Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
1.
Am J Transplant ; 16(2): 389-97, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26588186

ABSTRACT

Our understanding of the molecular pathways that control immune responses, particularly immunomodulatory molecules that control the extent and duration of an immune response, have led to new approaches in the field of transplantation immunology to induce allograft survival. These molecular pathways are being defined precisely in murine models and translated into clinical practice; however, many of the newly available drugs are human-specific reagents. Furthermore, many species-specific differences exist between mouse and human immune systems. Recent advances in the development of humanized mice, namely, immunodeficient mice engrafted with functional human immune systems, have led to the availability of a small animal model for the study of human immune responses. Humanized mice represent an important preclinical model system for evaluation of new drugs and identification of the mechanisms underlying human allograft rejection without putting patients at risk. This review highlights recent advances in the development of humanized mice and their use as preclinical models for the study of human allograft responses.


Subject(s)
Disease Models, Animal , Graft Rejection/prevention & control , Organ Transplantation , Transplantation Immunology , Animals , Graft Rejection/immunology , Humans , Mice , Prognosis
2.
Oncogene ; 35(9): 1111-21, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-25982283

ABSTRACT

IMP3 (insulin-like growth factor-2 mRNA binding protein 3) is an oncofetal protein whose expression is prognostic for poor outcome in several cancers. Although IMP3 is expressed preferentially in triple-negative breast cancer (TNBC), its function is poorly understood. We observed that IMP3 expression is significantly higher in tumor initiating than in non-tumor initiating breast cancer cells and we demonstrate that IMP3 contributes to self-renewal and tumor initiation, properties associated with cancer stem cells (CSCs). The mechanism by which IMP3 contributes to this phenotype involves its ability to induce the stem cell factor SOX2. IMP3 does not interact with SOX2 mRNA significantly or regulate SOX2 expression directly. We discovered that IMP3 binds avidly to SNAI2 (SLUG) mRNA and regulates its expression by binding to the 5' UTR. This finding is significant because SLUG has been implicated in breast CSCs and TNBC. Moreover, we show that SOX2 is a transcriptional target of SLUG. These data establish a novel mechanism of breast tumor initiation involving IMP3 and they provide a rationale for its association with aggressive disease and poor outcome.


Subject(s)
Biomarkers, Tumor/biosynthesis , RNA-Binding Proteins/biosynthesis , Transcription Factors/biosynthesis , Triple Negative Breast Neoplasms/genetics , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplastic Stem Cells , Prognosis , RNA, Messenger/biosynthesis , RNA-Binding Proteins/genetics , SOXB1 Transcription Factors/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Triple Negative Breast Neoplasms/pathology
3.
Clin Exp Immunol ; 179(3): 398-413, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25302633

ABSTRACT

Several ß cell antigens recognized by T cells in the non-obese diabetic (NOD) mouse model of type 1 diabetes (T1D) are also T cell targets in the human disease. While numerous antigen-specific therapies prevent diabetes in NOD mice, successful translation of rodent findings to patients has been difficult. A human leucocyte antigen (HLA)-transgenic mouse model incorporating human ß cell-specific T cells might provide a better platform for evaluating antigen-specific therapies. The ability to study such T cells is limited by their low frequency in peripheral blood and the difficulty in obtaining islet-infiltrating T cells from patients. We have worked to overcome this limitation by using lentiviral transduction to 'reprogram' primary human CD8 T cells to express three T cell receptors (TCRs) specific for a peptide derived from the ß cell antigen islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP265-273 ) and recognized in the context of the human class I major histocompatibility complex (MHC) molecule HLA-A2. The TCRs bound peptide/MHC multimers with a range of avidities, but all bound with at least 10-fold lower avidity than the anti-viral TCR used for comparison. One exhibited antigenic recognition promiscuity. The ß cell-specific human CD8 T cells generated by lentiviral transduction with one of the TCRs released interferon (IFN)-γ in response to antigen and exhibited cytotoxic activity against peptide-pulsed target cells. The cells engrafted in HLA-A2-transgenic NOD-scid IL2rγ(null) mice and could be detected in the blood, spleen and pancreas up to 5 weeks post-transfer, suggesting the utility of this approach for the evaluation of T cell-modulatory therapies for T1D and other T cell-mediated autoimmune diseases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Genetic Vectors/genetics , Immunotherapy, Adoptive/methods , Insulin-Secreting Cells/immunology , Lentivirus/genetics , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8-Positive T-Lymphocytes/transplantation , Cell Survival , Glucose-6-Phosphatase/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Jurkat Cells , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin-2/genetics , T-Lymphocytes, Cytotoxic/transplantation
4.
Stem Cell Res ; 13(3 Pt A): 404-12, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25310256

ABSTRACT

Preclinical research in animals often fails to adequately predict the outcomes observed in human patients. Chimeric animals bearing individual human tissues have been developed to provide improved models of human-specific cellular processes. Mice transplanted with human hematopoietic stem cells can be used to study human immune responses, infections of blood cells and processes of hematopoiesis. Animals with humanized livers are useful for modeling hepatotropic infections as well as drug metabolism and hepatotoxicity. However, many pathophysiologic processes involve both the liver and the hematolymphoid system. Examples include hepatitis C/HIV co-infection, immune mediated liver diseases, liver injuries with inflammation such as steatohepatitis and alcoholic liver disease. We developed a robust protocol enabling the concurrent double-humanization of mice with mature hepatocytes and human blood. Immune-deficient, fumarylacetoacetate hydrolase (Fah(-/-)), Rag2(-/-) and Il2rg(-/-) deficient animals on the NOD-strain background (FRGN) were simultaneously co-transplanted with adult human hepatocytes and hematopoietic stem cells after busulfan and Ad:uPA pre-conditioning. Four months after transplantation the average human liver repopulation exceeded 80% and hematopoietic chimerism also was high (40-80% in bone marrow). Importantly, human macrophages (Kupffer cells) were present in the chimeric livers. Double-chimeric FRGN mice will serve as a new model for disease processes that involve interactions between hepatocytes and hematolymphoid cells.


Subject(s)
Hematopoiesis , Hepatocytes/cytology , Animals , Antigens, CD/metabolism , Chimerism , Female , Hepatocytes/transplantation , Humans , Hydrolases/deficiency , Hydrolases/genetics , Hydrolases/metabolism , Immunohistochemistry , Liver/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Serum Albumin/metabolism
5.
Gene Ther ; 20(6): 658-69, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23076379

ABSTRACT

In vivo delivery is a major barrier to the use of molecular tools for gene modification. Here we demonstrate site-specific gene editing of human cells in vivo in hematopoietic stem cell-engrafted NOD.Cg-Prkdc(scid)IL2rγ(tm1Wjl) (abbreviated NOD-scid IL2rγ(null)) mice, using biodegradable nanoparticles loaded with triplex-forming peptide nucleic acids (PNAs) and single-stranded donor DNA molecules. In vitro screening showed greater efficacy of nanoparticles containing PNAs/DNAs together over PNA-alone or DNA-alone. Intravenous injection of particles containing PNAs/DNAs produced modification of the human CCR5 gene in hematolymphoid cells in the mice, with modification confirmed at the genomic DNA, mRNA and functional levels. Deep sequencing revealed in vivo modification of the CCR5 gene at frequencies of 0.43% in hematopoietic cells in the spleen and 0.05% in the bone marrow: off-target modification in the partially homologous CCR2 gene was two orders of magnitude lower. We also induced specific modification in the ß-globin gene using nanoparticles carrying ß-globin-targeted PNAs/DNAs, demonstrating this method's versatility. In vivo testing in an enhanced green fluorescent protein-ß-globin reporter mouse showed greater activity of nanoparticles containing PNAs/DNAs together over DNA only. Direct in vivo gene modification, such as we demonstrate here, would allow for gene therapy in systemic diseases or in cells that cannot be manipulated ex vivo.


Subject(s)
DNA/genetics , Gene Targeting , Gene Transfer Techniques , Nanoparticles/chemistry , Peptide Nucleic Acids/genetics , Animals , Cell Line , DNA/administration & dosage , DNA/chemistry , Genetic Therapy , Hematopoietic Stem Cells/cytology , Humans , Mice , Nanoparticles/administration & dosage , Peptide Nucleic Acids/administration & dosage , Peptide Nucleic Acids/chemistry , Receptors, CCR5/genetics
6.
Am J Transplant ; 10(8): 1738-48, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20659086

ABSTRACT

Viral infections have been associated with the rejection of transplanted allografts in humans and mice, and the induction of tolerance to allogeneic tissues in mice is abrogated by an ongoing viral infection and inhibited in virus-immune mice. One proposed mechanism for this 'heterologous immunity' is the induction of alloreactive T cell responses that cross-react with virus-derived antigens. These cross-reactive CD8 T cells are generated during acute viral infection and survive into memory, but their ability to partake in the immune response to allografts in vivo is not known. We show here that cross-reactive, virus-specific memory CD8 T cells from mice infected with LCMV proliferated in response to allografts. CD8 T cells specific to several LCMV epitopes proliferated in response to alloantigens, with the magnitude and hierarchy of epitope-specific responses varying with the private specificities of the host memory T cell repertoire, as shown by adoptive transfer studies. Last, we show that purified LCMV-specific CD8 T cells rejected skin allografts in SCID mice. These findings therefore implicate a potential role for heterologous immunity in virus-induced allograft rejection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Graft Rejection/virology , Lymphocytic choriomeningitis virus/immunology , Skin Transplantation/immunology , Adoptive Transfer , Animals , Epitopes, T-Lymphocyte/immunology , Isoantigens/immunology , Mice
7.
Cell Death Differ ; 17(5): 774-86, 2010 May.
Article in English | MEDLINE | ID: mdl-19911006

ABSTRACT

Endoplasmic reticulum (ER) stress-mediated cell death has an important role in the pathogenesis of chronic diseases, including diabetes and neurodegeneration. Although proapoptotic programs activated by ER stress have been extensively studied, identification and characterization of antiapoptotic programs that counteract ER stress are currently incomplete. Through the gene expression profiling of beta-cells lacking Wolfram syndrome 1 gene (WFS1), a causative gene for Wolfram syndrome, we discovered a novel antiapoptotic gene of the unfolded protein response (UPR), apoptosis antagonizing transcription factor (AATF). Here, we study the regulation of AATF, identify its target genes, and determine the basis for its antiapoptotic activities in response to ER stress. We show that AATF is induced by ER stress through the PERK-eIF2alpha pathway and transcriptionally activates the v-akt murine thymoma viral oncogene homolog 1 (AKT1) gene through signal transducer and activator of transcription 3 (Stat3), which sustains Akt1 activation and promotes cell survival. Ectopic expression of AATF or a constitutively active form of AKT1 confers on cells resistance to ER stress-mediated cell death, whereas RNAi-mediated knockdown of AATF or AKT1 renders cells sensitive to ER stress. We also discovered a positive crosstalk between the AATF and WFS1 signaling pathways. Thus, WFS1 deficiency or AATF deficiency mediates a self-perpetuating cycle of cell death. Our results reveal a novel antiapoptotic program relevant to the treatment of diseases caused by ER stress-mediated cell death.


Subject(s)
Apoptosis/physiology , Proto-Oncogene Proteins c-akt/genetics , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , Chromatin Immunoprecipitation , Humans , Immunoblotting , Immunoprecipitation , In Situ Nick-End Labeling , Lentivirus , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Nuclear Proteins , Oligonucleotide Array Sequence Analysis , Protein Binding/genetics , Protein Binding/physiology , Proto-Oncogene Proteins c-akt/metabolism , Rats , STAT3 Transcription Factor/metabolism , Transcription Factors/genetics
8.
Clin Exp Immunol ; 157(1): 104-18, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19659776

ABSTRACT

Immunodeficient non-obese diabetic (NOD)-severe combined immune-deficient (scid) mice bearing a targeted mutation in the gene encoding the interleukin (IL)-2 receptor gamma chain gene (IL2rgamma(null)) engraft readily with human peripheral blood mononuclear cells (PBMC). Here, we report a robust model of xenogeneic graft-versus-host-like disease (GVHD) based on intravenous injection of human PBMC into 2 Gy conditioned NOD-scid IL2rgamma(null) mice. These mice develop xenogeneic GVHD consistently (100%) following injection of as few as 5 x 10(6) PBMC, regardless of the PBMC donor used. As in human disease, the development of xenogeneic GVHD is highly dependent on expression of host major histocompatibility complex class I and class II molecules and is associated with severely depressed haematopoiesis. Interrupting the tumour necrosis factor-alpha signalling cascade with etanercept, a therapeutic drug in clinical trials for the treatment of human GVHD, delays the onset and progression of disease. This model now provides the opportunity to investigate in vivo mechanisms of xenogeneic GVHD as well as to assess the efficacy of therapeutic agents rapidly.


Subject(s)
Graft vs Host Disease/immunology , Interleukin Receptor Common gamma Subunit/genetics , Leukocytes, Mononuclear/transplantation , Major Histocompatibility Complex , Models, Animal , Animals , Etanercept , Female , Graft vs Host Disease/drug therapy , Humans , Immunoglobulin G/therapeutic use , Immunosuppressive Agents/therapeutic use , Injections, Intravenous , Leukocyte Common Antigens/analysis , Leukocytes, Mononuclear/immunology , Lymphocyte Culture Test, Mixed , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, Tumor Necrosis Factor/therapeutic use , Tissue Distribution , Transplantation, Heterologous , Whole-Body Irradiation
9.
Clin Exp Immunol ; 154(2): 270-84, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18785974

ABSTRACT

Immunodeficient hosts engrafted with human lymphohaematopoietic cells hold great promise as a preclinical bridge for understanding human haematopoiesis and immunity. We now describe a new immunodeficient radioresistant non-obese diabetic mice (NOD) stock based on targeted mutations in the recombination activating gene-1 (Rag1(null)) and interleukin (IL)-2 receptor common gamma chain (IL2rgamma(null)), and compare its ability to support lymphohaematopoietic cell engraftment with that achieved in radiosensitive NOD.CB17-Prkdc(scid) (NOD-Prkdc(scid)) IL2rgamma(null) mice. We observed that immunodeficient NOD-Rag1(null) IL2rgamma(null) mice tolerated much higher levels of irradiation conditioning than did NOD-Prkdc(scid) IL2rgamma(null) mice. High levels of human cord blood stem cell engraftment were observed in both stocks of irradiation-conditioned adult mice, leading to multi-lineage haematopoietic cell populations and a complete repertoire of human immune cells, including human T cells. Human peripheral blood mononuclear cells also engrafted at high levels in unconditioned adult mice of each stock. These data document that Rag1(null) and scid stocks of immunodeficient NOD mice harbouring the IL2rgamma(null) mutation support similar levels of human lymphohaematopoietic cell engraftment. NOD-Rag1(null) IL2rgamma(null) mice will be an important new model for human lymphohaematopoietic cell engraftment studies that require radioresistant hosts.


Subject(s)
Cord Blood Stem Cell Transplantation , Disease Models, Animal , Interleukin Receptor Common gamma Subunit/deficiency , Peripheral Blood Stem Cell Transplantation , Radiation Tolerance/immunology , Animals , Bone Marrow/immunology , Graft Survival/immunology , Humans , Immunophenotyping , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Radiation Tolerance/genetics , Spleen/immunology , Thymus Gland/immunology , Transplantation, Heterologous
10.
Diabetologia ; 51(8): 1449-56, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18563383

ABSTRACT

AIMS/HYPOTHESIS: To develop and validate a new immunodeficient mouse strain that spontaneously develops a non-autoimmune hyperglycaemia to serve as a diabetic host for human islets and human beta stem and progenitor cells without the need for induction of hyperglycaemia by toxic chemicals with their associated side effects. METHODS: We generated and characterised a new strain of immunodeficient spontaneously hyperglycaemic mice, the NOD-Rag1null Prf1null Ins2Akita strain and compared this strain with the NOD-scid Il2rgammanull (also known as Il2rg) immunodeficient strain rendered hyperglycaemic by administration of a single dose of streptozotocin. Hyperglycaemic mice were transplanted with human islets ranging from 1,000 to 4,000 islet equivalents (IEQ) and were monitored for normalisation of blood glucose levels. RESULTS: NOD-Rag1null Prf1null Ins2Akita mice developed spontaneous hyperglycaemia, similar to Ins2Akita-harbouring strains of immunocompetent mice. Histological examination of islets in the host pancreas validated the spontaneous loss of beta cell mass in the absence of mononuclear cell infiltration. Human islets transplanted into spontaneously diabetic NOD-Rag1null Prf1null Ins2Akita and chemically diabetic NOD-scid Il2rgammanull mice resulted in a return to euglycaemia that occurred with transplantation of similar beta cell masses. CONCLUSIONS/INTERPRETATION: The NOD-Rag1null Prf1null Ins2Akita mouse is the first immunodeficient, spontaneously hyperglycaemic mouse strain described that is based on the Ins2Akita mutation. This strain is suitable as hosts for human islet and human beta stem and progenitor cell transplantation in the absence of the need for pharmacological induction of diabetes. This strain of mice also has low levels of innate immunity and can be engrafted with a human immune system for the study of human islet allograft rejection.


Subject(s)
Hyperglycemia/genetics , Insulin-Secreting Cells/physiology , Islets of Langerhans Transplantation , Islets of Langerhans/physiology , Mutation , Animals , Cord Blood Stem Cell Transplantation , Disease Models, Animal , Humans , Mice , Mice, Inbred NOD , Receptors, Interleukin-2/genetics , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Transplantation, Heterologous
11.
Curr Top Microbiol Immunol ; 324: 25-51, 2008.
Article in English | MEDLINE | ID: mdl-18481451

ABSTRACT

There is a growing need for effective animal models to carry out experimental studies on human hematopoietic and immune systems without putting individuals at risk. Progress in development of small animal models for the in vivo investigation of human hematopoiesis and immunity has seen three major breakthroughs over the last three decades. First, CB 17-Prkdc(scid) (abbreviated CB 17-scid) mice were discovered in 1983, and engraftment of these mice with human fetal tissues (SCID-Hu model) and peripheral blood mononuclear cells (Hu-PBL-SCID model) was reported in 1988. Second, NOD-scid mice were developed and their enhanced ability to engraft with human hematolymphoid tissues as compared with CB17-scid mice was reported in 1995. NOD-scid mice have been the "gold standard" for studies of human hematolymphoid engraftment in small animal models over the last 10 years. Third, immunodeficient mice bearing a targeted mutation in the IL-2 receptor common gamma chain (IL2rgamma(null)) were developed independently by four groups between 2002 and 2005, and a major increase in the engraftment and function of human hematolymphoid cells as compared with NOD-scid mice has been reported. These new strains of immunodeficient IL2rgamma(null) mice are now being used for studies in human hematopoiesis, innate and adaptive immunity, autoimmunity, infectious diseases, cancer biology, and regenerative medicine. In this chapter, we discuss the current state of development of these strains of mice, the remaining deficiencies, and how approaches used to increase the engraftment and function of human hematolymphoid cells in CB 17-scid mice and in previous models based on NOD-scid mice may enhance human hematolymphoid engraftment and function in NOD-scid IL2rgamma(null) mice.


Subject(s)
Biomedical Research/methods , Disease Models, Animal , Animals , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Mice , Mice, Inbred NOD , Mice, SCID
12.
Am J Transplant ; 7(7): 1710-21, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17564635

ABSTRACT

Regulatory T cells (Treg) are important in peripheral tolerance, but their role in establishing and maintaining hematopoietic mixed chimerism and generating central tolerance is unclear. We now show that costimulation blockade using a donor-specific transfusion and anti-CD154 antibody applied to mice given bone marrow and simultaneously transplanted with skin allografts leads to hematopoietic chimerism and permanent skin allograft survival. Chimeric mice bearing intact skin allografts fail to generate effector/memory T cells against allogeneic targets as shown by the absence of IFNgamma-producing CD44(high)CD8+ T cells and in vivo cytotoxicity. Depletion of Tregs by injection of anti-CD4 or anti-CD25 antibody prior to costimulation blockade prevents chimerism, shortens skin allograft survival and leads to generation of effector/memory cytotoxic T cells. Depletion of Tregs by injection of anti-CD4 or anti-CD25 antibody two months after transplantation leads to loss of skin allografts even though mice remain chimeric and exhibit little in vivo cytotoxicity. In contrast, chimerism is lost, but skin allografts survive following naïve T-cell injection. We conclude that hematopoietic chimerism and peripheral tolerance may be maintained by different mechanisms in mixed hematopoietic chimeras.


Subject(s)
Bone Marrow Transplantation/immunology , Hematopoiesis , Immune Tolerance , Skin Transplantation/immunology , Transplantation Chimera , Transplantation Immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred CBA , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous
13.
Am J Transplant ; 6(5 Pt 1): 894-902, 2006 May.
Article in English | MEDLINE | ID: mdl-16611325

ABSTRACT

Costimulatory signals regulate T-cell activation. To investigate the role of costimulation in autoimmunity and transplantation, we studied the BB rat model of type 1 diabetes. Diabetes-prone BB (BBDP) rats spontaneously develop disease when 55-120 days of age. We observed that two anti-CD28 monoclonal antibodies (mAb) with different functional activities completely prevented diabetes in BBDP rats. Anti-CD154 mAb delayed diabetes, whereas treatment with CTLA4-Ig or anti-CD80 mAb accelerated disease. Anti-CD86 or anti-CD134L mAbs had no effect. Diabetes resistant BB (BBDR) rats are disease-free, but >95% of them develop diabetes after treatment with polyinosinic-polycytidylic acid and an mAb that depletes Treg cells. In the induced BBDR model, anti-CD154 mAb delayed onset of diabetes, whereas CTLA4-Ig, anti-CD134L or either of the anti-CD28 mAbs had little or no effect. In contrast, blockade of the CD134-CD134L pathway was highly effective for preventing autoimmune recurrence against syngeneic islet grafts in diabetic BBDR hosts. Blockade of the CD40-CD154 pathway was also effective, but less so. These data suggest that the effectiveness of costimulation blockade in the treatment of type 1 diabetes is dependent on both the costimulatory pathway targeted and the mechanism of induction, stage, intensity and duration of the pathogenic process.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/surgery , Islets of Langerhans Transplantation/immunology , Animals , CD28 Antigens/immunology , Diabetes Mellitus, Type 1/prevention & control , Disease Models, Animal , Immune Tolerance , Rats , Rats, Inbred BB , Recurrence
14.
Clin Immunol ; 109(2): 229-37, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14597222

ABSTRACT

The repeated administration of mercury to Brown Norway (BN) rats induces the production of autoantibodies to laminin 1 and other autoantigens, accompanied by renal deposition of immunoglobulins and a membranous glomerulonephropathy. A graft-versus-host-like (GVHL) syndrome, characterized by widespread necrotizing leukocytoclastic vasculitis of the bowel, skin, and other tissues, has also been observed after mercury treatment of BN rats. These findings have suggested that the autoimmunity caused by the administration of mercury to BN rats may result as a xenobiotic-induced GVHL effect under the control of OX22+ T lymphocytes. However, previous studies of mercury-induced autoimmunity have never reported any evidence of GVHL lesions. Therefore, we have carefully examined various tissues from a large group of BN rats injected with HgCl(2) to identify possible areas of inflammatory reactions that may have been unnoticed in previous investigations. In addition, we have determined by flow cytometry whether exposure to mercury results in percentage and numerical alterations of OX22+ or other lymphocyte subpopulations in lymphoid organs of HgCl(2)-treated BN rats. The present article confirms that mercury induces autoimmune responses to laminin 1 but does not corroborate the hypothesis of a GVHL syndrome regulated by OX22+ lymphocytes. First, changes in OX22+ cells during treatment with HgCl(2) were infrequent and had no significant correlation with the kinetics of autoimmune responses to laminin 1. Second, we detected no GVHL lesions in skin and intestine of mercury-treated BN rats.


Subject(s)
Autoantibodies/immunology , Graft vs Host Disease/immunology , Laminin/immunology , Mercuric Chloride/toxicity , Animals , Autoantibodies/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique, Direct , Glomerulonephritis, Membranous/immunology , Glomerulonephritis, Membranous/pathology , Graft vs Host Disease/pathology , Kidney/immunology , Kidney/pathology , Lymph Nodes/immunology , Male , Mercuric Chloride/immunology , Rats , Rats, Inbred BN , Spleen/immunology , Thymus Gland/immunology
15.
J Immunol ; 167(11): 6623-30, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11714833

ABSTRACT

Treatment of mice with a single donor-specific transfusion plus a brief course of anti-CD154 mAb uniformly induces donor-specific transplantation tolerance characterized by the deletion of alloreactive CD8+ T cells. Survival of islet allografts in treated mice is permanent, but skin grafts eventually fail unless recipients are thymectomized. To analyze the mechanisms underlying tolerance induction, maintenance, and failure in euthymic mice we created a new analytical system based on allo-TCR-transgenic hemopoietic chimeric graft recipients. Chimeras were CBA (H-2(k)) mice engrafted with small numbers of syngeneic TCR-transgenic KB5 bone marrow cells. These mice subsequently circulated a self-renewing trace population of anti-H-2(b)-alloreactive CD8+ T cells maturing in a normal microenvironment. With this system, we studied the maintenance of H-2(b) allografts in tolerized mice. We documented that alloreactive CD8+ T cells deleted during tolerance induction slowly returned toward pretreatment levels. Skin allograft rejection in this system occurred in the context of 1) increasing numbers of alloreactive CD8+ cells; 2) a decline in anti-CD154 mAb concentration to levels too low to inhibit costimulatory functions; and 3) activation of the alloreactive CD8+ T cells during graft rejection following deliberate depletion of regulatory CD4+ T cells. Rejection of healed-in allografts in tolerized mice appears to be a dynamic process dependent on the level of residual costimulation blockade, CD4+ regulatory cells, and activated alloreactive CD8+ thymic emigrants that have repopulated the periphery after tolerization.


Subject(s)
Mice, Inbred CBA/genetics , Mice, Inbred CBA/immunology , Models, Immunological , Radiation Chimera/immunology , Skin Transplantation/immunology , Skin Transplantation/methods , Transplantation Tolerance/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/blood , Blood Transfusion , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Survival/immunology , Clone Cells , Cricetinae , Female , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/genetics , Graft Survival/immunology , H-2 Antigens/genetics , Hematopoietic Stem Cells/immunology , Injections, Intravenous , Lymphocyte Activation/genetics , Lymphocyte Depletion , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Radiation Chimera/genetics , T-Lymphocyte Subsets/immunology
16.
Autoimmunity ; 33(3): 199-211, 2001 May.
Article in English | MEDLINE | ID: mdl-11683379

ABSTRACT

ART2a and ART2b are isoenzymes expressed on the surface of mature T cells and intraepithelial lymphocytes (IELs) in the rat. They exhibit both adenosine diphosphoribosyltransferase and nicotine adenine dinucleotide (NAD) glycohydrolase activities, and both can generate a transmembrane signal that modulates T cell activation. The presence or absence of ART2+ T cells modulates the expression of autoimmune diabetes in the BB rat. ART2 also circulates in a soluble form whose function is unknown. We tested the hypothesis that circulating ART2 protein regulates the expression of autoimmunity. We compared the kinetics, regulation, and source of soluble ART2 in normal rats and in rats with autoimmune diabetes. Basal levels of soluble ART2 varied greatly among strains of rats and were lowest in the diabetes-prone BB (BBDP/Wor) rat. In diabetes-resistant BB (BBDR/Wor) rats, administration of anti-ART2a antibody, which is known to induce diabetes, resulted in transient clearing of soluble ART2a that was followed rapidly by a rebound increase. Repeated treatment of BBDR/Wor rats with anti-ART2a antibody resulted in sustained supraphysiologic levels of soluble ART2a. Although the number of peripheral ART2a+ T cells is known to correlate with the expression of diabetes in BBDR/Wor rats, the level of soluble ART2a protein did not. The source of the soluble ART2 protein in the rat appeared to be the gut. The results suggest that ART2+ T cells and soluble ART2 protein may subserve different immunomodulatory functions.


Subject(s)
ADP Ribose Transferases , Diabetes Mellitus, Type 1/immunology , Histocompatibility Antigens/metabolism , Membrane Glycoproteins , Animals , Antibodies, Monoclonal/administration & dosage , Antibody Specificity , Antigens, Differentiation, T-Lymphocyte , Autoimmunity , CD5 Antigens/metabolism , CD8 Antigens/metabolism , Diabetes Mellitus, Type 1/enzymology , Female , Isoenzymes/antagonists & inhibitors , Isoenzymes/immunology , Isoenzymes/metabolism , Male , NAD+ Nucleosidase/antagonists & inhibitors , NAD+ Nucleosidase/immunology , NAD+ Nucleosidase/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/immunology , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Inbred BB , Rats, Inbred Strains , Rats, Nude , Solubility , Species Specificity , T-Lymphocytes/immunology
17.
J Immunol ; 167(7): 3600-9, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11564772

ABSTRACT

The intestinal lymphoid compartment of the rat is large and diverse, but the phenotype and functions of its constituent cell populations are not fully characterized. Using new methodology for the isolation and purification of rat intestinal intraepithelial lymphocytes (IELs), we previously identified a population of alphabeta- and gammadelta-TCR- NKR-P1A+ NK cells. These cells were almost completely restricted to the CD4-CD8- IEL population, and unlike peripheral NK cells in the rat, they were CD2-. We now report that rat intraepithelial NK (IENK) and peripheral NK cells are similar in morphology, in their ability to lyse NK-sensitive targets, and in their ability to suppress a one-way mixed lymphocyte culture. In contrast, however, intraepithelial and splenic NK cells differ markedly in two respects. First, IENK cells express high levels of ADP-ribosyltransferase 2 (a marker of regulatory T cells in the rat) and CD25, whereas peripheral NK cells do not. Second, unlike splenic NK cells, a substantial fraction of IENK cells appear to spontaneously secrete IL-4 and/or IFN-gamma. We conclude that the rat IEL compartment harbors a large population of NKR-P1A+CD3- cells that function as NK cells but display an activated phenotype and unusual cytokine profile that clearly distinguish them from splenic NK cells. Their phenotypic and functional characteristics suggest that these distinctive IENK cells may participate in the regulation of mucosal immunity.


Subject(s)
Immunity, Mucosal , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Intestinal Mucosa/immunology , Killer Cells, Natural/immunology , Lectins, C-Type , Aging , Animals , Antigens, Surface/analysis , CD3 Complex/analysis , Cells, Cultured , Cytotoxicity Tests, Immunologic , Female , Immunophenotyping , Killer Cells, Natural/classification , Killer Cells, Natural/cytology , Lymphocyte Culture Test, Mixed , Male , NK Cell Lectin-Like Receptor Subfamily B , Rats , Rats, Wistar , Spleen/immunology , Tumor Cells, Cultured
18.
J Immunol ; 167(4): 2049-59, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11489987

ABSTRACT

The presence of NAD-metabolizing enzymes (e.g., ADP-ribosyltransferase (ART)2) on the surface of immune cells suggests a potential immunomodulatory activity for ecto-NAD or its metabolites at sites of inflammation and cell lysis where extracellular levels of NAD may be high. In vitro, NAD inhibits mitogen-stimulated rat T cell proliferation. To investigate the mechanism of inhibition, the effects of NAD and its metabolites on T cell proliferation were studied using ART2a+ and ART2b+ rat T cells. NAD and ADP-ribose, but not nicotinamide, inhibited proliferation of mitogen-activated T cells independent of ART2 allele-specific expression. Inhibition by P2 purinergic receptor agonists was comparable to that induced by NAD and ADP-ribose; these compounds were more potent than P1 agonists. Analysis of the NAD-metabolizing activity of intact rat T cells demonstrated that ADP-ribose was the predominant metabolite, consistent with the presence of cell surface NAD glycohydrolase (NADase) activities. Treatment of T cells with phosphatidylinositol-specific phospholipase C removed much of the NADase activity, consistent with at least one NADase having a GPI anchor; ART2- T cell subsets contained NADase activity that was not releasable by phosphatidylinositol-specific phospholipase C treatment. Formation of AMP from NAD and ADP-ribose also occurred, a result of cell surface pyrophosphatase activity. Because AMP and its metabolite, adenosine, were less inhibitory to rat T cell proliferation than was NAD or ADP-ribose, pyrophosphatases may serve a regulatory role in modifying the inhibitory effect of ecto-NAD on T cell activation. These data suggest that T cells express multiple NAD and adenine nucleotide-metabolizing activities that together modulate immune function.


Subject(s)
ADP Ribose Transferases , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacology , Lymphocyte Activation , Membrane Glycoproteins , NAD+ Nucleosidase/metabolism , NAD/physiology , Pyrophosphatases/metabolism , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Adenosine/metabolism , Adenosine/pharmacology , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/pharmacology , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology , Animals , Antigens, Differentiation, T-Lymphocyte , Cell Membrane/enzymology , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Cholera Toxin/pharmacology , Female , Histocompatibility Antigens/biosynthesis , Lymphocyte Activation/drug effects , Male , Mitogens/pharmacology , NAD/metabolism , NAD+ Nucleosidase/physiology , Pertussis Toxin , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Phosphorus Radioisotopes/metabolism , Poly(ADP-ribose) Polymerases/biosynthesis , Pyrophosphatases/physiology , Rats , Rats, Inbred BB , Rats, Inbred WF , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Type C Phospholipases/pharmacology , Virulence Factors, Bordetella/pharmacology
19.
Clin Immunol ; 100(2): 134-43, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11465941

ABSTRACT

Type 1 diabetes in humans is a serious autoimmune disorder of children that is still poorly understood, unpreventable, and irreversible. Study of its animal models, notably the NOD mouse and BB rat, has generated a wealth of information concerning genetics and immunopathogenesis, but that information has still not altered the way in which we treat children with diabetes. In this review we attempt to identify the most promising avenues of continuing research in these models and the most important issues that must be faced by the designers of human therapies based on the animal dataset.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Disease Models, Animal , Animals , Autoimmunity , Diabetes Mellitus, Type 1/immunology , Humans
20.
Diabetes ; 50(5): 972-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11334440

ABSTRACT

Thymocytes from adult BB rats can adoptively transfer autoimmune diabetes to athymic recipients. It is also known that the development of BB rat T-cells is recapitulated in adult thymus organ cultures (ATOCs). Based on these observations, we tested the hypothesis that cells capable of the adoptive transfer of diabetes would be present in long-term ATOCs but could be rendered nondiabetogenic by co-culture with appropriate antigens. We observed that cells recovered from adult diabetes-resistant BB (BBDR) rat thymi cultured for up to 14 days can adoptively transfer disease to athymic WAG-rnu/rnu rats treated with polyinosinic: polycytidylic acid and a monoclonal antibody to preclude development of ART2a+ regulatory T-cells. Co-culture of adult BBDR thymi in the presence of BBDR thyrocytes had no effect on the ability of recovered cells to induce diabetes in 70-80% of adoptive recipients. In contrast, co-culture in the presence of islets prevented transfer of diabetes, on average, in >90% of recipients. Fresh islets, frozen islets, and islets pretreated with streptozotocin to deplete insulin were equally effective in preventing diabetes, but none prevented insulitis in nondiabetic recipients. Co-culture in the presence of islets was not associated with detectable alterations in phenotype or in the secretion of gamma-interferon or interleukin-4, either in cultures or in cells recovered from adoptive recipients. We conclude that islet antigens involved in the initiation of autoimmune diabetes in BB rats may be absent or deficient in BB rat thymi. Exposure of ATOCs to exogenous islets may lead to deletion or anergy of diabetogenic T-cells or to the positive selection of regulatory T-cells.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Adoptive Transfer , Animals , Cells, Cultured , Coculture Techniques , Diabetes Mellitus, Type 1/pathology , Immunity, Innate/immunology , Immunophenotyping , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Islets of Langerhans/cytology , Islets of Langerhans/pathology , Lymph Nodes/immunology , Lymphocyte Transfusion , Rats , Rats, Inbred BB , Rats, Nude , Receptors, Transferrin , Spleen/immunology , T-Lymphocytes/cytology , Thymus Gland/cytology , Time Factors , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...