Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Commun Biol ; 4(1): 344, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33727658

ABSTRACT

Adiponectin is a well-known insulin sensitizer and anti-inflammatory molecule, possessing therapeutic potentials in cardiovascular, metabolic and cancer diseases. Results of the present study demonstrate that adiponectin is expressed in a population of regulatory T-cells (Treg) resided within the thymic nurse cell (TNC) complexes. Adoptive transfer of adiponectin-expressing Treg precursors effectively attenuated obesity, improved glucose and insulin tolerance, prevented fatty liver injuries in wild-type mice fed a high-fat diet, and significantly inhibited breast cancer development in MMTV-PyVT transgenic mice. Within the TNC complexes, locally produced adiponectin bound to and regulated the expression as well as the distribution of CD100, a transmembrane lymphocyte semaphorin, in turn modulating the lymphoepithelial interactions to facilitate T-cell development and maturation. In summary, adiponectin plays an important role in the selection and development of T lymphocytes within the TNC complexes. Adiponectin-expressing Treg represent a promising candidate for adoptive cell immunotherapy against obesity-related metabolic and cancer diseases.


Subject(s)
Adiponectin/metabolism , Cell Differentiation , T-Lymphocytes, Regulatory/metabolism , Thymocytes/metabolism , Thymus Gland/metabolism , Adiponectin/genetics , Adoptive Transfer , Animals , Antigens, CD/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Cell Line, Tumor , Disease Models, Animal , Female , Glucose Intolerance/immunology , Glucose Intolerance/metabolism , Glucose Intolerance/prevention & control , Humans , Insulin Resistance , Mammary Tumor Virus, Mouse/genetics , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/immunology , Obesity/metabolism , Obesity/prevention & control , Phenotype , Semaphorins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Thymocytes/immunology , Thymocytes/transplantation , Thymus Gland/immunology
2.
Blood ; 135(25): 2252-2265, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32181817

ABSTRACT

Prolonged or enhanced expression of the proto-oncogene Lmo2 is associated with a severe form of T-cell acute lymphoblastic leukemia (T-ALL), designated early T-cell precursor ALL, which is characterized by the aberrant self-renewal and subsequent oncogenic transformation of immature thymocytes. It has been suggested that Lmo2 exerts these effects by functioning as component of a multi-subunit transcription complex that includes the ubiquitous adapter Ldb1 along with b-HLH and/or GATA family transcription factors; however, direct experimental evidence for this mechanism is lacking. In this study, we investigated the importance of Ldb1 for Lmo2-induced T-ALL by conditional deletion of Ldb1 in thymocytes in an Lmo2 transgenic mouse model of T-ALL. Our results identify a critical requirement for Ldb1 in Lmo2-induced thymocyte self-renewal and thymocyte radiation resistance and for the transition of preleukemic thymocytes to overt T-ALL. Moreover, Ldb1 was also required for acquisition of the aberrant preleukemic ETP gene expression signature in immature Lmo2 transgenic thymocytes. Co-binding of Ldb1 and Lmo2 was detected at the promoters of key upregulated T-ALL driver genes (Hhex, Lyl1, and Nfe2) in preleukemic Lmo2 transgenic thymocytes, and binding of both Ldb1 and Lmo2 at these sites was reduced following Cre-mediated deletion of Ldb1. Together, these results identify a key role for Ldb1, a nonproto-oncogene, in T-ALL and support a model in which Lmo2-induced T-ALL results from failure to downregulate Ldb1/Lmo2-nucleated transcription complexes which normally function to enforce self-renewal in bone marrow hematopoietic progenitors.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Self Renewal , DNA-Binding Proteins/physiology , LIM Domain Proteins/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Thymocytes/cytology , Adaptor Proteins, Signal Transducing/genetics , Adoptive Transfer , Animals , Antigens, CD/biosynthesis , Cell Transformation, Neoplastic , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Deletion , Gene Knock-In Techniques , LIM Domain Proteins/deficiency , LIM Domain Proteins/genetics , Lymphopoiesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Mas , RNA-Seq , Radiation Chimera , Radiation Tolerance , Thymocytes/metabolism , Thymocytes/radiation effects , Thymocytes/transplantation
3.
Clin Exp Immunol ; 192(3): 302-314, 2018 06.
Article in English | MEDLINE | ID: mdl-29431868

ABSTRACT

The tyrosine kinase zeta chain-associated protein of 70 kDa (ZAP-70) plays a key role in T cell development and signalling. In the absence of ZAP-70, T cell development is arrested in the CD4+ CD8+ double-positive stage, thus ZAP-70 homozygous knockout (ZAP-70-/- ) mice have no mature T cells in their peripheral lymphoid organs and blood, causing severe immunodeficiency. We investigated the early kinetics and long-term effects of wild-type thymocyte transfer on T cell repopulation in ZAP-70-/- mice. We used a single intraperitoneal (i.p.) injection to deliver donor thymocytes to the recipients. Here, we show that after i.p. injection donor thymocytes leave the peritoneum through milky spots in the omentum and home to the thymus, where donor-originated CD4- CD8- double-negative thymocytes most probably restore T cell development and the disrupted thymic architecture. Subsequently, newly developed, donor-originated, single-positive αß T cells appear in peripheral lymphoid organs, where they form organized T cell zones. The established chimerism was found to be stable, as donor-originated cells were present in transferred ZAP-70-/- mice as late as 8 months after i.p. injection. We demonstrate that a simple i.p. injection of ZAP-70+/+ thymocytes is a feasible method for the long-term reconstitution of T cell development in ZAP-70-deficient mice.


Subject(s)
Adoptive Transfer/methods , Immunologic Deficiency Syndromes/therapy , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/cytology , Thymocytes/transplantation , ZAP-70 Protein-Tyrosine Kinase/deficiency , Animals , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Lymphocyte Count , Mice , Mice, Knockout , Severe Combined Immunodeficiency/genetics , T-Lymphocytes/immunology , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/immunology
4.
Adv Clin Exp Med ; 25(2): 369-75, 2016.
Article in English | MEDLINE | ID: mdl-27627572

ABSTRACT

Medical science seems to be on the threshold of a revolution: It seems possible that in twenty years, doctors will be able to replace organs in the human body like parts in a car. This is thanks to the recent achievement of a team from the Medical Research Council Center for Regenerative Medicine in Edinburgh, Scotland - the group of researchers tried to regenerate the thymus gland in mice. The thymus gland is an essential organ for the development of the immune system, but very few people have any idea that it exists. In the literature and also in people's awareness, the fact is often that the thymus controls and harmonizes the entire immune system and the immune functioning of the organism. It is the primary donor of cells for the lymphatic system, much as bone marrow is the cell donor for the cardiovascular system. It is within the thymus that progenitor cells are created and then undergo maturation and differentiation into mature T cells. The thymus gland is located in the mediastinum, behind the sternum. It is composed of two identical lobes. Each lobe is divided into a central medulla and a peripheral cortex. The thymus is at its largest and most active during the neonatal and pre-adolescent periods. After this period the organ gradually disappears and is replaced by fat. In elderly individuals the thymus weighs 5 g. The aim of this work is to shed new light on this important immune defense organ, whose function is not confined to the destruction of nonfunctional T cells.


Subject(s)
Regeneration , Thymocytes/immunology , Thymus Gland/immunology , Age Factors , Aging/immunology , Animals , Clonal Selection, Antigen-Mediated , Humans , T-Lymphocytes/immunology , Thymocytes/transplantation , Thymus Gland/cytology , Thymus Gland/transplantation , Thymus Hormones/metabolism
5.
Arthritis Res Ther ; 18(1): 184, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27516061

ABSTRACT

BACKGROUND: Apoptotic cell-based therapies have been proposed to treat chronic inflammatory diseases. The aim of this study was to investigate the effect of intravenous (i.v.) apoptotic cell infusion in ongoing collagen-induced arthritis (CIA) and the interaction of this therapy with other treatments used in rheumatoid arthritis (RA), including methotrexate (MTX) or anti-TNF therapy. METHODS: The effects of i.v. apoptotic cell infusion were evaluated in a CIA mouse model in DBA/1 mice immunized with bovine type II collagen. The number and functions of antigen-presenting cells (APC), regulatory CD4(+) T cells (Treg), and circulating anti-collagen auto-antibodies were analyzed in CIA mice. RESULTS: Treatment of arthritic mice with i.v. apoptotic cell infusion significantly reduced the arthritis clinical score. This therapeutic approach modified T cell responses against the collagen auto-antigen with selective induction of collagen-specific Treg. In addition, we observed that APC from apoptotic-cell-treated animals were resistant to toll-like receptor ligand activation and favored ex vivo Treg induction, indicating APC reprogramming. Apoptotic cell injection-induced arthritis modulation was dependent on transforming growth factor (TGF)-ß, as neutralizing anti-TGF-ß antibody prevented the effects of apoptotic cells. Methotrexate did not interfere, while anti-TNF therapy was synergic with apoptotic-cell-based therapy. CONCLUSION: Overall, our data demonstrate that apoptotic-cell-based therapy is efficient in treating ongoing CIA, compatible with current RA treatments, and needs to be evaluated in humans in the treatment of RA.


Subject(s)
Apoptosis/immunology , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Cell- and Tissue-Based Therapy/methods , Thymocytes/transplantation , Animals , Antirheumatic Agents/pharmacology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Female , Methotrexate/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Tumor Necrosis Factor-alpha/antagonists & inhibitors
6.
Immunol Rev ; 271(1): 173-84, 2016 May.
Article in English | MEDLINE | ID: mdl-27088914

ABSTRACT

Thymus transplants were never used to correct T-cell intrinsic deficiencies, as it is generally believed that thymocytes have short intrinsic lifespans. This notion is based on multiple thymus transplantation experiments, where it was shown that thymus-resident cells were rapidly replaced by progenitors migrating from the bone marrow (BM). This substitution occurs even when bone marrow precursors are unable to generate T cells, as in Rag1/2(-) or severe combined immunodeficiency (SCID)-deficient mice. In contrast, two groups reported that neonatal thymi transplanted into mice that cannot respond to IL-7 harbor populations with extensive capacity to self-renew, which maintain continuous thymocyte generation for several months after surgery. The consequences of this self-renewal capacity differed in these two laboratories. We found that these thymus transplants rapidly reconstitute the full diversity of peripheral T-cell repertoires 1 month after surgery, the earliest time point studied. Moreover, transplantation experiments performed across major histocompatibility barriers show that allogeneic-transplanted thymi are not rejected, and allogeneic cells do not induce graft-versus-host disease, both syngeneic and allogeneic transplants inducing rapid protection from infection. These results indicate a potential use of neonatal thymus transplants to correct T-cell intrinsic deficiencies. The other group observed that continuous thymocyte renewal from BM precursors was fundamental to prevent tumor development. In the absence of this input, thymocytes from the transplanted thymus generated tumors with all the characteristics of T-cell acute lymphoblastic leukemia (T-ALL). Moreover, they suggested that the absence of BM competition was responsible for the T-ALLs developing in X-linked severe combined immunodeficiency (SCID)-X1 patients, deficient in the expression of IL2-Rγc . These patients were treated with autologous CD34(+) cells transfected with virus vectors expressing γc in the absence of myeloablation. We here review the potential therapeutic impact of thymus transplantation and compare the results of these two laboratories aiming to find an answer to the 'Dr Jekill versus Mr. Hyde' status of thymus transplantation experiments.


Subject(s)
Severe Combined Immunodeficiency/therapy , T-Lymphocytes/physiology , Thymocytes/physiology , Thymus Gland/immunology , Adenosine/analogs & derivatives , Adenosine/immunology , Animals , Cell Self Renewal , Clonal Selection, Antigen-Mediated , Humans , Immune Tolerance , Interleukin-7/immunology , Mice , Mice, SCID , Thymocytes/transplantation , Thymus Gland/transplantation , Transplantation, Homologous
7.
Mol Ther ; 24(6): 1090-1099, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26957223

ABSTRACT

Insertional oncogenesis due to retroviral (RV) vector integration has caused recurrent leukemia in multiple gene therapy trials, predominantly due to vector integration effects at the LMO2 locus. While currently available preclinical safety models have been used for evaluating vector safety, none have predicted or reproduced the recurrent LMO2 integrations seen in previous X-linked severe combined immunodeficiency (X-SCID) and Wiskott-Aldrich clinical gene therapy trials. We now describe a new assay for assessing vector safety that recapitulates naturally occurring insertions into Lmo2 and other T-cell proto-oncogenes leading to a preleukemic developmental arrest in primary murine thymocytes cultured in vitro. This assay was used to compare the relative oncogenic potential of a variety of gamma-RV and lentiviral vectors and to assess the risk conferred by various transcriptional elements contained in these genomes. Gamma-RV vectors that contained full viral long-terminal repeats were most prone to causing double negative 2 (DN2) arrest and led to repeated cases of Lmo2 pathway activation, while lentiviral vectors containing these same elements were significantly less prone to activate proto-oncogenes or cause DN2 arrest. This work provides a new preclinical assay that is especially relevant for assessing safety in SCID disorders and provides a new tool for designing safer RV vectors.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gammaretrovirus/genetics , Genetic Vectors/adverse effects , LIM Domain Proteins/genetics , Lentivirus/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Thymocytes/cytology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Disease Models, Animal , Humans , MEF2 Transcription Factors/genetics , Mice , Mutagenesis, Insertional , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Thymocytes/drug effects , Thymocytes/transplantation , Up-Regulation
9.
Cardiovasc Res ; 106(3): 443-52, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25681396

ABSTRACT

AIMS: To investigate whether activation of atheroprotective peritoneal B1a cells by apoptotic cells or phosphatidylserine liposomes (PSLs) can enhance their protective actions during atherosclerosis development. METHODS AND RESULTS: Male apolipoprotein E-knockout (ApoE-/-) mice were treated with apoptotic cells or PSLs at the beginning of 8-week high-fat diet. Intraperitoneally administered apoptotic cells attenuated atherosclerosis in hypercholesterolemic ApoE-/- mice by 53% and macrophage accumulation by 52%, effects mimicked by administering PSLs and abolished by B1a cell depletion by splenectomy. These effects were associated with reduced lesion CD4+ and CD8+ T cells, mRNAs of MCP-1, VCAM-1, TNF-α, IL-1ß, IL-12, and IL-18 while anti-inflammatory TGF-ß mRNA levels doubled. Apoptotic cells or PSLs increased B1a lymphocytes including TIM-1+ B1a cells in vivo and in vitro while other lymphocyte populations were unaffected. Total plasma IgM, anti-leucocyte, anti-CD3, anti-CD4, and anti-oxLDL IgM were elevated. IgM in atherosclerotic lesions was also elevated and this was associated with reduced lesion MDA-LDL (oxLDL), apoptotic cells and necrotic core size. These effects of activating B1a cells could be attributed to B1a-derived polyreactive IgM deposited in lesions that reduce inflammatory cytokines by lowering lesion ox-LDL via anti-oxLDL IgM, T-cells via anti-leucocyte, anti-CD3, and anti-CD4 IgM, apoptotic cells and necrotic core size via IgM binding to apoptotic cells and enhancing phagocytosis, which also elevates anti-inflammatory cytokines. CONCLUSION: Targeting B1a cell activation by PSLs may be a potentially potent therapeutic strategy to attenuate atherosclerosis and reduce the incidence of atherosclerosis-dependent myocardial infarction and stroke.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Apoptosis , Atherosclerosis/prevention & control , B-Lymphocyte Subsets/drug effects , Immunoglobulin M/biosynthesis , Lymphocyte Activation/drug effects , Phosphatidylserines/administration & dosage , Thymocytes/transplantation , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Immunoglobulin M/immunology , Inflammation Mediators/metabolism , Lipoproteins, LDL/metabolism , Liposomes , Male , Mice, Knockout , Necrosis , Phagocytosis , Phenotype , Splenectomy , Thymocytes/immunology , Thymocytes/metabolism , Thymocytes/pathology , Time Factors
10.
Leukemia ; 29(4): 927-38, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25283843

ABSTRACT

Lmo2 is an oncogenic transcription factor that is frequently overexpressed in T-cell acute leukemias, in particular poor prognosis early T-cell precursor-like (ETP-) acute lymphoblastic leukemia (ALL). The primary effect of Lmo2 is to cause self-renewal of developing CD4(-)CD8(-) (double negative, DN) T cells in the thymus, leading to serially transplantable thymocytes that eventually give rise to leukemia. These self-renewing thymocytes are intrinsically radioresistant implying that they may be a source of leukemia relapse after therapy. The homeobox transcription factor, Hhex, is highly upregulated in Lmo2-transgenic thymocytes and can phenocopy Lmo2 in inducing thymocyte self-renewal, implying that Hhex may be a key component of the Lmo2-induced self-renewal program. To test this, we conditionally deleted Hhex in the thymi of Lmo2-transgenic mice. Surprisingly, this did not prevent accumulation of DN thymocytes, nor alter the rate of overt leukemia development. However, deletion of Hhex abolished the transplantation capacity of Lmo2-transgenic thymocytes and overcame their radioresistance. We found that Hhex regulates Kit expression in Lmo2-transgenic thymocytes and that abrogation of Kit signaling phenocopied loss of Hhex in abolishing the transplantation capacity and radioresistance of these cells. Thus, targeting the Kit signaling pathway may facilitate the eradication of leukemia-initiating cells in immature T-cell leukemias in which it is expressed.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Expression Regulation, Leukemic , Homeodomain Proteins/genetics , LIM Domain Proteins/genetics , Proto-Oncogene Proteins c-kit/genetics , Thymocytes/metabolism , Thymus Gland/metabolism , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , CD4 Antigens/genetics , CD4 Antigens/metabolism , CD8 Antigens/genetics , CD8 Antigens/metabolism , Disease Models, Animal , Gamma Rays , Gene Deletion , LIM Domain Proteins/metabolism , Mice , Mice, Transgenic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-kit/metabolism , Radiation Tolerance , Signal Transduction , Thymocytes/pathology , Thymocytes/radiation effects , Thymocytes/transplantation , Thymus Gland/pathology , Thymus Gland/radiation effects , Transcription Factors/deficiency
11.
Proc Natl Acad Sci U S A ; 110(16): 6494-9, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23576744

ABSTRACT

Most Forkhead box P3(+) (Foxp3(+)) CD4 regulatory T cell (Treg) precursors are newly formed thymocytes that acquire Foxp3 expression on antigen encounter in the thymus. Differentiation of Treg, however, can also occur in the periphery. What limits this second layer of self- and nonself-reactive Treg production in physiological conditions remains to be understood. In this work, we tested the hypothesis that, similarly to thymic Treg, the precursors of peripheral Treg are immature T cells. We show that CD4(+)CD8(-)Foxp3(-) thymocytes and recent thymic emigrants (RTEs), contrarily to peripheral naïve mature cells, efficiently differentiate into Treg on transfer into lymphopenic mice. By varying donor and recipient mice and conducting ex vivo assays, we document that the preferential conversion of newly formed T cells does not require intrathymic preactivation, is cell-intrinsic, and correlates with low and high sensitivity to natural inhibitors and inducers of Foxp3 expression, such as IL-6, T-cell receptor triggering, and TGF-ß. Finally, ex vivo analysis of human thymocytes and peripheral blood T cells revealed that human RTE and newly developed T cells share an increased potential to acquire a FOXP3(bright)CD25(high) Treg phenotype. Our findings indicating that RTEs are the precursors of Tregs differentiated in the periphery should guide the design of Treg-based therapies.


Subject(s)
Cell Differentiation/immunology , Cell Movement/immunology , Precursor Cells, T-Lymphoid/cytology , T-Lymphocytes, Regulatory/cytology , Thymocytes/cytology , Animals , Cells, Cultured , Flow Cytometry , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Humans , Interleukin-6/immunology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/immunology , Statistics, Nonparametric , Thymocytes/immunology , Thymocytes/transplantation , Transforming Growth Factor beta/immunology
12.
Circ Res ; 113(1): 52-61, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23584255

ABSTRACT

RATIONALE: Apoptotic cell phagocytosis (efferocytosis) is mediated by specific receptors and is essential for resolution of inflammation. In chronic inflammation, apoptotic cell clearance is dysfunctional and soluble levels of several apoptotic cell receptors are elevated. Reports have identified proteolytic cleavage as a mechanism capable of releasing soluble apoptotic cell receptors, but the functional implications of their proteolysis are unclear. OBJECTIVE: To test the hypothesis that ADAM17-mediated cleavage of apoptotic cell receptors limits efferocytosis in vivo. METHODS AND RESULTS: In vivo comparison of macrophage efferocytosis in wild-type and Adam17-null hematopoietic chimeras demonstrates that ADAM17 deficiency leads to a 60% increase in efferocytosis and an enhanced anti-inflammatory phenotype in a model of peritonitis. In vitro uptake of phosphatidylserine liposomes identifies the dual-pass apoptotic cell receptor CD36 as a major contributor to enhanced efferocytosis, and CD36 surface levels are elevated on macrophages from Adam17-null mice. Further, temporal elevation of CD36 expression with inflammation may also contribute to its impact. Soluble CD36 from macrophage-conditioned media comprises 2 species based on Western blotting, and mass spectrometry identifies 3 N-terminal peptides that represent probable cleavage sites. Levels of soluble CD36 are decreased in Adam17-null conditioned media, providing evidence for involvement of ADAM17 in CD36 cleavage. Importantly, enhanced efferocytosis in vivo by macrophages lacking ADAM17 is CD36 dependent and accelerates macrophage clearance from the peritoneum, thus promoting resolution of inflammation and highlighting the impact of increased apoptotic cell uptake. CONCLUSIONS: Our studies demonstrate the importance of ADAM17-mediated proteolysis for in vivo efferocytosis regulation and suggest a possible mechanistic link between chronic inflammation and defective efferocytosis.


Subject(s)
ADAM Proteins/physiology , Apoptosis/physiology , CD36 Antigens/physiology , Macrophages, Peritoneal/enzymology , Peritonitis/enzymology , Phagocytosis/physiology , ADAM Proteins/deficiency , ADAM Proteins/genetics , ADAM17 Protein , Amino Acid Sequence , Animals , Bone Marrow Transplantation , CD36 Antigens/chemistry , Chimera , Culture Media, Conditioned/pharmacology , Dexamethasone/pharmacology , Embryonic Stem Cells/transplantation , Liposomes , Macrophages, Peritoneal/physiology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peritonitis/chemically induced , Peritonitis/pathology , Phenotype , Thioglycolates/toxicity , Thymocytes/pathology , Thymocytes/transplantation
13.
Eur J Immunol ; 43(5): 1185-94, 2013 May.
Article in English | MEDLINE | ID: mdl-23436244

ABSTRACT

Mammalian ortholog of Drosophila cell polarity protein, Dlg1, plays a critical role in neural synapse formation, epithelial cell homeostasis, and urogenital development. More recently, it has been proposed that Dlg1 may also be involved in the regulation of T-cell proliferation, migration, and Ag-receptor signaling. However, a requirement for Dlg1 in development and function of T lineage cells remains to be established. In this study, we investigated a role for Dlg1 during T-cell development and function using a combination of conditional Dlg1 KO and two different Cre expression systems where Dlg1 deficiency is restricted to the T-cell lineage only, or all hematopoietic cells. Here, using three different TCR models, we show that Dlg1 is not required during development and selection of thymocytes bearing functionally rearranged TCR transgenes. Moreover, Dlg1 is dispensable in the activation and proliferative expansion of Ag-specific TCR-transgenic CD4(+) and CD8(+) T cells in vitro and in vivo. Surprisingly, however, we show that Dlg1 is required for normal generation of memory T cells during endogenous response to cognate Ag. Thus, Dlg1 is not required for the thymocyte selection or the activation of primary T cells, however it is involved in the generation of memory T cells.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Lineage/immunology , Immunologic Memory , Nerve Tissue Proteins/immunology , Receptors, Antigen, T-Cell/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Polarity , Cell Proliferation , Discs Large Homolog 1 Protein , Gene Expression , Integrases , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptors, Antigen, T-Cell/genetics , SAP90-PSD95 Associated Proteins , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/transplantation , Thymus Gland/cytology , Thymus Gland/immunology
14.
Transplantation ; 94(11): 1095-102, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23222735

ABSTRACT

BACKGROUND: In many humanized mouse models, there are few T cells in the engrafted human cell, whereas the number of B cells is high. We attempted to overcome this limitation and investigate whether the entire process of human T cell development arose similarly to the process in humans, as previously reported. METHODS: To produce an advanced humanized mice model, we transplanted human fetal liver/thymus tissue subrenally and injected human CD34(+) stem cells intravenously into NOD/SCID/IL2Rgamma null (NSG) mice. RESULTS: Humanized mice transplanted with fetal thymus/liver tissues and fetal liver-derived CD34(+) stem cells (FLT+FLCD34) showed higher levels of human cells and T cells than mice transplanted with fetal liver-derived CD34(+) stem cells only (FLCD34). In the transplanted thymus tissue of FLT+FLCD34 mice, thymus seeding progenitors (TSPs), early thymic progenitors (ETPs), pre-T cells, and all the other human T cell populations were identified. In the periphery, FLT+FLCD34 mice have high levels of CD45RA(+) T cells; conversely, FLCD34 mice have higher levels of CD45RO(+) T cells. The CD45RO(+) T cells of FLCD34 mice proliferated rapidly after stimulation and exhibited innate T cells properties, expressing PLZF (promyelocytic leukemia zinc finger protein). CONCLUSION: Human T cells educated by mouse MHC II in mice without a human thymus differ from normal human T cells. On the basis of these findings, numerous T cell-tropic human diseases could be explored in our humanized mice and molecular aspects of human T cell development could be also studied extensively.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Liver Transplantation , Liver , T-Lymphocytes/transplantation , Thymus Gland/transplantation , Animals , Antigens, CD34/metabolism , Biomarkers/metabolism , Cell Proliferation , Flow Cytometry , Hematopoietic Stem Cells/immunology , Humans , Immunity, Innate , Immunologic Memory , Immunophenotyping/methods , Interferon-gamma/metabolism , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Kruppel-Like Transcription Factors/metabolism , Leukocyte Common Antigens/metabolism , Liver/embryology , Liver/immunology , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/transplantation , Promyelocytic Leukemia Zinc Finger Protein , T-Lymphocytes/immunology , Thymocytes/immunology , Thymocytes/transplantation , Thymus Gland/embryology , Thymus Gland/immunology , Time Factors
15.
Transplant Proc ; 43(6): 2325-33, 2011.
Article in English | MEDLINE | ID: mdl-21839264

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are professional antigen-presenting cells able to induce immunity or tolerance. The interactions of immature DCs with naive T lymphocytes induce peripheral tolerance through mechanisms that include anergy or deletion of lymphocytes or the generation of regulatory T cells. Because of the central role of DCs in the immune response, they are potential targets for the induction of experimental tolerance. Thus, the generation of immature (tolerogenic) DCs able to capture and present alloantigens to T cells represents an important aim in our efforts to achieve better transplant acceptance. METHODS: In this work, we generated immature DCs by using vitamin D(3) (VD3) during the process of DC differentiation. RESULTS: The VD3-DCs showed an immature phenotype characterized by a low expression of major histocompatibility complex antigens of class II, CD86, and CD80 molecules and the secretion of a tolerogenic cytokine pattern. Furthermore, we showed that VD3-DCs phagocytose apoptotic allogeneic cells efficiently without inducing DC maturation or activation. Most important, our experiments demonstrated that mice treated with VD3 produce immature DCs in vivo, and that DCs from VD3-treated mice immunized with allogeneic apoptotic cells maintained their tolerogenic phenotype. CONCLUSION: Our results show that allogeneic apoptotic cells in combination with VD3 generate DCs with tolerogenic characteristics that could be used to induce tolerance towards alloantigens.


Subject(s)
Apoptosis , Dendritic Cells/immunology , Immune Tolerance , Isoantigens/immunology , Phagocytosis , Thymocytes/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , B7-1 Antigen/immunology , Cell Differentiation , Cells, Cultured , Cholecalciferol/pharmacology , Cytokines/metabolism , Dendritic Cells/drug effects , Histocompatibility Antigens Class II/immunology , Immunophenotyping , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Necrosis , Phenotype , Thymocytes/pathology , Thymocytes/transplantation , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...