, although this correlation has not yet been corroborated in the literature.


Assuntos
Anticorpos Monoclonais/farmacologia , Inteligência Artificial , Citocinas/imunologia , Anticorpos Monoclonais/efeitos adversos , Bioensaio , Análise por Conglomerados , Árvores de Decisões , Humanos , Análise de Componente Principal , Síndrome
6.
Curr Pharm Biotechnol ; 14(2): 242-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23157711

RESUMO

CNTO 530 is an erythropoietin receptor agonist MIMETIBODYTM construct. CNTO 530 has been shown to be active in a number of rodent models of acquired anemia (e.g. renal insufficiency and chemotherapy induced anemia). We investigated the efficacy of CNTO 530 in murine models of ß-thalassemia and sickle cell anemia (Berkeley mice). ß- thalassemic mice are deficient in expression of α-globin chain and heterozygous mice are characterized by a clinical syndrome similar to the human ß-thalassemia intermedia. Berkeley mice are knocked out for murine alpha and beta globin and are transgenic for human alpha, beta (sickle) and gamma globin genes. Berkeley mice thus express human sickle hemoglobin A (HbS) and can also express human fetal hemoglobin. These mice express a severe compensated hypochromic microcytic anemia and display the sickle cell phenotype. To test the effectiveness of CNTO 530, mice from both genotypes received a single subcutaneous (s.c.) dose of CNTO 530 or darbepoetin-α (as a comparator) at 10,000 U/kg, a dose shown to cause a similar increase in reticulocytes and hemoglobin in normal mice. Hematologic parameters were evaluated over time. CNTO 530, but not darbepoetin-α, increased reticulocytes, red blood cells and total hemoglobin in ß- thalassemic mice. In Berkeley mice CNTO 530 showed an increase in reticulocytes, red blood cells, F-cells, total hemoglobin and fetal hemoglobin. In conclusion, CNTO 530 is effective in murine models of ß-thalassemia and sickle cell anemia. These data suggest that CNTO 530 may have beneficial effects in patients with genetically mediated hemoglobinopathies.


Assuntos
Anemia Falciforme/tratamento farmacológico , Receptores da Eritropoetina/agonistas , Proteínas Recombinantes de Fusão/uso terapêutico , Talassemia beta/tratamento farmacológico , Anemia Falciforme/sangue , Animais , Darbepoetina alfa , Modelos Animais de Doenças , Contagem de Eritrócitos , Eritropoetina/análogos & derivados , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Feminino , Hematínicos/farmacologia , Hematínicos/uso terapêutico , Hemoglobinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/farmacologia , Talassemia beta/sangue
7.
J Immunotoxicol ; 9(1): 43-55, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22299716

RESUMO

Many immunosuppressive drugs are associated with an increased risk of neoplasia, principally non-melanoma skin cancers and B-cell lymphomas. However, only 6 of the 13 immunosuppressive drugs tested in 2 year bioassays increased the incidence of neoplasia. For example, the 2-year bioassays conducted with cyclosporine (CSA), an International Agency for Research on Cancer (IARC) Group 1 human carcinogen, were negative. The purpose of these investigations was to use transplanted tumor models in immunocompetent, syngeneic mice to gain insight into the failure of the 2-year bioassay to show an increased incidence of neoplasia with CSA. C3H HeN mice were used in a battery of assays with a transplanted squamous cell carcinoma (SCC VII cells) or a B-cell, lymphoma (38C13 cells) cells to study effects of CSA on local growth and metastases, experimental metastases, and progression of established metastases. Mice received CSA twice weekly by subcutaneous (SC) injection at doses of 0.5, 5, or 50 mg/kg; controls received the CSA vehicle. CSA had a modest inhibitory effect on SC tumors initiated by 38C13 cells and on intramuscular tumors initiated by SCC VII cells. CSA also decreased the number of lung colonies and decreased the size, growth fraction and vascularity of established lung metastases initiated by SCC VII cells. In contrast, CSA increased progressive growth of metastases to the sentinel lymph node from an intramuscular SCC VII tumor, but had no effect cellular traffic to the node. In conclusion, CSA at doses up to 50 mg/kg did not facilitate tumor progression and it partially inhibited tumor growth, suggesting that suppression of tumor progression may partially explain the failure of CSA to act as a carcinogen in 2 year bioassays.


Assuntos
Carcinoma de Células Escamosas/secundário , Ciclosporina/toxicidade , Imunossupressores/toxicidade , Neoplasias Pulmonares/secundário , Linfoma de Células B/patologia , Neoplasias Musculares/patologia , Animais , Testes de Carcinogenicidade , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/prevenção & controle , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclosporina/administração & dosagem , Progressão da Doença , Relação Dose-Resposta a Droga , Feminino , Imunossupressores/administração & dosagem , Injeções Subcutâneas , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/prevenção & controle , Metástase Linfática , Linfoma de Células B/imunologia , Linfoma de Células B/prevenção & controle , Camundongos , Camundongos Endogâmicos C3H , Neoplasias Musculares/imunologia , Neoplasias Musculares/prevenção & controle , Invasividade Neoplásica , Medição de Risco , Fatores de Tempo , Carga Tumoral/efeitos dos fármacos
8.
Int Immunopharmacol ; 11(11): 1697-705, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21689786

RESUMO

Anti-CD28 superagonist (SA) mediated cytokine release syndrome (CRS), an adverse event resulting in systemic release of cytokines, is an emergent issue in drug development. CRS is of potential concern for all monoclonal antibodies (mAbs) particularly those directed against cell surface targets on lymphocytes. Concern regarding patient safety requires development of novel methods to predict these adverse reactions. Due to the inability of animal studies to predict CRS, we have developed a whole blood in vitro screen to support First in Human studies and assess the potential for mAbs to cause anti-CD28 SA-like CRS. For this purpose we have immobilized marketed mAbs, whose potential for causing CRS and milder infusion reactions is known, on Protein A beads and used these beads to stimulate cytokine release. After culture, supernatants are harvested and frozen for later multiplex analysis of cytokines using Searchlight™ technology. We have employed hierarchicalluster analysis (HCA) to allow comparison of 12 different cytokine levels across numerous donors, treatments, and experiments. Results conclusively distinguish test mAb responses from an anti-CD28 superagonist mAb response. As part of a global analysis of preclinical data, the results of this assay can facilitate entry into First in Human clinical trials, help with selection of starting doses and may allow more rapid dose escalation using smaller cohorts.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Antígenos CD28/imunologia , Citocinas/sangue , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/sangue , Ensaio de Imunoadsorção Enzimática/métodos , Doenças do Sistema Imunitário/sangue , Análise por Conglomerados , Citocinas/imunologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/imunologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/metabolismo , Ensaio de Imunoadsorção Enzimática/estatística & dados numéricos , Humanos , Doenças do Sistema Imunitário/induzido quimicamente , Doenças do Sistema Imunitário/metabolismo , Valor Preditivo dos Testes , Receptores Fc/metabolismo , Síndrome
9.
J Pharmacol Toxicol Methods ; 63(3): 236-49, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21156211

RESUMO

INTRODUCTION: Immunosuppressive drugs are associated with an increased risk of infections and in some cases neoplasia, particularly non-melanoma skin cancers. This paper describes the development of a model to test the effects of immunosuppressive drugs on local invasion and metastases of a squamous cell carcinoma in syngeneic, immunocompetent mice. METHODS: SCC VII cells were labeled with 655 quantum dots (QDs), injected intramuscularly into C3H HEN mice and traffic and progressive growth in the draining popliteal lymph node were evaluated. RESULTS: SCC VII cells express RAE-1, an NKG2D ligand, and were sensitive to natural killer (NK) cells in vitro. QDs were stable in SCC VII cells and showed no evidence of toxicity to the cells. In vivo, confocal microscopy showed that QD-labeled SCC VII cells could migrate to the draining node and microfluorimetry showed progressive traffic of QDs to the node. There was no evidence of systemic toxicity of QDs. Primary immunosuppression in SCID and SCID-beige mice and treatment of normal mice with immunosuppressive agents (anti-asialoGM1 and cyclophosphamide) can enhance traffic of QDs and/or metastases to the draining lymph node. In contrast, cyclosporine had no effect on traffic or metastases. CONCLUSION: This model of local invasion and metastases may be useful in immunotoxicology for identifying and characterizing the hazard posed by selective immunosuppressive drugs.


Assuntos
Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/secundário , Imunossupressores/toxicidade , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/secundário , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Citometria de Fluxo , Imuno-Histoquímica , Metástase Linfática , Camundongos , Camundongos Endogâmicos C3H , Camundongos SCID , Transplante de Neoplasias
10.
Int J Toxicol ; 29(5): 435-66, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20884856

RESUMO

Many immunosuppressive drugs are associated with an increased risk of B-cell lymphoma, squamous cell carcinoma, and Kaposi sarcoma. Thirteen immunosuppressive drugs have been tested in 2-year carcinogenicity studies (abatacept; azathioprine; busulfan; cyclophosphamide; cyclosporine; dexamethasone; everolimus; leflunomide; methotrexate; mycophenolate mofetil; prednisone; sirolimus; and tacrolimus) and in additional models including neonatal and genetically modified mice; chemical, viral, ultraviolet, and ionizing radiation co-carcinogenesis, and in models with transplanted tumor cells. The purpose of this review is to outline the mechanisms by which immunosuppressive drugs can influence neoplasia, to summarize the available preclinical data on the 13 drugs, and to critically review the performance of the models. A combination of primary tumor and metastasis assays conducted with transplanted cells may provide the highest value for hazard identification and can be applied on a case-by-case basis. However, for both small molecules and therapeutic proteins, determining the relative risk to patients from preclinical data remains problematic. Classifying immunosuppressive drugs based on their mechanism of action and hazard identification from preclinical studies and a prospective pharmacovigilance program to monitor carcinogenic risk may be a feasible way to manage patient safety during the clinical development program and postmarketing.


Assuntos
Vigilância Imunológica/efeitos dos fármacos , Terapia de Imunossupressão/efeitos adversos , Imunossupressores/efeitos adversos , Imunossupressores/farmacologia , Neoplasias/epidemiologia , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Vigilância Imunológica/fisiologia , Imunossupressores/classificação , Neoplasias/fisiopatologia , Transplante de Órgãos , Vigilância de Produtos Comercializados , Testes de Toxicidade Crônica/métodos
11.
Curr Opin Drug Discov Devel ; 13(1): 124-35, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20047153

RESUMO

Infusion reactions and cytokine release syndrome (CRS) are an emerging issue in drug development and are of particular importance with the development of new therapeutic proteins. Increasing concerns regarding patient safety require a better understanding of the mechanism involved and the development of novel methods for preventing and predicting such reactions and CRS. This review discusses developments during the past few years in understanding the mechanisms that cause infusion reactions and CRS, advances in approaches to prevent CRS, the reason why preclinical animal models are unreliable predictors of CRS, and new developments in the design and analysis of in vitro screening systems for the prediction of CRS.


Assuntos
Citocinas/metabolismo , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/induzido quimicamente , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/imunologia , Doenças do Sistema Imunitário/induzido quimicamente , Doenças do Sistema Imunitário/metabolismo , Animais , Análise por Conglomerados , Citocinas/imunologia , Humanos , Receptores Fc/metabolismo
12.
Cell Immunol ; 258(1): 65-71, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19410243

RESUMO

Mouse studies demonstrated that infusion of CD4+CD25+ regulatory T cells (Tregs) prevented graft versus host disease (GVHD) lethality after bone marrow transplantation (BMT). But the potential impact of human Tregs on GVHD has not been well demonstrated. In this study, we demonstrated that human Tregs enriched from peripheral blood of healthy donors could be expanded ex vivo to clinically relevant cell numbers in 2-3 weeks while maintaining Foxp3, CD25, CTLA-4, and CD62L expression as well as in vitro suppressive function. Furthermore, injection of human PBL into NOD/SCID mice induced lethal xenogenic GVHD, but co-transfer of expanded human Tregs with human PBL significantly enhanced survival, reduced GVHD symptoms, and inhibited human IgG/IgM production in the NOD/SCID mice. These results demonstrated that ex vivo expanded human Tregs retained their in vivo suppressive activity and prevented lethal xenogeneic GVHD, revealing the therapeutic potential of expanded human Tregs for GVHD.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/transplante , Transplante Heterólogo/imunologia , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Antígeno CTLA-4 , Técnicas de Cultura de Células , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Doença Enxerto-Hospedeiro/metabolismo , Doença Enxerto-Hospedeiro/mortalidade , Humanos , Subunidade alfa de Receptor de Interleucina-2/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Selectina L/imunologia , Selectina L/metabolismo , Camundongos , Camundongos SCID , Linfócitos T Reguladores/metabolismo
13.
Eur J Immunol ; 39(2): 612-20, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19180473

RESUMO

CD4(+)CD25(+)FOXP3(+) Treg cells require TCR engagement for suppressive function, thus ensuring that suppression occurs only in the presence of specific antigens; however, to date no studies have addressed the function of self-antigen-specific Treg in humans. These studies were designed to determine whether peripheral generation and function of islet antigen-specific adaptive Treg are defective in human subjects with type 1 diabetes (T1D). Islet antigen-specific adaptive Treg were induced in vitro by activation of CD4(+)FOXP3(-) T cells with glutamic acid decarboxylase and islet-specific glucose-6-phosphate catalytic subunit-related protein peptides in the context of T1D-associated HLA-DRbeta alleles. Antigen-specific Treg were characterized using flow cytometry for FOXP3 and class II tetramer and assessed for the ability to inhibit proliferation. These adaptive Treg were then compared with influenza-specific Treg from the same study population. The function of tetramer(+) cells that expressed FOXP3 was similar for both influenza and islet antigens generated from control and T1D subjects. In fact, the potency of suppression correlated with FOXP3 expression, not antigen specificity. Thus, these data suggest that development of functional adaptive Treg can occur in response to islet antigens and activation of islet-specific Treg may potentially be used as a targeted immunotherapy in T1D.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Ilhotas Pancreáticas/imunologia , Linfócitos T Reguladores/imunologia , Antígenos CD4/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Glucose-6-Fosfatase/imunologia , Glucose-6-Fosfatase/metabolismo , Glutamato Descarboxilase/imunologia , Glutamato Descarboxilase/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Subunidade alfa de Receptor de Interleucina-2/imunologia , Fragmentos de Peptídeos/imunologia , Linfócitos T Reguladores/metabolismo
14.
Proc Natl Acad Sci U S A ; 102(11): 4103-8, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15753318

RESUMO

Antigen-specificity is a hallmark of adaptive T cell-mediated immune responses. CD4+CD25+FOXP3+ regulatory T cells (T(R)) also require activation through the T cell receptor for function. Although these cells require antigen-specific activation, they are generally able to suppress bystander T cell responses once activated. This raises the possibility that antigen-specific T(R) may be useful therapeutically by localizing generalized suppressive activity to tissues expressing select target antigens. Here, we demonstrate that T(R) specific for particular peptide-MHC complexes can be generated from human CD4+CD25- T cells in vitro and isolated by using HLA class II tetramers. Influenza hemagglutinin epitopes were used to generate hemagglutinin-specific T(R), which required cognate antigen for activation but which subsequently suppressed noncognate bystander T cell responses as well. These findings have implications for the generation of therapeutic regulatory T cells in disease, and also suggest an important mechanism by which T cells may be regulated at the site of inflammation.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Receptores de Interleucina-2/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Memória Imunológica/imunologia
15.
Cell Immunol ; 230(1): 33-43, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15541717

RESUMO

This study focused on synthesis of MHC class II glycoproteins (MHCII) by rat CD4(+) T-helper cells. During activation in Con A and IL-2, purified rat splenic CD4(+) T cells expressed abundant surface MHCII together with transcripts for I-A alpha/beta, invariant chain, and the type III and type IV MHC class II transactivator (CIITA). Activated thymic CD8(+)CD4(-) and CD8(+)CD4(+) T cells exhibited essentially the same phenotype. MHCII synthesis by CD4(+) T cells enabled presentation of myelin basic protein (MBP) to antigen-specific responders. T cell expression of MHCII was due to direct biosynthesis rather than adsorption from professional APC; indeed, T cell-mediated expression of MHCII was optimal in the absence of professional APC. Despite periodic reactivation with Con A during 3-4 weeks of culture, CD4(+) T cells repressed MHCII synthesis and reverted to a MHCII(-) phenotype. These short-term lines resembled established lines of MBP-specific T cells in that mitogenic activation elicited extensive blastogenesis without MHCII synthesis. Activation-dependent synthesis of MHCII however was partially restored in lines of mitogen-stimulated T cells when the cultures were reconstituted with irradiated splenic APC. These data indicate that most naive rat CD4(+) T cells exhibit activation-dependent synthesis of MHCII whereas continuously propagated T cells require an APC-derived signal to support MHCII synthesis.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/imunologia , Ativação Linfocitária , Transdução de Sinais , Animais , Linfócitos T CD4-Positivos/citologia , Diferenciação Celular , Células Cultivadas , Concanavalina A/imunologia , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/genética , Memória Imunológica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Lew , Baço/imunologia , Baço/metabolismo
16.
J Clin Invest ; 112(9): 1437-43, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14597769

RESUMO

CD4+CD25+ regulatory T (TR) cells have been described in both humans and mice. In mice, TR are thymically derived, and lack of TR leads to organ-specific autoimmunity. Recently, the forkhead/winged helix transcription factor, FoxP3, has been shown to be important for the function of TR cells in mice. In this study, human TR cells were examined and, in results similar to those of studies done in mice, expression of FoxP3 was found exclusively in CD4+CD25+ T cells and correlated with the suppressive activity of these cells. In contrast to the mouse studies, activation of human CD4+CD25- T cells led to expression of FoxP3. Expression of FoxP3 in activated human CD4+CD25+ cells also correlated with suppression of proliferation by these cells in freshly isolated CD4+CD25- T cells from the same donor. This suppression was cell-contact dependent and cytokine independent. Thus, in humans, during activation of CD4+CD25- T cells in an immune response, two populations of cells may arise, effector CD4+CD25+ and regulatory CD4+CD25+ T cells, with expression of FoxP3 correlated with regulatory activity. These data also raise the possibility that a failure to generate peripheral TR cells properly may contribute to autoimmune disease and suggest a possible therapeutic role for FoxP3 in the treatment of such diseases.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Proteínas de Ligação a DNA/fisiologia , Ativação Linfocitária , Receptores de Interleucina-2/análise , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/genética , Fatores de Transcrição Forkhead , Humanos , RNA Mensageiro/análise , Receptores de Antígenos de Linfócitos T/fisiologia
17.
Cell Immunol ; 218(1-2): 13-25, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12470610

RESUMO

This study provides evidence that both rat and mouse thymic and splenic T cells express significant levels of MHC class II glycoproteins (MHCII) in vivo. Derivation of rat and mouse chimeras revealed that a major source of MHCII on thymic T cells was acquired from radioresistant host APC. Expression of MHC on thymic T cells appeared physiologically relevant because presentation of rat myelin basic protein (RMBP) by nonadherent, radiosensitive thymic T cells was associated with the adoptive transfer of tolerance. Mature MBP-specific effector T cells isolated from the CNS in both rat and mouse models of EAE also expressed significant levels of MHCII. Adoptive transfer of activated B10.PL MBP/I-A(u)-restricted TCR transgenic T cells into F1(C57BL/6 x B10.PL) mice revealed acquisition of allogeneic I-A(b) on encephalitogenic CNS-derived T cells. Overall, this study indicates that immature and mature T cells in rats and mice acquire functional MHCII in vivo during thymic development and pathogenic inflammation.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos/imunologia , Sistema Nervoso Central/imunologia , Encefalomielite Autoimune Experimental/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Fragmentos de Peptídeos/imunologia , Subpopulações de Linfócitos T/imunologia , Timo/citologia , Transferência Adotiva , Animais , Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/patologia , Cobaias , Tolerância Imunológica , Interleucina-2/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Básica da Mielina/imunologia , Especificidade de Órgãos , Quimera por Radiação , Tolerância a Radiação , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Lew , Especificidade da Espécie , Baço/citologia , Baço/imunologia , Subpopulações de Linfócitos T/metabolismo , Timo/crescimento & desenvolvimento , Timo/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA