Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cytokine ; 85: 101-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27309676

ABSTRACT

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.


Subject(s)
Cytokines/blood , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Biological Assay/methods , Humans , Immune System Diseases/blood , Immune System Diseases/drug therapy
2.
J Immunotoxicol ; 13(2): 226-34, 2016.
Article in English | MEDLINE | ID: mdl-26001195

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell , Neoplasms, Experimental , Skin Neoplasms , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Drug Screening Assays, Antitumor/methods , Mice , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/pathology
3.
J Immunotoxicol ; 12(4): 330-41, 2015.
Article in English | MEDLINE | ID: mdl-25412621

ABSTRACT

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.


Subject(s)
Herpesviridae Infections/transmission , Models, Biological , Rhadinovirus/pathogenicity , Tumor Virus Infections/transmission , Animals , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Mice , Mice, Inbred BALB C , Mice, SCID , Rhadinovirus/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/pathology
4.
J Immunotoxicol ; 12(1): 1-15, 2015.
Article in English | MEDLINE | ID: mdl-24512328

ABSTRACT

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.


Subject(s)
Epstein-Barr Virus Infections/immunology , Graft Rejection/prevention & control , Herpesvirus 4, Human/immunology , Herpesvirus 8, Human/immunology , Immunosuppressive Agents/administration & dosage , Lymphoproliferative Disorders/immunology , Organ Transplantation , Rhadinovirus/immunology , Sarcoma, Kaposi/immunology , Animals , Carcinogenesis , Disease Models, Animal , Graft Rejection/etiology , Humans , Immunomodulation , Immunosuppressive Agents/adverse effects , Recurrence , Virus Activation/drug effects
5.
Int Immunopharmacol ; 22(2): 465-79, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25107440

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/pharmacology , Artificial Intelligence , Cytokines/immunology , Antibodies, Monoclonal/adverse effects , Biological Assay , Cluster Analysis , Decision Trees , Humans , Principal Component Analysis , Syndrome
6.
Curr Pharm Biotechnol ; 14(2): 242-8, 2013.
Article in English | MEDLINE | ID: mdl-23157711

ABSTRACT

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.


Subject(s)
Anemia, Sickle Cell/drug therapy , Receptors, Erythropoietin/agonists , Recombinant Fusion Proteins/therapeutic use , beta-Thalassemia/drug therapy , Anemia, Sickle Cell/blood , Animals , Darbepoetin alfa , Disease Models, Animal , Erythrocyte Count , Erythropoietin/analogs & derivatives , Erythropoietin/pharmacology , Erythropoietin/therapeutic use , Female , Hematinics/pharmacology , Hematinics/therapeutic use , Hemoglobins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recombinant Fusion Proteins/pharmacology , beta-Thalassemia/blood
7.
J Immunotoxicol ; 9(1): 43-55, 2012.
Article in English | MEDLINE | ID: mdl-22299716

ABSTRACT

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.


Subject(s)
Carcinoma, Squamous Cell/secondary , Cyclosporine/toxicity , Immunosuppressive Agents/toxicity , Lung Neoplasms/secondary , Lymphoma, B-Cell/pathology , Muscle Neoplasms/pathology , Animals , Carcinogenicity Tests , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/prevention & control , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclosporine/administration & dosage , Disease Progression , Dose-Response Relationship, Drug , Female , Immunosuppressive Agents/administration & dosage , Injections, Subcutaneous , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lymphatic Metastasis , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/prevention & control , Mice , Mice, Inbred C3H , Muscle Neoplasms/immunology , Muscle Neoplasms/prevention & control , Neoplasm Invasiveness , Risk Assessment , Time Factors , Tumor Burden/drug effects
8.
Int Immunopharmacol ; 11(11): 1697-705, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21689786

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/adverse effects , CD28 Antigens/immunology , Cytokines/blood , Drug-Related Side Effects and Adverse Reactions/blood , Enzyme-Linked Immunosorbent Assay/methods , Immune System Diseases/blood , Cluster Analysis , Cytokines/immunology , Drug-Related Side Effects and Adverse Reactions/immunology , Drug-Related Side Effects and Adverse Reactions/metabolism , Enzyme-Linked Immunosorbent Assay/statistics & numerical data , Humans , Immune System Diseases/chemically induced , Immune System Diseases/metabolism , Predictive Value of Tests , Receptors, Fc/metabolism , Syndrome
9.
J Pharmacol Toxicol Methods ; 63(3): 236-49, 2011.
Article in English | MEDLINE | ID: mdl-21156211

ABSTRACT

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.


Subject(s)
Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/secondary , Immunosuppressive Agents/toxicity , Neoplasms, Experimental/immunology , Neoplasms, Experimental/secondary , Animals , Cell Culture Techniques , Cell Line, Tumor , Flow Cytometry , Immunohistochemistry , Lymphatic Metastasis , Mice , Mice, Inbred C3H , Mice, SCID , Neoplasm Transplantation
10.
Int J Toxicol ; 29(5): 435-66, 2010.
Article in English | MEDLINE | ID: mdl-20884856

ABSTRACT

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.


Subject(s)
Immunologic Surveillance/drug effects , Immunosuppression Therapy/adverse effects , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacology , Neoplasms/epidemiology , Animals , Drug Evaluation, Preclinical/methods , Humans , Immunologic Surveillance/physiology , Immunosuppressive Agents/classification , Neoplasms/physiopathology , Organ Transplantation , Product Surveillance, Postmarketing , Toxicity Tests, Chronic/methods
11.
Curr Opin Drug Discov Devel ; 13(1): 124-35, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20047153

ABSTRACT

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.


Subject(s)
Cytokines/metabolism , Drug-Related Side Effects and Adverse Reactions/chemically induced , Drug-Related Side Effects and Adverse Reactions/immunology , Immune System Diseases/chemically induced , Immune System Diseases/metabolism , Animals , Cluster Analysis , Cytokines/immunology , Humans , Receptors, Fc/metabolism
12.
Cell Immunol ; 258(1): 65-71, 2009.
Article in English | MEDLINE | ID: mdl-19410243

ABSTRACT

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.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Transplantation, Heterologous/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , CTLA-4 Antigen , Cell Culture Techniques , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/metabolism , Graft vs Host Disease/mortality , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , L-Selectin/immunology , L-Selectin/metabolism , Mice , Mice, SCID , T-Lymphocytes, Regulatory/metabolism
13.
Eur J Immunol ; 39(2): 612-20, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19180473

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , T-Lymphocytes, Regulatory/immunology , CD4 Antigens/immunology , Diabetes Mellitus, Type 1/metabolism , Glucose-6-Phosphatase/immunology , Glucose-6-Phosphatase/metabolism , Glutamate Decarboxylase/immunology , Glutamate Decarboxylase/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Peptide Fragments/immunology , T-Lymphocytes, Regulatory/metabolism
14.
Proc Natl Acad Sci U S A ; 102(11): 4103-8, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15753318

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Receptors, Interleukin-2/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunologic Memory/immunology
15.
Cell Immunol ; 230(1): 33-43, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15541717

ABSTRACT

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.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation , Signal Transduction , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation , Cells, Cultured , Concanavalin A/immunology , Flow Cytometry , Histocompatibility Antigens Class II/genetics , Immunologic Memory , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred BN , Rats, Inbred Lew , Spleen/immunology , Spleen/metabolism
16.
J Clin Invest ; 112(9): 1437-43, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14597769

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , DNA-Binding Proteins/physiology , Lymphocyte Activation , Receptors, Interleukin-2/analysis , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Forkhead Transcription Factors , Humans , RNA, Messenger/analysis , Receptors, Antigen, T-Cell/physiology
17.
Cell Immunol ; 218(1-2): 13-25, 2002.
Article in English | MEDLINE | ID: mdl-12470610

ABSTRACT

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.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Histocompatibility Antigens Class II/immunology , Peptide Fragments/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Adoptive Transfer , Animals , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Guinea Pigs , Immune Tolerance , Interleukin-2/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Basic Protein/immunology , Organ Specificity , Radiation Chimera , Radiation Tolerance , Rats , Rats, Inbred BN , Rats, Inbred Lew , Species Specificity , Spleen/cytology , Spleen/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/growth & development , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...