Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 19(3): 279-290, 2018 03.
Article in English | MEDLINE | ID: mdl-29434353

ABSTRACT

Deletion of master regulators of the B cell lineage reprograms B cells into T cells. Here we found that the transcription factor Hoxb5, which is expressed in uncommitted hematopoietic progenitor cells but is not present in cells committed to the B cell or T cell lineage, was able to reprogram pro-pre-B cells into functional early T cell lineage progenitors. This reprogramming started in the bone marrow and was completed in the thymus and gave rise to T lymphocytes with transcriptomes, hierarchical differentiation, tissue distribution and immunological functions that closely resembled those of their natural counterparts. Hoxb5 repressed B cell 'master genes', activated regulators of T cells and regulated crucial chromatin modifiers in pro-pre-B cells and ultimately drove the B cell fate-to-T cell fate conversion. Our results provide a de novo paradigm for the generation of functional T cells through reprogramming in vivo.


Subject(s)
B-Lymphocytes/cytology , Cell Lineage/immunology , Cellular Reprogramming/immunology , Homeodomain Proteins/immunology , T-Lymphocytes/cytology , Animals , Cell Differentiation , Cell Lineage/genetics , Cellular Reprogramming/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Precursor Cells, B-Lymphoid/cytology
2.
Nat Immunol ; 19(9): 1036, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29915300

ABSTRACT

In the version of this article initially published, some identification of the supplementary information was incorrect. The items originally called Supplementary Tables 1, 2, 3, 4 and 5 should be Source Data Figures 1, 2, 4, 5 and 7, respectively; those originally called Supplementary Tables 6, 7 and 8 should be Supplementary Tables 1, 2 and 3, respectively; and those originally called Source Data Figures 1, 2, 4, 5 and 7 should be Supplementary Tables 4, 5, 6, 7 and 8, respectively. The errors have been corrected in the HTML version of the article.

3.
J Immunol ; 211(5): 885-894, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37486211

ABSTRACT

IFN-γ is a pleiotropic cytokine that plays a controversial role in regulatory T cell (Treg) activity. In this study, we sought to understand how IFN-γ receptor (IFN-γR) signaling affects donor Tregs following allogeneic hematopoietic cell transplant (allo-HCT), a potentially curative therapy for leukemia. We show that IFN-γR signaling inhibits Treg expansion and conversion of conventional T cells (Tcons) to peripheral Tregs in both mice and humans. Mice receiving IFN-γR-deficient allo-HCT showed markedly reduced graft-versus-host disease (GVHD) and graft-versus-leukemia (GVL) effects, a trend associated with increased frequencies of Tregs, compared with recipients of wild-type allo-HCT. In mice receiving Treg-depleted allo-HCT, IFN-γR deficiency-induced peripheral Treg conversion was effective in preventing persistent GVHD while minimally affecting GVL effects. Thus, impairing IFN-γR signaling in Tcons may offer a promising strategy for achieving GVL effects without refractory GVHD. Similarly, in a human PBMC-induced xenogeneic GVHD model, significant inhibition of GVHD and an increase in donor Tregs were observed in mice cotransferred with human CD4 T cells that were deleted of IFN-γR1 by CRISPR/Cas9 technology, providing proof-of-concept support for using IFN-γR-deficient T cells in clinical allo-HCT.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia , Mice , Humans , Animals , T-Lymphocytes, Regulatory , Transplantation, Homologous , Leukocytes, Mononuclear , Mice, Knockout
4.
Mol Pharm ; 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38953708

ABSTRACT

The coronavirus (COVID-19) pandemic has underscored the critical role of mRNA-based vaccines as powerful, adaptable, readily manufacturable, and safe methodologies for prophylaxis. mRNA-based treatments are emerging as a hopeful avenue for a plethora of conditions, encompassing infectious diseases, cancer, autoimmune diseases, genetic diseases, and rare disorders. Nonetheless, the in vivo delivery of mRNA faces challenges due to its instability, suboptimal delivery, and potential for triggering undesired immune reactions. In this context, the development of effective drug delivery systems, particularly nanoparticles (NPs), is paramount. Tailored with biophysical and chemical properties and susceptible to surface customization, these NPs have demonstrated enhanced mRNA delivery in vivo and led to the approval of several NPs-based formulations for clinical use. Despite these advancements, the necessity for developing a refined, targeted NP delivery system remains imperative. This review comprehensively surveys the biological, translational, and clinical progress in NPs-mediated mRNA therapeutics for both the prevention and treatment of diverse diseases. By addressing critical factors for enhancing existing methodologies, it aims to inform the future development of precise and efficacious mRNA-based therapeutic interventions.

5.
J Immunol ; 208(2): 420-428, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34903640

ABSTRACT

There are urgent needs for humanized mouse models of viral respiratory diseases to study immunopathogenesis and therapeutic interventions. Although human immune system (HIS) mice permit analysis in real time of human immune responses in vivo, evolutionary divergences preclude their usefulness for the respiratory viruses that do not infect mouse lungs. In this study, we sought to use HIS mice with human lung (HL) tissue xenografts (HISL mice) to address this issue. The grafted HL tissue maintained histologically normal structure, and populated with human tissue-resident immune cells, including CD11c+ dendritic cells and CD4+ and CD8+ tissue-resident memory T cells. HISL mice showed a marked expansion of tissue-resident memory T cells and generation of viral Ag-specific T cells in the HL xenografts, and production of antiviral IgM and IgG Abs upon immunization of the HL xenograft by H1N1 influenza viruses. RNA-seq analysis on H1N1-infected and control HL xenografts identified a total of 5089 differentially expressed genes with enrichments for genes involved in respiratory diseases, viral infections, and associated immune responses. Furthermore, prophylactic viral exposures resulted in protection against subsequent lethal challenge by intranasal viral inoculation. This study supports the usefulness of this preclinical model in exploring the immunopathology and therapies of respiratory viral diseases.


Subject(s)
Antibodies, Viral/blood , Dendritic Cells/immunology , Influenza A Virus, H1N1 Subtype/immunology , Memory T Cells/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Heterografts , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Immunologic Memory/immunology , Lung/cytology , Lung/immunology , Lung Transplantation/methods , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Orthomyxoviridae Infections/immunology , Vaccination
6.
Eur J Immunol ; 52(10): 1640-1647, 2022 10.
Article in English | MEDLINE | ID: mdl-35976660

ABSTRACT

There is an urgent need for animal models of coronavirus disease 2019 to study immunopathogenesis and test therapeutic intervenes. In this study, we showed that NOD/SCID IL2rg-/- (NSG) mice engrafted with human lung (HL) tissue (NSG-L mice) could be infected efficiently by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and that live virus capable of infecting Vero cells was found in the HL grafts and multiple organs from infected NSG-L mice. RNA-Sequencing identified a series of differentially expressed genes, which are enriched in viral defense responses, chemotaxis, IFN stimulation and pulmonary fibrosis, between HL grafts from infected and control NSG-L mice. Furthermore, when infected with SARS-CoV-2, humanized mice with both human immune system (HIS) and autologous HL grafts (HISL mice) had bodyweight loss and hemorrhage and immune cell infiltration in HL grafts, which were not observed in immunodeficient NSG-L mice, indicating the development of anti-viral immune responses in these mice. In support of this possibility, the infected HISL mice showed bodyweight recovery and lack of detectable live virus at the later time. These results demonstrate that NSG-L and HISL mice are susceptible to SARS-CoV-2 infection, offering a useful in vivo model for studying SARS-CoV-2 infection and the associated immune response and immunopathology, and testing anti-SARS-CoV-2 therapies.


Subject(s)
COVID-19 , Animals , Chlorocebus aethiops , Disease Models, Animal , Humans , Immunity , Lung , Mice , Mice, Inbred NOD , Mice, SCID , RNA , SARS-CoV-2 , Vero Cells
7.
Xenotransplantation ; 30(5): e12824, 2023.
Article in English | MEDLINE | ID: mdl-37695083

ABSTRACT

BACKGROUND: Blood vessels that contain endothelial cells (ECs) on the surface are in direct contact with host blood and are the first target of xenograft rejection. Currently, our understanding of human anti-pig vessel immune responses is primarily based on in vitro assays using pig ECs. Therefore, it is necessary to develop an animal model that permits in vivo study of human immunological rejection of pig vessels. METHODS: Pig artery tissues (PAT) were transplanted into human immune system (HIS) mice or immunodeficient NSG mice (as controls). Intragraft human immune cell infiltration and antibody deposition were quantified using histology and immunohistochemistry. Donor antigen-specific immune responses were quantified using a mixed lymphocyte reaction and a complement-dependent killing assay. RESULTS: Pig CD31+ ECs were detected and increased 2-fold from weeks 3 to 5 in PAT xenografts from immunodeficient NSG mice. However, compared with NSG mice, PAT xenografts in HIS mice had significantly lower numbers of porcine CD31+ ECs and showed a marked reduction from week 3 to week 5. PAT xenograft rejection in HIS mice is associated with intensive infiltration of human immune cells, deposition of human IgM and IgG antibodies, and the formation of a tertiary lymphoid structure. Robust donor pig antigen-specific human T cells and antibody responses were detected in PAT-transplanted HIS mice. CONCLUSION: We have developed a humanized mouse model to evaluate human anti-pig xenoimmune responses by PAT transplantation in vivo. This model is expected to facilitate the refinement of pig gene-editing strategies (the expression on EC surface) and the testing of local immunosuppressive strategies for clinical pig organ xenotransplantation.


Subject(s)
Endothelial Cells , Graft Rejection , Humans , Animals , Swine , Mice , Transplantation, Heterologous , Arteries/transplantation , Immunosuppressive Agents
8.
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
9.
Am J Transplant ; 22(9): 2246-2253, 2022 09.
Article in English | MEDLINE | ID: mdl-35373451

ABSTRACT

Thrombospondin-1 (TSP-1) is a key mediator of renal ischemia-reperfusion injury (IRI), a major cause of kidney dysfunction under various disease conditions and a risk factor of renal allograft rejection. In this study, we developed a nanotechnology-based therapy targeting TSP-1 to prevent renal IRI. A biocompatible nanoparticle (NP) capable of specific binding to TSP-1 was prepared by conjugating NPs with TSP-1-binding (LSKL) peptides. LSKL/NPs not only effectively adsorbed recombinant TSP-1 proteins in vitro, but also efficiently neutralized TSP-1 in mice undergoing renal IRI. IRI-induced elevation of TSP-1 in the kidney was significantly inhibited by post-IR treatment with LSKL/NPs, but not free LSKL or NPs. Furthermore, TSP-1 proteins adsorbed on LSKL/NPs were functionally inactive and unable to induce apoptosis in renal tubular epithelial cells. Importantly, LSKL/NPs induced strong protection against renal IRI, as shown by markedly diminished serum creatinine levels and improved histological lesions of the kidney. Thus, LSKL/NPs provide a useful means of depleting and inactivating TSP-1 and a potential therapy for renal IRI.


Subject(s)
Kidney Transplantation , Nanoparticles , Reperfusion Injury , Animals , Apoptosis , Kidney/pathology , Kidney Transplantation/adverse effects , Mice , Mice, Inbred C57BL , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Thrombospondin 1/antagonists & inhibitors , Thrombospondin 1/metabolism , Thrombospondin 1/pharmacology
10.
Immunol Cell Biol ; 100(8): 624-635, 2022 09.
Article in English | MEDLINE | ID: mdl-35662247

ABSTRACT

Humanized mice reconstituted with a functional human immune system (HIS) are instrumental in studying human immunity and immune disorders in vivo. The poor or lack of cross-reactivity between mouse cytokines and human cells limits the development and/or function of human immune cell subsets including human myeloid, natural killer and B cells. Here we explored the potential to achieve long-term production of human cytokines in immunodeficient mice using a transposon-plasmid-based hydrodynamic injection approach. We constructed a transposon-plasmid carrying five human cytokine coding sequences (named PB-5F), and observed that four of the cytokines (granulocyte-macrophage colony-stimulating factor, interleukin (IL)-15, IL-6 and IL-3) were detectable in sera and three (granulocyte-macrophage colony-stimulating factor, IL-15 and IL-6) showed long-term production in immunodeficient mice that received a single hydrodynamic injection of PB-5F plus the transposase plasmid (Super PB). Furthermore, a single injection of PB-5F/Super PB markedly enhanced the reconstitution of human myeloid cells and natural killer cells, and promoted human B-cell maturation in HIS mice. Taken together, our data revealed that hydrodynamic injection of the PB-5F/Super PB vectors may serve as a convenient and efficacious means to promote human immune function in HIS mice.


Subject(s)
Cytokines , Granulocyte-Macrophage Colony-Stimulating Factor , Animals , Cytokines/genetics , Humans , Hydrodynamics , Interleukin-6/genetics , Killer Cells, Natural , Mice , Plasmids/genetics
11.
J Autoimmun ; 119: 102612, 2021 05.
Article in English | MEDLINE | ID: mdl-33611150

ABSTRACT

We evaluated the role of the thymus in development of multi-organ autoimmunity in human immune system (HIS) mice. T cells were essential for disease development and the same T cell clones with varying phenotypes infiltrated multiple tissues. De novo-generated hematopoietic stem cell (HSC)-derived T cells were the major disease drivers, though thymocytes pre-existing in grafted human thymi contributed if not first depleted. HIS mice with a native mouse thymus developed disease earlier than thymectomized mice with a thymocyte-depleted human thymus graft. Defective structure in the native mouse thymus was associated with impaired negative selection of thymocytes expressing a transgenic TCR recognizing a self-antigen. Disease developed without direct recognition of antigens on recipient mouse MHC. While human thymus grafts had normal structure and negative selection, failure to tolerize human T cells recognizing mouse antigens presented on HLA molecules may explain eventual disease development. These new insights have implications for human autoimmunity and suggest methods of avoiding autoimmunity in next-generation HIS mice.


Subject(s)
Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmunity , Disease Susceptibility/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Animals , Antigens , Autoimmune Diseases/pathology , Biomarkers , Clonal Selection, Antigen-Mediated/immunology , Disease Models, Animal , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Immunophenotyping , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Mice , Mice, Knockout , Mice, Transgenic , Organ Specificity/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
Xenotransplantation ; 28(3): e12657, 2021 05.
Article in English | MEDLINE | ID: mdl-33111471

ABSTRACT

Liver xenotransplantation (LXT) is greatly impeded by severe thrombocytopenia, anemia, and coagulopathy. Hepatic phagocytic cells are thought to play an important role in LXT-induced thrombocytopenia and anemia. In this study, we investigated whether the lack of recipient CD47-donor SIRPα interaction, which is known to induce xenograft rejection by macrophages, exacerbates platelet and RBC depletion following LXT. We first addressed this question in the absence of anti-donor immune responses using a syngeneic mouse liver transplantation (LT) model. Neither wild-type (WT) nor CD47KO B6 mice developed thrombocytopenia following LT from WT B6 donors. Although a moderate decline in RBCs was detected following LT, there was no significant difference in RBC counts between WT and CD47KO recipients. Because mouse CD47 is cross-reactive with rat SIRPα, we then compared thrombocytopenia and anemia between WT and CD47KO mice following rat LXT. Unlike syngeneic mouse LT, significant thrombocytopenia and anemia were detected following rat LXT. However, the severities of both platelet and RBC depletions were comparable between WT and CD47KO recipients. Furthermore, WT and CD47KO recipients showed a similar extent of early platelet activation. Our results indicate that CD47-SIRPα signaling does not significantly affect the loss of platelets or RBCs following LXT, suggesting that the limited cross-reactivity between recipient CD47 and donor SIRPα is not a significant risk factor for LXT-induced thrombocytopenia and anemia.


Subject(s)
Anemia , Thrombocytopenia , Anemia/etiology , Animals , CD47 Antigen , Heterografts , Liver , Mice , Mice, Knockout , Phagocytosis , Rats , Receptors, Immunologic , Risk Factors , Thrombocytopenia/etiology , Transplantation, Heterologous
13.
BMC Gastroenterol ; 21(1): 84, 2021 Feb 23.
Article in English | MEDLINE | ID: mdl-33622256

ABSTRACT

BACKGROUND: Hepatic cavernous hemangioma is the most common type of benign liver tumor. Although ruptures and hemorrhages of hepatic hemangioma are rare complications, they are associated with high mortality. Most practitioners only pay more attention to abdominal hemorrhages caused by the rupture of hepatic hemangiomas. However, spontaneous intracapsular hemorrhages can often be neglected and poorly understood. CASE PRESENTATION: A 65-year-old man was referred to our institution with right upper quadrant pain, which had occurred suddenly and without a history of recent trauma. The blood test results were normal. Magnetic resonance imaging (MRI) of the abdomen showed a cystic mass in the right liver lobe. Considering the possibility of hepatic cystadenoma with hemorrhage, the patient underwent a right hepatic lobectomy. The pathological findings unexpectedly revealed intratumoral hemorrhage of hepatic hemangioma. The patient recovered well and was discharged eight days after surgery. CONCLUSIONS: Intracapsular hemorrhage of hepatic cavernous hemangioma is challenging to diagnose and has a high potential risk of rupture. MRI is beneficial for diagnosing subacute internal hemorrhage cases, and it is recommended to undergo surgery for patients with a definitive diagnosis.


Subject(s)
Hemangioma, Cavernous , Hemangioma , Liver Neoplasms , Aged , Hemangioma, Cavernous/complications , Hemangioma, Cavernous/diagnostic imaging , Hemangioma, Cavernous/surgery , Hemorrhage/etiology , Humans , Liver Neoplasms/complications , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/surgery
14.
Am J Transplant ; 20(6): 1538-1550, 2020 06.
Article in English | MEDLINE | ID: mdl-31883299

ABSTRACT

Xenogeneic porcine islet transplantation is a promising potential therapy for type 1 diabetes (T1D). Understanding human immune responses against porcine islets is crucial for the design of optimal immunomodulatory regimens for effective control of xenogeneic rejection of porcine islets in humans. Humanized mice are a valuable tool for studying human immune responses and therefore present an attractive alternative to human subject research. Here, by using a pig-to-humanized mouse model of xenogeneic islet transplantation, we described the human immune response to transplanted porcine islets, a process characterized by dense islet xenograft infiltration of human CD45+ cells comprising activated human B cells, CD4+ CD44+ IL-17+ Th17 cells, and CD68+ macrophages. In addition, we tested an experimental immunomodulatory regimen in promoting long-term islet xenograft survival, a triple therapy consisting of donor splenocytes treated with ethylcarbodiimide (ECDI-SP), and peri-transplant rituximab and rapamycin. We observed that the triple therapy effectively inhibited graft infiltration of T and B cells as well as macrophages, promoted transitional B cells both in the periphery and in the islet xenografts, and provided a superior islet xenograft protection. Our study therefore indicates an advantage of donor ECDI-SP treatment in controlling human immune cells in promoting long-term islet xenograft survival.


Subject(s)
Islets of Langerhans Transplantation , Th17 Cells , Animals , B-Lymphocytes , Graft Rejection/prevention & control , Graft Survival , Mice , Swine , Transplantation, Heterologous
15.
Circ Res ; 123(11): e35-e47, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30571460

ABSTRACT

RATIONALE: The mechanisms driving atherothrombotic risk in individuals with JAK2 V617F ( Jak2 VF) positive clonal hematopoiesis or myeloproliferative neoplasms are poorly understood. OBJECTIVE: The goal of this study was to assess atherosclerosis and underlying mechanisms in hypercholesterolemic mice with hematopoietic Jak2 VF expression. METHODS AND RESULTS: Irradiated low-density lipoprotein receptor knockout ( Ldlr-/-) mice were transplanted with bone marrow from wild-type or Jak2 VF mice and fed a high-fat high-cholesterol Western diet. Hematopoietic functions and atherosclerosis were characterized. After 7 weeks of Western diet, Jak2 VF mice showed increased atherosclerosis. Early atherosclerotic lesions showed increased neutrophil adhesion and content, correlating with lesion size. After 12 weeks of Western diet, Jak2 VF lesions showed increased complexity, with larger necrotic cores, defective efferocytosis, prominent iron deposition, and costaining of erythrocytes and macrophages, suggesting erythrophagocytosis. Jak2 VF erythrocytes were more susceptible to phagocytosis by wild-type macrophages and showed decreased surface expression of CD47, a "don't-eat-me" signal. Human JAK2VF erythrocytes were also more susceptible to erythrophagocytosis. Jak2 VF macrophages displayed increased expression and production of proinflammatory cytokines and chemokines, prominent inflammasome activation, increased p38 MAPK (mitogen-activated protein kinase) signaling, and reduced levels of MerTK (c-Mer tyrosine kinase), a key molecule mediating efferocytosis. Increased erythrophagocytosis also suppressed efferocytosis. CONCLUSIONS: Hematopoietic Jak2 VF expression promotes early lesion formation and increased complexity in advanced atherosclerosis. In addition to increasing hematopoiesis and neutrophil infiltration in early lesions, Jak2 VF caused cellular defects in erythrocytes and macrophages, leading to increased erythrophagocytosis but defective efferocytosis. These changes promote accumulation of iron in plaques and increased necrotic core formation which, together with exacerbated proinflammatory responses, likely contribute to plaque instability.


Subject(s)
Atherosclerosis/genetics , Erythrocytes/metabolism , Janus Kinase 2/genetics , Macrophages/metabolism , Phagocytosis , Adult , Aged , Animals , Atherosclerosis/blood , Atherosclerosis/metabolism , CD47 Antigen/genetics , CD47 Antigen/metabolism , Cytokines/genetics , Cytokines/metabolism , Female , Hematopoiesis , Humans , Iron/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neutrophils/metabolism , c-Mer Tyrosine Kinase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Proc Natl Acad Sci U S A ; 114(41): 10954-10959, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28874533

ABSTRACT

There is an urgent and unmet need for humanized in vivo models of type 1 diabetes to study immunopathogenesis and immunotherapy, and in particular antigen-specific therapy. Transfer of patient blood lymphocytes to immunodeficient mice is associated with xenogeneic graft-versus-host reactivity that complicates assessment of autoimmunity. Improved models could identify which human T cells initiate and participate in beta-cell destruction and help define critical target islet autoantigens. We used humanized mice (hu-mice) containing robust human immune repertoires lacking xenogeneic graft-versus-host reactivity to address this question. Hu-mice constructed by transplantation of HLA-DQ8+ human fetal thymus and CD34+ cells into HLA-DQ8-transgenic immunodeficient mice developed hyperglycemia and diabetes after transfer of autologous HLA-DQ8/insulin-B:9-23 (InsB:9-23)-specific T-cell receptor (TCR)-expressing human CD4+ T cells and immunization with InsB:9-23. Survival of the infused human T cells depended on the preexisting autologous human immune system, and pancreatic infiltration by human CD3+ T cells and insulitis were observed in the diabetic hu-mice, provided their islets were stressed by streptozotocin. This study fits Koch's postulate for pathogenicity, demonstrating a pathogenic role of islet autoreactive CD4+ T-cell responses in type 1 diabetes induction in humans, underscores the role of the target beta-cells in their immunological fate, and demonstrates the capacity to initiate disease with T cells, recognizing the InsB:9-23 epitope in the presence of islet inflammation. This preclinical model has the potential to be used in studies of the pathogenesis of type 1 diabetes and for testing of clinically relevant therapeutic interventions.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , HLA-DQ Antigens/immunology , Insulin-Secreting Cells/immunology , Insulin/immunology , Peptide Fragments/immunology , Animals , Autoimmunity , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic
17.
Xenotransplantation ; 26(2): e12459, 2019 03.
Article in English | MEDLINE | ID: mdl-30136356

ABSTRACT

CD47 is a ubiquitously expressed transmembrane glycoprotein that plays a complex role in regulation of cell survival and function. We have previously shown that the interspecies incompatibility of CD47 plays an important role in triggering rejection of cellular xenografts by macrophages. However, the role of CD47 in solid organ transplantation remains undetermined. Here, we explored this question in mouse models of heart allotransplantation. We observed that the lack of CD47 in donor hearts had no deleterious effect on graft survival in syngeneic or single MHC class I-mismatched recipients, in which both wild-type (WT) and CD47 knockout (CD47 KO) mouse hearts survived long term with no sign of rejection. Paradoxically, elimination of donor CD47 was beneficial for graft survival in signal MHC class II- and class I- plus class II-mismatched combinations, in which CD47 KO donor hearts showed significantly improved survival compared to WT donor hearts. Similarly, CD47 KO donor hearts were more resistant than WT hearts to humoral rejection in α1,3-galactosyltransferase-deficient mice. Moreover, a significant prolongation of WT allografts was observed in recipient mice treated with antibodies against a CD47 ligand thrombospondin-1 (TSP1) or with TSP1 deficiency, indicating that TSP1-CD47 signaling may stimulate vascularized allograft rejection. Thus, unlike cellular transplantation, donor CD47 expression may accelerate the rejection of vascularized allografts.


Subject(s)
Allografts/immunology , CD47 Antigen/immunology , Graft Rejection/immunology , Graft Survival/immunology , Thrombospondin 1/immunology , Animals , CD47 Antigen/genetics , Cell Transplantation/methods , Graft Rejection/genetics , Graft Survival/genetics , Heart Transplantation/methods , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Thrombospondin 1/genetics , Transplantation, Heterologous/methods , Transplantation, Homologous/methods
18.
Am J Hematol ; 93(6): 786-793, 2018 06.
Article in English | MEDLINE | ID: mdl-29603337

ABSTRACT

Persistent low levels of disease in bone marrow, an immunoprivileged tissue, are responsible for relapse following allogeneic hematopoietic cell transplantation. Using mouse models carrying primary human acute lymphoblast leukemia derived from MLL-AF9-overexpressing human hematopoietic stem cells, we demonstrate that allogeneic lymphocyte infusion (ALI)-mediated graft-vs.-leukemia effects selectively spare leukemia cells in the bone marrow. The resistance of leukemia cells to ALI within bone marrow is due to the immunosuppressive status of the tissue, as ALI achieved a significantly increased complete remission rate when leukemia cells were dislodged from bone marrow by treatment with a CXCR4 antagonist AMD3100. Adoptive transfer experiments confirmed that the frequency of leukemia-initiating cells in bone marrow was significantly decreased in the recipients treated with ALI plus AMD3100 compared to those receiving ALI only. These findings indicate that the immunoprivileged nature of bone marrow is largely responsible for relapse after immunotherapies, and that treatment with AMD3100 may offer a clinically-practical approach to improving the outcome of adoptive allogeneic cell therapy.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, CXCR4/antagonists & inhibitors , Animals , Benzylamines , Cell Line, Tumor , Cyclams , Heterocyclic Compounds/therapeutic use , Heterografts , Humans , Immunotherapy, Adoptive/methods , Lymphocyte Transfusion/methods , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Transplantation, Homologous
19.
Nat Rev Immunol ; 7(7): 519-31, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17571072

ABSTRACT

Xenotransplantation using pigs as the transplant source has the potential to resolve the severe shortage of human organ donors. Although the development of relatively non-toxic immunosuppressive or tolerance-inducing regimens will be required to justify clinical trials using pig organs, recent advances in our understanding of the biology of xenograft rejection and zoonotic infections, and the generation of alpha1,3-galactosyltransferase-deficient pigs have moved this approach closer to clinical application. This Review highlights the major obstacles impeding the translation of xenotransplantation into clinical therapies and the potential solutions, providing a perspective on the future of clinical xenotransplantation.


Subject(s)
Transplantation, Heterologous/immunology , Transplantation, Heterologous/trends , Animals , Antibody Formation/immunology , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Immune Tolerance/immunology , T-Lymphocytes/immunology
20.
Immunol Rev ; 258(1): 30-44, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24517424

ABSTRACT

Allogeneic hematopoietic cell transplantation (allo-HCT) is increasingly being performed to treat patients with hematologic malignancies. However, separating the beneficial graft-versus-tumor (GVT) or graft-versus-leukemia effects from graft-versus-host disease (GVHD) has been difficult and remains a significant challenge toward improving therapeutic efficacy and reducing toxicity of allo-HCT. GVHD is induced by donor T cells that also mediate potent anti-tumor responses. However, despite the largely shared effector mechanisms, extensive animal studies have demonstrated the potential of dissociating the GVT effect from GVHD. Also in many clinical cases, long-term remission was achieved following allo-HCT, without significant GVHD. A better mechanistic understanding of the immunopathophysiology of GVHD and GVT effects may potentially help to improve allo-HCT as well as maximize the benefit of GVT effects while minimizing GVHD. In this article, we review the role of IFN-γ in regulation of alloresponses following allo-HCT, with a focus on the mechanisms of how this cytokine may separate GVHD from GVT effects.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Tumor Effect/immunology , Hematologic Neoplasms/surgery , Hematopoietic Stem Cell Transplantation , Interferon-gamma/immunology , T-Lymphocytes/immunology , Animals , Graft vs Host Disease/metabolism , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Interferon-gamma/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL