Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Immunol ; 208(7): 1652-1663, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35315788

ABSTRACT

Immunodeficient mice reconstituted with immune systems from patients, or personalized immune (PI) mice, are powerful tools for understanding human disease. Compared with immunodeficient mice transplanted with human fetal thymus tissue and fetal liver-derived CD34+ cells administered i.v. (Hu/Hu mice), PI mice, which are transplanted with human fetal thymus and adult bone marrow (aBM) CD34+ cells, demonstrate reduced levels of human reconstitution. We characterized APC and APC progenitor repopulation in human immune system mice and detected significant reductions in blood, bone marrow (BM), and splenic APC populations in PI compared with Hu/Hu mice. APC progenitors and hematopoietic stem cells (HSCs) were less abundant in aBM CD34+ cells compared with fetal liver-derived CD34+ cell preparations, and this reduction in APC progenitors was reflected in the BM of PI compared with Hu/Hu mice 14-20 wk posttransplant. The number of HSCs increased in PI mice compared with the originally infused BM cells and maintained functional repopulation potential, because BM from some PI mice 28 wk posttransplant generated human myeloid and lymphoid cells in secondary recipients. Moreover, long-term PI mouse BM contained functional T cell progenitors, evidenced by thymopoiesis in thymic organ cultures. Injection of aBM cells directly into the BM cavity, transgenic expression of hematopoietic cytokines, and coinfusion of human BM-derived mesenchymal stem cells synergized to enhance long-term B cell and monocyte levels in PI mice. These improvements allow a sustained time frame of 18-22 wk where APCs and T cells are present and greater flexibility for modeling immune disease pathogenesis and immunotherapies in PI mice.


Subject(s)
Bone Marrow , Hematopoietic Stem Cell Transplantation , Animals , Bone Marrow Cells , Hematopoietic Stem Cells , Humans , Liver , Mice
2.
J Allergy Clin Immunol ; 149(5): 1755-1771, 2022 05.
Article in English | MEDLINE | ID: mdl-34695489

ABSTRACT

BACKGROUND: Generation of thymic tissue from pluripotent stem cells would provide therapies for acquired and congenital thymic insufficiency states. OBJECTIVES: This study aimed to generate human thymic epithelial progenitors from human embryonic stem cells (hES-TEPs) and to assess their thymopoietic function in vivo. METHODS: This study differentiated hES-TEPs by mimicking developmental queues with FGF8, retinoic acid, SHH, Noggin, and BMP4. Their function was assessed in reaggregate cellular grafts under the kidney capsule and in hybrid thymi by incorporating them into swine thymus (SwTHY) grafts implanted under the kidney capsules of immunodeficient mice that received human hematopoietic stem and progenitor cells (hHSPCs) intravenously. RESULTS: Cultured hES-TEPs expressed FOXN1 and formed colonies expressing EPCAM and both cortical and medullary thymic epithelial cell markers. In thymectomized immunodeficient mice receiving hHSPCs, hES-TEPs mixed with human thymic mesenchymal cells supported human T-cell development. Hypothesizing that support from non-epithelial thymic cells might allow long-term function of hES-TEPs, the investigators injected them into SwTHY tissue, which supports human thymopoiesis in NOD severe combined immunodeficiency IL2Rγnull mice receiving hHSPCs. hES-TEPs integrated into SwTHY grafts, enhanced human thymopoiesis, and increased peripheral CD4+ naive T-cell reconstitution. CONCLUSIONS: This study has developed and demonstrated in vivo thymopoietic function of hES-TEPs generated with a novel differentiation protocol. The SwTHY hybrid thymus model demonstrates beneficial effects on human thymocyte development of hES-TEPs maturing in the context of a supportive thymic structure.


Subject(s)
Epithelial Cells , Thymocytes , Animals , Cell Differentiation , Epithelial Cells/physiology , Epithelium , Humans , Mice , Mice, Inbred NOD , Thymus Gland
3.
J Clin Invest ; 131(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33630757

ABSTRACT

In humans receiving intestinal transplantation (ITx), long-term multilineage blood chimerism often develops. Donor T cell macrochimerism (≥4%) frequently occurs without graft-versus-host disease (GVHD) and is associated with reduced rejection. Here we demonstrate that patients with macrochimerism had high graft-versus-host (GvH) to host-versus-graft (HvG) T cell clonal ratios in their allografts. These GvH clones entered the circulation, where their peak levels were associated with declines in HvG clones early after transplant, suggesting that GvH reactions may contribute to chimerism and control HvG responses without causing GVHD. Consistently, donor-derived T cells, including GvH clones, and CD34+ hematopoietic stem and progenitor cells (HSPCs) were simultaneously detected in the recipients' BM more than 100 days after transplant. Individual GvH clones appeared in ileal mucosa or PBMCs before detection in recipient BM, consistent with an intestinal mucosal origin, where donor GvH-reactive T cells expanded early upon entry of recipient APCs into the graft. These results, combined with cytotoxic single-cell transcriptional profiles of donor T cells in recipient BM, suggest that tissue-resident GvH-reactive donor T cells migrated into the recipient circulation and BM, where they destroyed recipient hematopoietic cells through cytolytic effector functions and promoted engraftment of graft-derived HSPCs that maintain chimerism. These mechanisms suggest an approach to achieving intestinal allograft tolerance.


Subject(s)
Graft vs Host Disease/immunology , Intestines/transplantation , Lymphopoiesis/immunology , Organ Transplantation , T-Lymphocytes/immunology , Transplantation Chimera/immunology , Allografts , Female , Graft vs Host Disease/pathology , Humans , Intestines/immunology , Intestines/pathology , Male , T-Lymphocytes/pathology
4.
Eur J Immunol ; 50(1): 138-141, 2020 01.
Article in English | MEDLINE | ID: mdl-31583677

ABSTRACT

We developed a rapid method to remove the native mouse thymus from NSG mice, which allowed us to compare the behavior of human immune cells in the presence or absence of human T cells in human immune system mice. Removing the native mouse thymus is critical for studies of human thymopiesis in grafted thymic tissue in humanized mice.


Subject(s)
Thymectomy/methods , Thymus Gland/immunology , Thymus Gland/transplantation , Transplantation, Heterologous/methods , Animals , Humans , Mice , Mice, Inbred NOD , Mice, SCID
5.
Cell Rep ; 29(12): 3916-3932.e5, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31851923

ABSTRACT

Non-recirculating tissue-resident memory T cells (TRMs) are the predominant T cell subset in diverse tissue sites, where they mediate protective immune responses in situ. Here, we reveal a role for TRM in maintaining immune homeostasis in the human pancreas through interactions with resident macrophages and the PD-1/PD-L1 inhibitory pathway. Using tissues obtained from organ donors, we identify that pancreas T cells comprise CD8+PD-1hi TRMs, which are phenotypically, functionally, and transcriptionally distinct compared to TRMs in neighboring jejunum and lymph node sites. Pancreas TRMs cluster with resident macrophages throughout the exocrine areas; TRM effector functions are enhanced by macrophage-derived co-stimulation and attenuated by the PD-1/PD-L1 pathways. Conversely, in samples from chronic pancreatitis, TRMs exhibit reduced PD-1 expression and reduced interactions with macrophages. These findings suggest important roles for PD-1 and TRM-macrophage interactions in controlling tissue homeostasis and immune dysfunctions underlying inflammatory disease, with important implications for PD-1-based immunotherapies.


Subject(s)
B7-H1 Antigen/metabolism , Immunologic Memory/physiology , Pancreas/metabolism , Pancreatitis/immunology , Pancreatitis/metabolism , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocyte Subsets/metabolism , B7-H1 Antigen/genetics , CD58 Antigens/metabolism , Humans , Immunity, Mucosal/genetics , Immunity, Mucosal/physiology , Immunologic Memory/genetics , Macrophages/immunology , Macrophages/metabolism , Pancreas/immunology , Pancreas/pathology , Pancreatitis/genetics , Programmed Cell Death 1 Receptor/genetics , Signal Transduction , T-Lymphocyte Subsets/immunology
6.
Cell Stem Cell ; 25(2): 225-240.e7, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31178255

ABSTRACT

The specific cellular physiology of hematopoietic stem cells (HSCs) is underexplored, and their maintenance in vitro remains challenging. We discovered that culture of HSCs in low calcium increased their maintenance as determined by phenotype, function, and single-cell expression signature. HSCs are endowed with low intracellular calcium conveyed by elevated activity of glycolysis-fueled plasma membrane calcium efflux pumps and a low-bone-marrow interstitial fluid calcium concentration. Low-calcium conditions inhibited calpain proteases, which target ten-eleven translocated (TET) enzymes, of which TET2 was required for the effect of low calcium conditions on HSC maintenance in vitro. These observations reveal a physiological feature of HSCs that can be harnessed to improve their maintenance in vitro.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/physiology , Proto-Oncogene Proteins/metabolism , Animals , Calpain/metabolism , Cell Self Renewal , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats , Dioxygenases , Glycolysis , Hematopoiesis , Homeostasis , Humans , Male , Mice , Mice, Inbred C57BL , Single-Cell Analysis , Transcriptome
7.
Blood Adv ; 1(23): 2007-2018, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-29296847

ABSTRACT

B cells play a major role in antigen presentation and antibody production in the development of autoimmune diseases, and some of these diseases disproportionally occur in females. Moreover, immune responses tend to be stronger in female vs male humans and mice. Because it is challenging to distinguish intrinsic from extrinsic influences on human immune responses, we used a personalized immune (PI) humanized mouse model, in which immune systems were generated de novo from adult human hematopoietic stem cells (HSCs) in immunodeficient mice. We assessed the effect of recipient sex and of donor autoimmune diseases (type 1 diabetes [T1D] and rheumatoid arthritis [RA]) on human B-cell development in PI mice. We observed that human B-cell levels were increased in female recipients regardless of the source of human HSCs or the strain of immunodeficient recipient mice. Moreover, mice injected with T1D- or RA-derived HSCs displayed B-cell abnormalities compared with healthy control HSC-derived mice, including altered B-cell levels, increased proportions of mature B cells and reduced CD19 expression. Our study revealed an HSC-extrinsic effect of recipient sex on human B-cell reconstitution. Moreover, the PI humanized mouse model revealed HSC-intrinsic defects in central B-cell tolerance that recapitulated those in patients with autoimmune diseases. These results demonstrate the utility of humanized mouse models as a tool to better understand human immune cell development and regulation.

8.
PLoS One ; 9(1): e86129, 2014.
Article in English | MEDLINE | ID: mdl-24465914

ABSTRACT

Thymic epithelial cells (TECs) are critical for the normal development and function of the thymus. Here, we examined the developmental stages of TECs using quantitative assessment of the cortical and medullary markers Keratin 5 and Keratin 8 (K5 and K8) respectively, in normal and gain/loss of function mutant animals. Gain of function mice overexpressed RANKL in T cells, whereas loss of function animals lacked expression of Traf6 in TECs (Traf6ΔTEC). Assessment of K5 and K8 expression in conjunction with other TEC markers in wild type mice identified novel cortical and medullary TEC populations, expressing different combinations of these markers. RANKL overexpression led to expansion of all medullary TECs (mTECs) and enlargement of the thymic medulla. This in turn associated with a block in thymocyte development and loss of CD4+ CD8+, CD4+ and CD8+ thymocytes. In contrast, Traf6 deletion inhibited the production of most TEC populations including cortical TECs (cTECs), defined by absence of UEA-1 binding and LY51 expression, but had no apparent effect on thymocyte development. These results reveal a large degree of heterogeneity within the TEC compartment and the existence of several populations exhibiting concomitant expression of cortical, medullary and epithelial markers and whose production is regulated by RANKL and Traf6.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/metabolism , RANK Ligand/metabolism , TNF Receptor-Associated Factor 6/metabolism , Thymus Gland/cytology , Aging/physiology , Animals , Animals, Newborn , Antigens, Neoplasm/metabolism , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Cell Proliferation , Epithelial Cell Adhesion Molecule , Gene Deletion , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Time Factors , Transcription Factors/metabolism , AIRE Protein
9.
J Clin Invest ; 123(8): 3510-24, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23867620

ABSTRACT

TRAF6, an E3 ubiquitin protein ligase, plays a critical role in T cell tolerance by regulating medullary thymic epithelial cell (mTEC) development. mTECs regulate T cell tolerance by ectopically expressing self-antigens and eliminating autoreactive T cells in the thymus. Here we show that mice with mTEC depletion due to conditional deletion of Traf6 expression in murine thymic epithelial cells (Traf6ΔTEC mice) showed a surprisingly narrow spectrum of autoimmunity affecting the liver. The liver inflammation in Traf6ΔTEC mice exhibited all the histological and immunological characteristics of human autoimmune hepatitis (AIH). The role of T cells in AIH establishment was supported by intrahepatic T cell population changes and AIH development after transfer of liver T cells into immunodeficient mice. Despite a 50% reduction in natural Treg thymic output, peripheral tolerance in Traf6ΔTEC mice was normal, whereas compensatory T regulatory mechanisms were evident in the liver of these animals. These data indicate that mTECs exert a cell-autonomous role in central T cell tolerance and organ-specific autoimmunity, but play a redundant role in peripheral tolerance. These findings also demonstrate that Traf6ΔTEC mice are a relevant model with which to study the pathophysiology of AIH, as well as autoantigen-specific T cell responses and regulatory mechanisms underlying this disease.


Subject(s)
Epithelial Cells/pathology , Hepatitis, Autoimmune/pathology , Thymus Gland/pathology , Adoptive Transfer , Animals , Antibodies, Antinuclear/metabolism , Disease Models, Animal , Epithelial Cells/immunology , Hepatitis, Autoimmune/immunology , Humans , Immune Tolerance , Liver/immunology , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasma Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , T-Lymphocytes, Regulatory/immunology , TNF Receptor-Associated Factor 6/deficiency , TNF Receptor-Associated Factor 6/genetics , Thymus Gland/immunology
10.
Immunol Res ; 54(1-3): 177-90, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22454100

ABSTRACT

During thymocyte development bone marrow-derived precursors in the thymus undergo a series of differentiation steps to produce self-tolerant, mature T lymphocytes. The thymus contains two functionally distinct anatomical compartments, consisting of a centrally located medulla surrounded by the thymic cortex. These compartments in turn are comprised of two major cellular components: (1) the T lymphoid compartment of developing thymocytes and (2) the thymic stroma consisting mainly of thymic epithelial cells (TECs). These epithelial cells are further separated into cortical and medullary TECs (cTECs and mTECs) based on their localization within the thymic cortex or medulla respectively. Reciprocal interactions between thymocytes and epithelial cells are required for the development of both cellular components into a functional thymic organ. Thymocytes provide trophic factors for the development of a complex three-dimensional epithelial cell network, while epithelial cells regulate T cell development through expression and presentation of self-antigens on major histocompatibility molecules. Our work focuses on how thymic epithelial cells regulate T cell development and function and on elucidating the mechanisms of thymic epithelial cell differentiation. Here we review current knowledge and provide our own insight into the development, differentiation and antigen presenting properties of TECs. We focus specifically on how mTECs regulate T cell repertoire selection and central tolerance.


Subject(s)
Antigen-Presenting Cells/immunology , Epithelial Cells/immunology , T-Lymphocytes/immunology , Thymus Gland/cytology , Animals , Antigen Presentation/immunology , Autoantigens/immunology , Epithelial Cells/cytology , Humans , Immune Tolerance
11.
J Exp Med ; 207(5): 999-1013, 2010 May 10.
Article in English | MEDLINE | ID: mdl-20404100

ABSTRACT

Medullary thymic epithelial cells (mTECs) play an important role in T cell tolerance and prevention of autoimmunity. Mice deficient in expression of the signaling protein Sin exhibit exaggerated immune responses and multitissue inflammation. Here, we show that Sin is expressed in the thymic stroma, specifically in mTECs. Sin deficiency led to thymic stroma-dependent autoimmune manifestations shown by radiation chimeras and thymic transplants in nude mice, and associated with defective mTEC-mediated elimination of thymocytes in a T cell receptor transgenic model of negative selection. Lack of Sin expression correlated with a disorganized medullary architecture and fewer functionally mature mTECs under steady-state conditions. Additionally, Sin deficiency inhibited the expansion of mTECs in response to in vivo administration of keratinocyte growth factor (KGF). These results identify Sin as a novel regulator of mTEC development and T cell tolerance, and suggest that Sin is important for homeostatic maintenance of the medullary epithelium in the adult thymus.


Subject(s)
Phosphoproteins/physiology , Thymus Gland/cytology , Thymus Gland/immunology , Adaptor Proteins, Signal Transducing , Animals , Autoimmunity/immunology , Cell Differentiation/immunology , Cell Differentiation/physiology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Fibroblast Growth Factor 7/pharmacology , Homeostasis , Immune Tolerance , Immunologic Memory , Immunosuppression Therapy , Inflammation/etiology , Major Histocompatibility Complex , Mice , Mice, Knockout , Mice, Nude , Phosphoproteins/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/immunology , Thymus Gland/drug effects
12.
Immunity ; 25(6): 907-18, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17174122

ABSTRACT

Leukocyte migration and trafficking is dynamically regulated by various chemokine and adhesion molecules and is vital to the proper function of the immune system. We describe a role for the Cas and Hef-1-associated signal transducer in hematopoietic cells (Chat-H) as a critical regulator of T lymphocyte migration, by using lentivirus-mediated RNA interference (RNAi). Impaired migration of Chat-H-depleted cells coincided with defective inside-out signaling shown by diminished chemokine-induced activation of the Rap-1 GTPase and integrin-mediated adhesion. Localization of Chat-H to the plasma membrane, association with its binding partner Crk-associated substrate in lymphocytes (CasL), and Chat-H-mediated CasL serine-threonine phosphorylation were required for T cell migration. These results identify Chat-H as a critical signaling intermediate acting upstream of Rap1 to regulate chemokine-induced adhesion and migration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion/immunology , Chemotaxis, Leukocyte/immunology , Models, Immunological , T-Lymphocytes/metabolism , Adaptor Proteins, Signal Transducing/immunology , Animals , Enzyme Activation/physiology , Immunoblotting , Integrins/immunology , Integrins/metabolism , Mice , Mice, Inbred C57BL , Protein Isoforms/immunology , Protein Isoforms/metabolism , RNA, Small Interfering , Signal Transduction/immunology , T-Lymphocytes/immunology , rap1 GTP-Binding Proteins/immunology , rap1 GTP-Binding Proteins/metabolism
13.
Mol Cell Biol ; 25(24): 11035-46, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16314525

ABSTRACT

Our studies have concentrated on elucidating the role of the signaling protein Sin in T-lymphocyte function. We have previously shown that Sin overexpression inhibits T-lymphocyte development and activation. Here we show that Sin-deficient mice exhibit exaggerated immune responses characterized by enhanced cytokine secretion and T-cell-dependent antibody production. Excessive T-cell responses in young mice correlate with spontaneous development of inflammatory lesions in different organs of aged Sin(-/-) mice, particularly the small intestine. The intestinal inflammation is characterized by T- and B-cell infiltrates in the lamina propria, which correlate with crypt enlargement and marked villus expansion and/or damage. Similar to the human intestinal inflammatory disorder Crohn's disease (CD), and in contrast to most mouse models of mucosal inflammation, inflammatory lesions in the gastrointestinal tract of Sin(-/-) mice are restricted to the small bowel. Taken together, these results suggest that Sin regulates immune system and T-lymphocyte function and that immune system dysfunction in the absence of Sin may underlie the pathogenesis of tissue-specific inflammation and enteropathies such as CD.


Subject(s)
Enterocolitis/immunology , Lymphocyte Activation , Phosphoproteins/physiology , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Enterocolitis/genetics , Granuloma/genetics , Intestinal Mucosa/pathology , Liver/pathology , Male , Mice , Mice, Knockout , Phosphoproteins/deficiency , Phosphoproteins/genetics , Receptors, Interleukin-2/analysis , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...