Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cell ; 134(4): 577-86, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18691745

ABSTRACT

Evaluation of the therapeutic potential of RNAi for HIV infection has been hampered by the challenges of siRNA delivery and lack of suitable animal models. Using a delivery method for T cells, we show that siRNA treatment can dramatically suppress HIV infection. A CD7-specific single-chain antibody was conjugated to oligo-9-arginine peptide (scFvCD7-9R) for T cell-specific siRNA delivery in NOD/SCIDIL2rgamma-/- mice reconstituted with human lymphocytes (Hu-PBL) or CD34+ hematopoietic stem cells (Hu-HSC). In HIV-infected Hu-PBL mice, treatment with anti-CCR5 (viral coreceptor) and antiviral siRNAs complexed to scFvCD7-9R controlled viral replication and prevented the disease-associated CD4 T cell loss. This treatment also suppressed endogenous virus and restored CD4 T cell counts in mice reconstituted with HIV+ peripheral blood mononuclear cells. Moreover, scFvCD7-9R could deliver antiviral siRNAs to naive T cells in Hu-HSC mice and effectively suppress viremia in infected mice. Thus, siRNA therapy for HIV infection appears to be feasible in a preclinical animal model.


Subject(s)
HIV Infections/genetics , HIV Infections/therapy , RNA Interference , RNA, Small Interfering/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, CD7/metabolism , Disease Models, Animal , Gene Expression , HIV-1/genetics , HIV-1/metabolism , Humans , Immunoglobulin Fragments/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Viral/metabolism
3.
Curr Protoc ; 4(6): e1073, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38924322

ABSTRACT

Traditional skin sampling methods include punch or shave biopsies to produce a solid tissue sample for analysis. These biopsy procedures are painful, require anesthesia, and leave permanent scars. This unit describes a suction blister skin biopsy method that can be used in place of traditional biopsy methodologies as a minimally invasive, non-scarring skin sampling technique. The induction of suction blisters uses an instrument with a chamber that applies negative pressure and gentle heat to the skin. Blister formation occurs within 1 hr, producing up to five blisters, each 10 mm in diameter per biopsy site. Blister fluid can be extracted and centrifuged to retrieve cells from the epidermis and upper dermis for flow cytometry, single-cell RNA sequencing, cell culture, and more without the need for digestion protocols. In addition, the blister fluid can be used to measure soluble proteins and metabolites. This unit describes the preparation of supplies and subjects, the suction blister biopsy procedure and blister formation, fluid extraction, and post-blistering care. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Preparation of supplies and subject Basic Protocol 2: Suction blister biopsy procedure and formation Basic Protocol 3: Blister fluid extraction Basic Protocol 4: Post-blister care and clean up.


Subject(s)
Blister , Skin , Humans , Blister/pathology , Suction , Biopsy/methods , Skin/pathology , Specimen Handling/methods
4.
J Immunol ; 182(5): 3278-84, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234226

ABSTRACT

Fulminant inflammation in the liver is often accompanied by the accumulation of IFN-gamma-producing T cells. The BALB/c-Tgfb1(-/-) mouse exhibits extensive, spontaneously developing necroinflammation in the liver, accompanied by the accumulation of IFN-gamma-producing CD4(+) and CD8(+) T cells. Liver damage depends on the presence of an intact Ifng gene. We determined the relevant cellular source(s) of IFN-gamma. In Tgfb1(-/-) liver, CD4(+) T cells were more numerous than CD8(+) T cells and NK cells, and produced more IFN-gamma. Depletion of CD4(+) T cells eliminated both the elevation in plasma IFN-gamma and aspartate aminotransferase, whereas depletion of CD8(+) T cells did not. Rag1(-/-)Tgfb1(-/-) mice exhibited neither IFN-gamma elevation nor tissue damage, indicating that NK cells are not sufficient. IFN-gamma was required for strong overexpression of class II genes but not for CD4(+) T cell activation, oligoclonal expansion, or accumulation in the liver. The T cell inhibitory molecule PD-L1 was strongly expressed in Tgfb1(-/-) livers, ruling out a lack of PD-L1 expression as an explanation for aberrant liver T cell activation. Finally, whereas Tgfb1(-/-) CD4(+) T cells overexpressed Fas ligand, hepatocellular damage was observed in Fas(lpr/lpr)Tgfb1(-/-) mice, indicating that liver pathology is Fas independent. We conclude that liver damage in this model of fulminant autoimmune hepatitis is driven by CD4(+) T cell production of IFN-gamma, is independent of both CD8(+) T cells and the Fas ligand/Fas pathway, and is not explained by a lack of PD-L1 expression.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Liver Failure, Acute/immunology , Liver Failure, Acute/pathology , fas Receptor , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Fas Ligand Protein/genetics , Fas Ligand Protein/physiology , Hepatitis, Autoimmune/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/physiology , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Failure, Acute/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Transforming Growth Factor beta1/genetics , fas Receptor/genetics , fas Receptor/physiology
5.
J Immunol ; 182(9): 5547-59, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380803

ABSTRACT

Activation of TLR4 by administration of LPS shortens the survival of skin allografts in mice treated with costimulation blockade through a CD8 T cell-dependent, MyD88-dependent, and type I IFN receptor-dependent pathway. The effect of TLR activation on the establishment of allogeneic hematopoietic chimerism in mice treated with costimulation blockade is not known. Using a costimulation blockade protocol based on a donor-specific transfusion (DST) and a short course of anti-CD154 mAb, we show that LPS administration at the time of DST matures host alloantigen-presenting dendritic cells, prevents the establishment of mixed allogeneic hematopoietic chimerism, and shortens survival of donor-specific skin allografts. LPS mediates its effects via a mechanism that involves both CD4(+) and CD8(+) T cells and results from signaling through either the MyD88 or the type I IFN receptor pathways. We also document that timing of LPS administration is critical, as injection of LPS 24 h before treatment with DST and anti-CD154 mAb does not prevent hematopoietic engraftment but administration the day after bone marrow transplantation does. We conclude that TLR4 activation prevents the induction of mixed allogeneic hematopoietic chimerism through type I IFN receptor and MyD88-dependent signaling, which leads to the up-regulation of costimulatory molecules on host APCs and the generation of alloreactive T cells. These data suggest that distinct but overlapping cellular and molecular mechanisms control the ability of TLR agonists to block tolerance induction to hematopoietic and skin allografts in mice treated with costimulation blockade.


Subject(s)
Chimerism , Hematopoietic Stem Cell Transplantation , Immunosuppression Therapy , Isoantigens/genetics , Lipopolysaccharides/administration & dosage , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Toll-Like Receptors/agonists , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Bone Marrow Transplantation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carrier Proteins/administration & dosage , Graft Survival/genetics , Graft Survival/immunology , Immunosuppression Therapy/methods , Interferon Type I/biosynthesis , Interferon Type I/metabolism , Interferon Type I/physiology , Isoantigens/immunology , Isoantigens/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/physiology , Poly I-C/administration & dosage , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Skin Transplantation/immunology , Toll-Like Receptors/administration & dosage , Toll-Like Receptors/metabolism
6.
8.
Exp Biol Med (Maywood) ; 233(8): 997-1012, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18653783

ABSTRACT

Umbilical cord blood (UCB) is increasingly being used for human hematopoietic stem cell (HSC) transplantation in children but often requires pooling multiple cords to obtain sufficient numbers for transplantation in adults. To overcome this limitation, we have used an ex vivo two-week culture system to expand the number of hematopoietic CD34(+) cells in cord blood. To assess the in vivo function of these expanded CD34(+) cells, cultured human UCB containing 1 x 10(6) CD34(+) cells were transplanted into conditioned NOD-scid IL2rgamma(null) mice. The expanded CD34(+) cells displayed short- and long-term repopulating cell activity. The cultured human cells differentiated into myeloid, B-lymphoid, and erythroid lineages, but not T lymphocytes. Administration of human recombinant TNFalpha to recipient mice immediately prior to transplantation promoted human thymocyte and T-cell development. These T cells proliferated vigorously in response to TCR cross-linking by anti-CD3 antibody. Engrafted TNFalpha-treated mice generated antibodies in response to T-dependent and T-independent immunization, which was enhanced when mice were co-treated with the B cell cytokine BLyS. Ex vivo expanded CD34(+) human UCB cells have the capacity to generate multiple hematopoietic lineages and a functional human immune system upon transplantation into TNFalpha-treated NOD-scid IL2rgamma(null) mice.


Subject(s)
Cord Blood Stem Cell Transplantation , Interleukin Receptor Common gamma Subunit/deficiency , Animals , Antigens, CD34/blood , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Culture Techniques/methods , Female , Hematopoiesis , Humans , Infant, Newborn , Interleukin Receptor Common gamma Subunit/genetics , Lymphocyte Activation , Lymphopoiesis , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Recombinant Proteins/administration & dosage , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transplantation Conditioning , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/administration & dosage
9.
Ann N Y Acad Sci ; 1103: 77-89, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17332083

ABSTRACT

There are many rodent models of autoimmune diabetes that have been used to study the pathogenesis of human type 1 diabetes (T1D), including the non-obese diabetic (NOD) mouse, the biobreeding (BB) rat, and the transgenic mouse models. However, mice and rats are not humans, and these rodent models do not completely recapitulate the autoimmune pathogenesis of the human disease. In addition, many of the reagents, tools, and therapeutics proposed for use in humans may be species specific and cannot be investigated in rodents. Researchers have used nonhuman primates to more closely mimic the human immune system and, to study species-specific therapeutics, but these studies are associated with additional ethical and economic constraints and, to date, no model of autoimmune diabetes in this species has been described. New animal models are needed that will permit the in vivo investigation of human immune systems and analyses of the pathogenesis of human T1D without putting individuals at risk. To fill this need, we are developing humanized mouse models for the in vivo study of T1D. These models are based on our newly generated stock of NOD-scid IL2rgamma(null) mice, which engraft at higher levels with human hematolymphoid cells and exhibit enhanced function of the engrafted human immune systems compared with previous humanized mouse models. Overall, development of these new generations of humanized mice should facilitate in vivo studies of the human immune system as well as permit the investigation of the pathogenesis and effector phases of human T1D.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Animals , Disease Models, Animal , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Reproducibility of Results
10.
Ann N Y Acad Sci ; 1103: 90-3, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17376822

ABSTRACT

The use of "humanized" mice represents an appealing translational model for studies of the pathogenesis of immune-mediated diseases and for the evaluation of potential therapeutics. The utility of humanized mice depends on their ability to model the human immune system with high fidelity, and, in this respect, previous models have fallen short. The recently developed NOD-scid Il2rgamma(null) mouse, however, exhibits greatly enhanced ability to support the engraftment of human peripheral blood mononuclear cells. Herein, we describe the challenges of recapitulating human immunity in humanized mice and features of NOD-scid Il2rgamma(null) mice that help overcome them.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Leukocytes, Mononuclear/immunology , Mice, Inbred NOD/genetics , Mice, SCID/genetics , Animals , Crosses, Genetic , Disease Models, Animal , Humans , Isoantigens/immunology , Mice , Mice, Inbred BALB C
11.
Diabetes ; 52(2): 321-6, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12540603

ABSTRACT

The loss of self-tolerance leading to autoimmune type 1 diabetes in the NOD mouse model involves at least 19 genetic loci. In addition to their genetic defects in self-tolerance, NOD mice resist peripheral transplantation tolerance induced by costimulation blockade using donor-specific transfusion and anti-CD154 antibody. Hypothesizing that these two abnormalities might be related, we investigated whether they could be uncoupled through a genetic approach. Diabetes-resistant NOD and C57BL/6 stocks congenic for various reciprocally introduced Idd loci were assessed for their ability to be tolerized. Surprisingly, in NOD congenic mice that are almost completely protected from diabetes, costimulation blockade failed to prolong skin allograft survival. In reciprocal C57BL/6 congenic mice with NOD-derived Idd loci, skin allograft survival was readily prolonged by costimulation blockade. These data indicate that single or multiple combinations of evaluated Idd loci that dramatically reduce diabetes frequency do not correct resistance to peripheral transplantation tolerance induced by costimulation blockade. We suggest that mechanisms controlling autoimmunity and transplantation tolerance in NOD mice are not completely overlapping and are potentially distinct, or that the genetic threshold for normalizing the transplantation tolerance defect is higher than that for preventing autoimmune diabetes.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Skin Transplantation/immunology , Transplantation Tolerance/genetics , Animals , Antibodies, Monoclonal/therapeutic use , Antibody Specificity , Blood Transfusion , CD40 Ligand/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Genetic Predisposition to Disease , Graft Survival/immunology , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Time Factors
12.
Diabetes ; 53(8): 1972-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277375

ABSTRACT

NOD mice develop type 1 autoimmune diabetes and exhibit genetically dominant resistance to transplantation tolerance induction. These two phenotypes are genetically separable. Costimulation blockade fails to prolong skin allograft survival in (NOD x C57BL/6)F1 mice and in NOD-related strains made diabetes-resistant by congenic introduction of protective major histocompatibility complex (MHC) or non-MHC Idd region genes. Here, we tested the hypothesis that the genetic basis for the resistance of NOD mice to skin allograft tolerance also applies to islet allografts. Surprisingly, costimulation blockade induced permanent islet allograft survival in (NOD x C57BL/6)F1 mice but not in NOD mice. After costimulation blockade, islet allograft survival was prolonged in diabetes-resistant NOD.B6 Idd3 mice and shortened in diabetes-free C57BL/6 mice congenic for the NOD Idd3 variant. Islet allograft tolerance could not be induced in diabetes-resistant NOD.B10 Idd5 and NOD.B10 Idd9 mice. The data demonstrate that 1) NOD mice resist islet allograft tolerance induction; 2) unlike skin allografts, resistance to islet allograft tolerance is a genetically recessive trait; 3) an Idd3 region gene(s) is an important determinant of islet allograft tolerance induction; and 4) there may be overlap in the mechanism by which the Idd3 resistance locus improves self-tolerance and the induction of allotolerance.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Graft Survival/physiology , Islets of Langerhans Transplantation/physiology , Alleles , Animals , CD40 Ligand/immunology , Combined Modality Therapy , Diabetes Mellitus, Type 1/surgery , Genetic Variation , Immunotherapy , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Transplantation, Homologous
13.
Transplantation ; 78(11): 1601-8, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15591948

ABSTRACT

BACKGROUND: Treatment with anti-CD154 monoclonal antibody (mAb) plus a donor-specific transfusion (DST) of spleen cells prolongs skin allograft survival in mice through a mechanism involving deletion of host alloreactive CD8(+) T cells. It is unknown if other lymphohematopoietic cell populations can be used as a DST. METHODS: Murine recipients of allogeneic skin grafts on day 0 were either untreated or given a DST on day -7 plus 4 doses of anti-CD154 mAb on days -7, -4, 0, and +4. Deletion of CD8(+) alloreactive cells was measured using "synchimeric" CBA recipients, which circulate trace populations of TCR transgenic alloreactive CD8(+) T cells. RESULTS: Transfusion of splenocytes, thymocytes, lymph node cells, or buffy coat cells led to prolonged skin allograft survival in recipients treated with anti-CD154 mAb. In contrast, bone marrow DST failed to delete host alloreactive CD8(+) T cells and was associated with brief skin allograft survival. Transfusions consisting of bone marrow-derived dendritic cells or a mixture of splenocytes and bone marrow cells were also ineffective. CONCLUSIONS: Donor-specific transfusions of splenocytes, thymocytes, lymph node cells, or buffy coat cells can prolong skin allograft survival in recipients treated with costimulation blockade. Bone marrow cells fail to serve this function, in part by failing to delete host alloreactive CD8(+) T cells, and they may actively interfere with the function of a spleen cell DST. The data suggest that transplantation tolerance induction protocols that incorporate bone marrow cells to serve as a DST may not be effective.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Blood Transfusion , Bone Marrow Cells/physiology , CD40 Ligand/physiology , Graft Survival , Skin Transplantation , Animals , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/transplantation , Lymphocyte Depletion , Metrizamide/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Spleen/cytology , Transplantation, Homologous
14.
Ann N Y Acad Sci ; 1005: 148-56, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14679049

ABSTRACT

Recent advances in islet transplantation have enabled physicians to cure type 1 autoimmune diabetes, but at the cost of lifelong immunosuppression with its attendant side effects and long-term health risks. To eliminate the need for immunosuppression, researchers have developed methods for inducing tolerance to transplanted allografts. Tolerance-based transplantation using costimulation blockade has proven remarkably successful in many animal model systems. The most widely used animal model system for studying islet transplantation in type 1-like autoimmune diabetes is the NOD mouse. Unfortunately, this strain has proven resistant to costimulation blockade-based transplantation tolerance protocols that are successful in chemically diabetic mice given islet grafts. It has been assumed that resistance to transplantation tolerance in the NOD mouse is (1) related to autoimmunity directed against its pancreatic beta cells, (2) a consequence of that autoimmunity, and (3) under the control of the same genes that control autoimmunity. In this review, we provide arguments to challenge these assumptions. We describe a new animal model and a new conceptual framework based on data indicating that the mechanisms responsible for resistance to transplantation tolerance and beta cell autoimmunity are not identical. We believe that the recent discoveries we describe will have important implications for the development of tolerance-based transplantation therapies and their translation from the laboratory to the clinic.


Subject(s)
Immune Tolerance , Islets of Langerhans Transplantation , Islets of Langerhans/immunology , Animals , Mice , Mice, Inbred NOD
15.
PLoS One ; 7(11): e49213, 2012.
Article in English | MEDLINE | ID: mdl-23155466

ABSTRACT

Despite increasing evidence that autoreactive CD8 T-cells are involved in both the initiation of type 1 diabetes (T1D) and the destruction of beta-cells, direct evidence for their destructive role in-vivo is lacking. To address a destructive role for autoreactive CD8 T-cells in human disease, we assessed the pathogenicity of a CD8 T-cell clone derived from a T1D donor and specific for an HLA-A2-restricted epitope of islet-specific glucose-6-phosphatase catalytic-subunit related protein (IGRP). HLA-A2/IGRP tetramer staining revealed a higher frequency of IGRP-specific CD8 T-cells in the peripheral blood of recent onset human individuals than of healthy donors. IGRP(265-273)-specific CD8 T-cells that were cloned from the peripheral blood of a recent onset T1D individual were shown to secrete IFNγ and Granzyme B after antigen-specific activation and lyse HLA-A2-expressing murine islets in-vitro. Lytic capacity was also demonstrated in-vivo by specific killing of peptide-pulsed target cells. Using the HLA-A2 NOD-scid IL2rγ(null) mouse model, HLA-A2-restricted IGRP-specific CD8 T-cells induced a destructive insulitis. Together, this is the first evidence that human HLA-restricted autoreactive CD8 T-cells target HLA-expressing beta-cells in-vivo, demonstrating the translational value of humanized mice to study mechanisms of disease and therapeutic intervention strategies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , HLA-A2 Antigen/immunology , Insulin-Secreting Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Epitopes/immunology , Epitopes/metabolism , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred NOD , Mice, Transgenic , T-Lymphocytes, Cytotoxic/metabolism
16.
PLoS One ; 4(10): e7251, 2009 Oct 05.
Article in English | MEDLINE | ID: mdl-19802382

ABSTRACT

BACKGROUND: The lack of a suitable animal model to study viral and immunological mechanisms of human dengue disease has been a deterrent to dengue research. METHODOLOGY/PRINCIPAL FINDINGS: We sought to establish an animal model for dengue virus (DENV) infection and immunity using non-obese diabetic/severe combined immunodeficiency interleukin-2 receptor gamma-chain knockout (NOD-scid IL2rgamma(null)) mice engrafted with human hematopoietic stem cells. Human CD45(+) cells in the bone marrow of engrafted mice were susceptible to in vitro infection using low passage clinical and established strains of DENV. Engrafted mice were infected with DENV type 2 by different routes and at multiple time points post infection, we detected DENV antigen and RNA in the sera, bone marrow, spleen and liver of infected engrafted mice. Anti-dengue IgM antibodies directed against the envelope protein of DENV peaked in the sera of mice at 1 week post infection. Human T cells that developed following engraftment of HLA-A2 transgenic NOD-scid IL2rgamma(null) mice with HLA-A2(+) human cord blood hematopoietic stem cells, were able to secrete IFN-gamma, IL-2 and TNF-alpha in response to stimulation with three previously identified A2 restricted dengue peptides NS4b 2353((111-119)), NS4b 2423((181-189)), and NS4a 2148((56-64)). CONCLUSIONS/SIGNIFICANCE: This is the first study to demonstrate infection of human cells and functional DENV-specific T cell responses in DENV-infected humanized mice. Overall, these mice should be a valuable tool to study the role of prior immunity on subsequent DENV infections.


Subject(s)
Dengue Virus/immunology , Fetal Blood/virology , HLA-A2 Antigen/immunology , Hematopoiesis , Hematopoietic Stem Cells/virology , Interleukin-2/immunology , Mice, SCID/immunology , Animals , Dengue/immunology , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Peptides/chemistry , RNA/metabolism
17.
Diabetes ; 58(1): 165-73, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18984741

ABSTRACT

OBJECTIVE: NOD mice model human type 1 diabetes and are used to investigate tolerance induction protocols for islet transplantation in a setting of autoimmunity. However, costimulation blockade-based tolerance protocols have failed in prolonging islet allograft survival in NOD mice. RESEARCH DESIGN AND METHODS: To investigate the underlying mechanisms, we studied the ability of costimulation blockade to prolong islet allograft survival in congenic NOD mice bearing insulin-dependent diabetes (Idd) loci that reduce the frequency of diabetes. RESULTS: The frequency of diabetes is reduced in NOD.B6 Idd3 mice and is virtually absent in NOD.B6/B10 Idd3 Idd5 mice. Islet allograft survival in NOD.B6 Idd3 mice treated with costimulation blockade is prolonged compared with NOD mice, and in NOD.B6/B10 Idd3 Idd5, mice islet allograft survival is similar to that achieved in C57BL/6 mice. Conversely, some Idd loci were not beneficial for the induction of transplantation tolerance. Alloreactive CD8 T-cell depletion in (NOD x CBA)F1 mice treated with costimulation blockade was impaired compared with similarly treated (C57BL/6.H2(g7) x CBA)F1 mice. Injection of exogenous interleukin (IL)-2 into NOD mice treated with costimulation prolonged islet allograft survival. NOD.B6 Idd3 mice treated with costimulation blockade deleted alloreactive CD8 T-cells and exhibited prolonged islet allograft survival. CONCLUSIONS: Il2 is the Idd3 diabetes susceptibility gene and can influence the outcome of T-cell deletion and islet allograft survival in mice treated with costimulation blockade. These data suggest that Idd loci can facilitate induction of transplantation tolerance by costimulation blockade and that IL-2/Idd3 is a critical component in this process.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Islets of Langerhans/immunology , Animals , Antibodies, Monoclonal/pharmacology , CD40 Ligand/immunology , Cytotoxicity, Immunologic/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/surgery , Flow Cytometry , Graft Survival/drug effects , Graft Survival/genetics , Islets of Langerhans Transplantation/methods , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Mice , Mice, Congenic , Mice, Inbred C3H , Mice, Inbred NOD , Transplantation, Homologous
18.
Curr Protoc Immunol ; Chapter 15: 15.21.1-15.21.21, 2008 May.
Article in English | MEDLINE | ID: mdl-18491294

ABSTRACT

"Humanized" mice are a promising translational model for studying human hematopoiesis and immunity. Their utility has been enhanced by the development of new stocks of immunodeficient hosts, most notably mouse strains such as NOD-scid IL2rgamma(null) mice that lack the IL-2 receptor common gamma chain. These stocks of mice lack adaptive immune function, display multiple defects in innate immunity, and support heightened levels of human hematolymphoid engraftment. Humanized mice can support studies in many areas of immunology, including autoimmunity, transplantation, infectious diseases, and cancer. These models are particularly valuable in experimentation where there is no appropriate small animal model of the human disease, as in the case of certain viral infections. This unit details the creation of humanized mice by engraftment of immunodeficient mice with hematopoietic stem cells or peripheral blood mononuclear cells, provides methods for evaluating engraftment, and discusses considerations for choosing the appropriate model system to meet specific goals.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cell Transplantation , Immunity, Innate , Immunocompetence , Interleukin Receptor Common gamma Subunit/metabolism , Models, Animal , Peripheral Blood Stem Cell Transplantation , Animals , Animals, Newborn , Female , Flow Cytometry , Hematopoiesis/genetics , Humans , Immunity, Innate/genetics , Immunocompetence/genetics , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Spleen/immunology , Transplantation, Heterologous
19.
Ann N Y Acad Sci ; 1150: 46-53, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19120266

ABSTRACT

Our understanding of the basic biology of diabetes has been guided by observations made using animal models, particularly rodents. However, humans are not mice, and outcomes predicted by murine studies are not always representative of actual outcomes in the clinic. In particular, investigators studying diabetes have relied heavily on mouse and rat models of autoimmune type 1-like diabetes, and experimental results using these models have not been representative of many of the clinical trials in type 1 diabetes. In this article, we describe the availability of new models of humanized mice for the study of three areas of diabetes. These include the use of humanized mice for the study of (1) human islet stem and progenitor cells, (2) human islet allograft rejection, and (3) human immunity and autoimmunity. These humanized mouse models provide an important preclinical bridge between in vitro studies and rodent models and the translation of discoveries in these model systems to the clinic.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Disease Models, Animal , Insulin-Secreting Cells/physiology , Mice , Animals , Autoimmunity/physiology , Diabetes Mellitus, Type 1/immunology , Humans , Immune System/physiology , Insulin-Secreting Cells/transplantation , Mice, SCID , Stem Cells/physiology , Transplantation, Heterologous/methods
20.
Ann N Y Acad Sci ; 1150: 149-51, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19120285

ABSTRACT

We investigated the mechanisms by which Toll-like receptor (TLR) agonists affect the induction of mixed chimerism and skin allograft survival in mice treated with co-stimulation blockade (CB). We report that TLR agonists prevent the generation of mixed chimerism by breaking tolerance in the alloreactive CD4(+) and CD8(+) T cell compartments, and that type I interferon (IFN) is important in this process. Understanding how environmental perturbations affect CB-induced transplantation tolerance may lead to more effective regimens that can be used as an approach for the treatment of type I diabetes, for which the transplantation of pancreatic islets is a promising therapy.


Subject(s)
Chimerism/drug effects , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Toll-Like Receptors/agonists , Transplantation Tolerance/drug effects , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Poly I-C/pharmacology , Receptor, Interferon alpha-beta/genetics , Skin Transplantation/immunology , Skin Transplantation/veterinary , Toll-Like Receptor 3/agonists , Toll-Like Receptor 4/agonists
SELECTION OF CITATIONS
SEARCH DETAIL