Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 250
Filter
1.
Blood Adv ; 3(19): 2870-2882, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31585951

ABSTRACT

γδT cells are key players in cancer immune surveillance because of their ability to recognize malignant transformed cells, which makes them promising therapeutic tools in the treatment of cancer. However, the biological mechanisms of how γδT-cell receptors (TCRs) interact with their ligands are poorly understood. Within this context, we describe the novel allo-HLA-restricted and CD8α-dependent Vγ5Vδ1TCR. In contrast to the previous assumption of the general allo-HLA reactivity of a minor fraction of γδTCRs, we show that classic anti-HLA-directed, γδTCR-mediated reactivity can selectively act on hematological and solid tumor cells, while not harming healthy tissues in vitro and in vivo. We identified the molecular interface with proximity to the peptide-binding groove of HLA-A*24:02 as the essential determinant for recognition and describe the critical role of CD8 as a coreceptor. We conclude that alloreactive γδT-cell repertoires provide therapeutic opportunities, either within the context of haplotransplantation or as individual γδTCRs for genetic engineering of tumor-reactive T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/genetics , Animals , Humans , Mice
2.
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
3.
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
4.
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
5.
Leukemia ; 29(6): 1390-401, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25533804

ABSTRACT

Although anaplastic large-cell lymphomas (ALCL) carrying anaplastic lymphoma kinase (ALK) have a relatively good prognosis, aggressive forms exist. We have identified a novel translocation, causing the fusion of the TRAF1 and ALK genes, in one patient who presented with a leukemic ALK+ ALCL (ALCL-11). To uncover the mechanisms leading to high-grade ALCL, we developed a human patient-derived tumorgraft (hPDT) line. Molecular characterization of primary and PDT cells demonstrated the activation of ALK and nuclear factor kB (NFkB) pathways. Genomic studies of ALCL-11 showed the TP53 loss and the in vivo subclonal expansion of lymphoma cells, lacking PRDM1/Blimp1 and carrying c-MYC gene amplification. The treatment with proteasome inhibitors of TRAF1-ALK cells led to the downregulation of p50/p52 and lymphoma growth inhibition. Moreover, a NFkB gene set classifier stratified ALCL in distinct subsets with different clinical outcome. Although a selective ALK inhibitor (CEP28122) resulted in a significant clinical response of hPDT mice, nevertheless the disease could not be eradicated. These data indicate that the activation of NFkB signaling contributes to the neoplastic phenotype of TRAF1-ALK ALCL. ALCL hPDTs are invaluable tools to validate the role of druggable molecules, predict therapeutic responses and implement patient specific therapies.


Subject(s)
Drug Resistance, Neoplasm , Lymphoma, Large-Cell, Anaplastic/genetics , NF-kappa B/metabolism , Receptor Protein-Tyrosine Kinases/genetics , TNF Receptor-Associated Factor 1/genetics , Translocation, Genetic/genetics , Anaplastic Lymphoma Kinase , Animals , Blotting, Western , Flow Cytometry , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunoprecipitation , In Situ Hybridization, Fluorescence , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/mortality , Mice , Mice, Inbred NOD , NF-kappa B/genetics , Positive Regulatory Domain I-Binding Factor 1 , Proteasome Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , TNF Receptor-Associated Factor 1/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
6.
Clin Exp Immunol ; 174(3): 372-88, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23869841

ABSTRACT

Immunodeficient mice bearing targeted mutations in the IL2rg gene and engrafted with human immune systems are effective tools for the study of human haematopoiesis, immunity, infectious disease and transplantation biology. The most robust human immune model is generated by implantation of human fetal thymic and liver tissues in irradiated recipients followed by intravenous injection of autologous fetal liver haematopoietic stem cells [often referred to as the BLT (bone marrow, liver, thymus) model]. To evaluate the non-obese diabetic (NOD)-scid IL2rγ(null) (NSG)-BLT model, we have assessed various engraftment parameters and how these parameters influence the longevity of NSG-BLT mice. We observed that irradiation and subrenal capsule implantation of thymus/liver fragments was optimal for generating human immune systems. However, after 4 months, a high number of NSG-BLT mice develop a fatal graft-versus-host disease (GVHD)-like syndrome, which correlates with the activation of human T cells and increased levels of human immunoglobulin (Ig). Onset of GVHD was not delayed in NSG mice lacking murine major histocompatibility complex (MHC) classes I or II and was not associated with a loss of human regulatory T cells or absence of intrathymic cells of mouse origin (mouse CD45(+) ). Our findings demonstrate that NSG-BLT mice develop robust human immune systems, but that the experimental window for these mice may be limited by the development of GVHD-like pathological changes.


Subject(s)
Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Thymus Gland/transplantation , Animals , Disease Models, Animal , Hematopoietic Stem Cells , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Leukocyte Common Antigens/metabolism , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Inbred NOD , Molecular Sequence Data , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology
7.
Cell Death Dis ; 4: e567, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23559001

ABSTRACT

Reorganization of spared neural network connections is one of the most important processes for restoring impaired function after brain injury. However, plasticity is quite limited in the adult brain due to the presence of inhibitory molecules and a lack of intrinsic neuronal signals for axonal growth. Src homology 2-containing phosphatase (SHP)-1 has been shown to have a role in axon growth inhibition. Here, we tested the hypothesis that SHP-1 negatively affects axonal reorganization. We observed that unilateral motor cortex injury led to increased expression and activity of SHP-1 in the contralesional cortex. In this model, corticospinal axons originating from the contralesional cortex sprouted into the denervated side of the cervical spinal cord after injury. We observed that the number of sprouting fibers was increased in SHP-1-deficient heterozygous viable motheaten (+/me(v)) mice, which show reduced SHP-1 activity, and in wild-type mice treated with an SHP inhibitor. Motor function recovery of impaired forelimb was enhanced in +/me(v) mice. Collectively, our results indicate that downregulation of SHP-1 activity promotes corticospinal tract sprouting and functional recovery after brain injury.


Subject(s)
Brain Injuries/drug therapy , Enzyme Inhibitors/pharmacology , Motor Cortex/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors , Pyramidal Tracts/drug effects , Quinolines/pharmacology , Recovery of Function , Animals , Axons/drug effects , Axons/physiology , Brain Injuries/metabolism , Down-Regulation , Forelimb/drug effects , Forelimb/physiology , Heterozygote , Male , Mice , Motor Cortex/injuries , Motor Cortex/physiology , Nerve Fibers/drug effects , Nerve Fibers/physiology , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Pyramidal Tracts/enzymology , Pyramidal Tracts/injuries
8.
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
9.
Clin Exp Immunol ; 166(2): 269-80, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21985373

ABSTRACT

Graft-versus-host disease (GVHD) is a life-threatening complication of human allogeneic haematopoietic stem cell transplantation. Non-obese diabetic (NOD)-scid IL2rγ(null) (NSG) mice injected with human peripheral blood mononuclear cells (PBMC) engraft at high levels and develop a robust xenogeneic (xeno)-GVHD, which reproduces many aspects of the clinical disease. Here we show that enriched and purified human CD4 T cells engraft readily in NSG mice and mediate xeno-GVHD, although with slower kinetics compared to injection of whole PBMC. Moreover, purified human CD4 T cells engraft but do not induce a GVHD in NSG mice that lack murine MHC class II (NSG-H2-Ab1(tm1Gru), NSG-Ab°), demonstrating the importance of murine major histocompatibility complex (MHC) class II in the CD4-mediated xeno-response. Injection of purified human CD4 T cells from a DR4-negative donor into a newly developed NSG mouse strain that expresses human leucocyte antigen D-related 4 (HLA-DR4) but not murine class II (NSG-Ab° DR4) induces an allogeneic GVHD characterized by weight loss, fur loss, infiltration of human cells in skin, lung and liver and a high level of mortality. The ability of human CD4 T cells to mediate an allo-GVHD in NSG-Ab° DR4 mice suggests that this model will be useful to investigate acute allo-GVHD pathogenesis and to evaluate human specific therapies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Graft vs Host Disease , Receptors, Interleukin-2/immunology , Animals , CD4-Positive T-Lymphocytes/transplantation , Genes, MHC Class II , Graft vs Host Disease/immunology , HLA-DR4 Antigen/immunology , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Transplantation, Heterologous
10.
Vet Pathol ; 48(2): 495-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20817888

ABSTRACT

Spontaneous morbidity primarily affecting female breeders in 3 independent breeding colonies of NSG (NOD.Cg-Prkdc(scid) I12rg(tm1Wjl) /SzJ) mice prompted an investigation to uncover the cause of disease. Necropsies were performed on 264 (157 female and 107 male) spontaneously sick, experimentally unmanipulated NSG mice. In sum, 42 mice (15.9%) had acute or chronic renal inflammatory lesions, of which 12 had concurrent histologic evidence of an ascending urinary tract infection. From 94 kidneys cultured for bacterial organisms, 23 (24.5%) grew Enterococcus sp and 19 (20.2%) grew Klebsiella oxytoca. Female mice were twice more likely than males to present with nephritis. These findings indicate that bacterial nephritis is a major contributor to morbidity in the NSG strain.


Subject(s)
Animals, Laboratory , Enterococcus , Klebsiella oxytoca , Nephritis/veterinary , Opportunistic Infections/veterinary , Rodent Diseases/epidemiology , Rodent Diseases/microbiology , Animals , Female , Male , Mice , Mice, Inbred Strains , Morbidity , Nephritis/microbiology , Opportunistic Infections/epidemiology , Opportunistic Infections/microbiology
11.
Leukemia ; 24(11): 1859-66, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739953

ABSTRACT

In order to develop a xenograft model to determine the efficacy of new therapies against primary human precursor-B acute lymphoblastic leukemia (ALL) stem cells (LSCs), we used the highly immunodeficient non-obese diabetic (NOD).Cg-Prkdc(scid)IL2rg(tmlWjl)/SzJ (NOD-severe combined immune deficient (scid) IL2rg(-/-)) mouse strain. Intravenous transplantation of 2 of 2 ALL cell lines and 9 of 14 primary ALL cases generated leukemia-like proliferations in recipient mice by 1-7 months after transplant. Leukemias were retransplantable, and the immunophenotypes, gene rearrangements and expression profiles were identical or similar to those of the original primary samples. NOD-scid mice transplanted with the same primary samples developed similar leukemias with only a slightly longer latency than did NOD-scid-IL2Rg(-/-) mice. In this highly sensitive NOD-scid-IL2Rg(-/-)-based assay, 1-100 unsorted primary human ALL cells from five of five tested patients, four of whom eventually experienced leukemia relapse, generated leukemias in recipient mice. This very high frequency of LSCs suggests that a hierarchical LSC model is not valuable for poor-outcome ALL.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Stem Cells/pathology , Animals , Cell Line, Tumor/immunology , Cell Line, Tumor/pathology , Child , Humans , Leukemia, B-Cell/immunology , Leukemia, B-Cell/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Recurrence , Splenomegaly/pathology , Transplantation, Heterologous , Treatment Outcome
12.
Gene Ther ; 17(2): 238-49, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19829316

ABSTRACT

Human embryonic stem cells (hESCs) provide a novel source of hematopoietic and other cell populations suitable for gene therapy applications. Preclinical studies to evaluate engraftment of hESC-derived hematopoietic cells transplanted into immunodeficient mice demonstrate only limited repopulation. Expression of a drug-resistance gene, such as Tyr22-dihydrofolate reductase (Tyr22-DHFR), coupled to methotrexate (MTX) chemotherapy has the potential to selectively increase the engraftment of gene-modified, hESC-derived cells in mouse xenografts. Here, we describe the generation of Tyr22-DHFR-GFP-expressing hESCs that maintain pluripotency, produce teratomas and can differentiate into MTXr-hemato-endothelial cells. We demonstrate that MTX administered to nonobese diabetic/severe combined immunodeficient/IL-2Rgammac(null) (NSG) mice after injection of Tyr22-DHFR-hESC-derived cells significantly increases human CD34(+) and CD45(+) cell engraftment in the bone marrow (BM) and peripheral blood of transplanted MTX-treated mice. These results demonstrate that MTX treatment supports selective, long-term engraftment of Tyr22-DHFR cells in vivo, and provides a novel approach for combined human cell and gene therapy.


Subject(s)
Embryonic Stem Cells/metabolism , Methotrexate/pharmacology , Stem Cell Transplantation/methods , Tetrahydrofolate Dehydrogenase/genetics , Animals , Bone Marrow , Cell Differentiation , Cell Line , Drug Resistance , Genetic Therapy/methods , Graft Survival , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Teratoma/genetics , Tetrahydrofolate Dehydrogenase/metabolism
14.
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
15.
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
16.
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
17.
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
18.
Curr Top Microbiol Immunol ; 324: 87-94, 2008.
Article in English | MEDLINE | ID: mdl-18481454

ABSTRACT

Biomedical research including immunology and stem cell biology has developed greatly because of the evolving technology of gene modification and conventional transplantation methods using the most common experimental laboratory animal, the mouse. To translate promising research findings based on mouse research into clinical medicine, however, we need to clarify whether similar events take place in humans. In the study of hematology and immunology, humanized mice provide a unique and efficient experimental system to evaluate differentiation, function, and interaction of human blood cells or immune components. Here we review the latest experimental findings in the fields of immunology, stem cell biology, and regenerative medicine using humanized mice.


Subject(s)
Bone Marrow/physiology , Cell Differentiation , Hematopoietic Stem Cells/physiology , Models, Animal , Regeneration , Animals , Hematopoietic Stem Cell Transplantation , Humans , Mice , Mice, SCID
19.
Leukemia ; 22(6): 1207-13, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18418410

ABSTRACT

The presence of rare malignant stem cells supplying a hierarchy of malignant cells has recently been reported. In human acute myelogenous leukemia (AML), the leukemia stem cells (LSCs) have been phenotypically restricted within the CD34+CD38- fraction. To understand the origin of malignant cells in primary human B-precursor acute lymphocytic leukemia (B-ALL), we established a novel in vivo xenotransplantation model. Purified CD34+CD38+CD19+, CD34+CD38-CD19+ and CD34+CD38-CD19- bone marrow (BM) or peripheral blood (PB) cells from three pediatric B-ALL patients were intravenously injected into sublethally irradiated newborn NOD/SCID/IL2rgamma(null) mice. We found that both CD34+CD38+CD19+ and CD34+CD38-CD19+ cells initiate B-ALL in primary recipients, whereas the recipients of CD34+CD38-CD10-CD19- cells showed normal human hematopoietic repopulation. The extent of leukemic infiltration into the spleen, liver and kidney was similar between the recipients transplanted with CD34+CD38+CD19+ cells and those transplanted with CD34+CD38-CD19+ cells. In each of the three cases studied, transplantation of CD34+CD38+CD19+ cells resulted in the development of B-ALL in secondary recipients, demonstrating self-renewal capacity. The identification of CD34+CD38+CD19+ self-renewing B-ALL cells proposes a hierarchy of leukemia-initiating cells (LICs) distinct from that of AML. Recapitulation of patient B-ALL in NOD/SCID/IL2rgamma(null) recipients provides a powerful tool for directly studying leukemogenesis and for developing therapeutic strategies.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Antigens, CD19/metabolism , Antigens, CD34/metabolism , Hematopoietic Stem Cells/pathology , Neoplastic Stem Cells/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Animals, Newborn , Cell Differentiation , Cell Lineage , Child , Flow Cytometry , Graft Survival , Humans , Immunophenotyping , Infant , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/transplantation , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured , Whole-Body Irradiation
20.
Parasite Immunol ; 29(11): 567-74, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17944746

ABSTRACT

In this communication, we examine the determinants and duration of memory responses against filarial parasites using an intraperitoneal mouse model of Brugia pahangi infection. We assessed the role of T cells in the memory response against B. pahangi larvae by transferring splenic T cells from wild-type mice primed with L3 into T-cell-deficient mice. We found that mice reconstituted with primed T cells cleared intraperitoneal infections with infective larvae in an accelerated manner. To determine the components that may be responsible for the memory response, we transferred unfractionated T cells or purified CD4+ T cells or CD8+ T cells from BALB/cByJ mice primed a month earlier with L3 into T-cell-deficient BALB/c TCRbeta-/- mice. Recipients were challenged 10 days after adoptive transfer. Our data demonstrated that while either CD4+ or CD8+ T cells are able to confer some level of protection, both are required for an optimal recall response. To evaluate the longevity of the memory response, we primed several groups of wild-type mice at different times over a year. These mice were then challenged with a single injection of B. pahangi L3. The gap between the priming and second injections of larvae ranged between 4 and 60 weeks. We found that the memory responses in BALB/cByJ mice lasted over a year whereas those in C57BL/6 mice waned more rapidly.


Subject(s)
Brugia pahangi/immunology , Filariasis/immunology , Adoptive Transfer/methods , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Filariasis/parasitology , Filariasis/prevention & control , Immunologic Memory/immunology , Larva/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...