Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
Am J Physiol Heart Circ Physiol ; 327(2): H533-H544, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38995212

ABSTRACT

The thymus, where T lymphocytes develop and mature, is sensitive to insults such as tissue ischemia or injury. The insults can cause thymic atrophy and compromise T-cell development, potentially impairing adaptive immunity. The objective of this study was to investigate whether myocardial infarction (MI) induces thymic injury to impair T lymphopoiesis and to uncover the underlying mechanisms. When compared with sham controls, MI mice at day 7 post-MI exhibited smaller thymus, lower cellularity, as well as less thymocytes at different developmental stages, indicative of T-lymphopoiesis impairment following MI. Accordingly, the spleen of MI mice has less T cells and recent thymic emigrants (RTEs), implying that the thymus of MI mice releases fewer mature thymocytes than sham controls. Interestingly, the secretory function of splenic T cells was not affected by MI. Further experiments showed that the reduction of thymocytes in MI mice was due to increased thymocyte apoptosis. Removal of adrenal glands by adrenalectomy (ADX) prevented MI-induced thymic injury and dysfunction, whereas corticosterone supplementation in ADX + MI mice reinduced thymic injury and dysfunction, indicating that glucocorticoids mediate thymic damage triggered by MI. Eosinophils play essential roles in thymic regeneration postirradiation, and eosinophil-deficient mice exhibit impaired thymic recovery after sublethal irradiation. Interestingly, the thymus was fully regenerated in both wild-type and eosinophil-deficient mice at day 14 post-MI, suggesting that eosinophils are not critical for thymus regeneration post-MI. In conclusion, our study demonstrates that MI-induced glucocorticoids trigger thymocyte apoptosis and impair T lymphopoiesis, resulting in less mature thymocyte release to the spleen.NEW & NOTEWORTHY The thymus is essential for maintaining whole body T-cell output. Thymic injury can adversely affect T lymphopoiesis and T-cell immune response. This study demonstrates that MI induces thymocyte apoptosis and compromises T lymphopoiesis, resulting in fewer releases of mature thymocytes to the spleen. This process is mediated by glucocorticoids secreted by adrenal glands. Therefore, targeting glucocorticoids represents a novel approach to attenuate post-MI thymic injury.


Subject(s)
Adrenalectomy , Apoptosis , Lymphopoiesis , Mice, Inbred C57BL , Myocardial Infarction , Thymus Gland , Animals , Thymus Gland/pathology , Thymus Gland/immunology , Thymus Gland/drug effects , Myocardial Infarction/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/immunology , Myocardial Infarction/physiopathology , Male , Thymocytes/metabolism , Thymocytes/pathology , Thymocytes/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Glucocorticoids/pharmacology , Eosinophils/metabolism , Eosinophils/immunology , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Disease Models, Animal , Mice , Corticosterone/blood
2.
Cell Rep ; 43(6): 114261, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38776224

ABSTRACT

Thymocyte development requires precise control of PI3K-Akt signaling to promote proliferation and prevent leukemia and autoimmune disorders. Here, we show that ablating individual clusters of the miR-17∼92 family has a negligible effect on thymocyte development, while deleting the entire family severely impairs thymocyte proliferation and reduces thymic cellularity, phenocopying genetic deletion of Dicer. Mechanistically, miR-17∼92 expression is induced by Myc-mediated pre-T cell receptor (TCR) signaling, and miR-17∼92 promotes thymocyte proliferation by suppressing the translation of Pten. Retroviral expression of miR-17∼92 restores the proliferation and differentiation of Myc-deficient thymocytes. Conversely, partial deletion of the miR-17∼92 family significantly delays Myc-driven leukemogenesis. Intriguingly, thymocyte-specific transgenic miR-17∼92 expression does not cause leukemia or lymphoma but instead aggravates skin inflammation, while ablation of the miR-17∼92 family ameliorates skin inflammation. This study reveals intricate roles of the miR-17∼92 family in balancing thymocyte development, leukemogenesis, and autoimmunity and identifies those microRNAs (miRNAs) as potential therapeutic targets for leukemia and autoimmune diseases.


Subject(s)
Autoimmunity , Leukemia , MicroRNAs , Thymocytes , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Thymocytes/metabolism , Thymocytes/pathology , Autoimmunity/genetics , Mice , Leukemia/pathology , Leukemia/genetics , Cell Proliferation , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Cell Differentiation/genetics , Signal Transduction , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinogenesis/metabolism
3.
Leukemia ; 37(5): 1039-1047, 2023 05.
Article in English | MEDLINE | ID: mdl-36997676

ABSTRACT

Hematopoietic stem cells (HSCs) and leukemia stem cells (LSCs) have robust self-renewal potential, which is responsible for sustaining normal and malignant hematopoiesis, respectively. Although considerable efforts have been made to explore the regulation of HSC and LSC maintenance, the underlying molecular mechanism remains obscure. Here, we observe that the expression of thymocyte-expressed, positive selection-associated 1 (Tespa1) is markedly increased in HSCs after stresses exposure. Of note, deletion of Tespa1 results in short-term expansion but long-term exhaustion of HSCs in mice under stress conditions due to impaired quiescence. Mechanistically, Tespa1 can interact with CSN subunit 6 (CSN6), a subunit of COP9 signalosome, to prevent ubiquitination-mediated degradation of c-Myc protein in HSCs. As a consequence, forcing c-Myc expression improves the functional defect of Tespa1-null HSCs. On the other hand, Tespa1 is identified to be highly enriched in human acute myeloid leukemia (AML) cells and is essential for AML cell growth. Furthermore, using MLL-AF9-induced AML model, we find that Tespa1 deficiency suppresses leukemogenesis and LSC maintenance. In summary, our findings reveal the important role of Tespa1 in promoting HSC and LSC maintenance and therefore provide new insights on the feasibility of hematopoietic regeneration and AML treatment.


Subject(s)
Leukemia, Myeloid, Acute , Thymocytes , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Thymocytes/pathology
4.
BMC Cancer ; 22(1): 1216, 2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36434543

ABSTRACT

BACKGROUND: Ovarian cancer is one of the most lethal gynecologic malignancies with a dismal prognosis that poses a serious threat to human health, highlighting the need for more knowledge about what is required for identifying some biomarkers for early diagnosis, prediction of prognosis and disease monitoring. TOX, a critical transcription factor related to the development of malignancies that contributing to lymphocytes not just T cells, had been proved prognostic value in some spectrum of cancers. Here, we aimed to study the prognostic role of TOX in ovarian cancer. RESULTS: We found that TOX was not only expressed in CD8 T cells but also tumor cells. TOX expression score was higher in ovarian cancer tissues and correlated with survival status. Survival analysis revealed that ovarian cancer patients with high TOX expression score generally shorter overall survival and disease-free survival times. Univariate and Multivariate Cox demonstrated that TOX expression score could be used as an independent prognostic factor for patients with ovarian cancer. CONCLUSION: TOX expression in ovarian cancer could be a promising tool for predict overall survival of ovarian cancer patients.


Subject(s)
Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/pathology , Thymocytes/pathology , Carcinoma, Ovarian Epithelial , Lymphocyte Activation , Prognosis
5.
Proc Natl Acad Sci U S A ; 119(32): e2201493119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35921443

ABSTRACT

Understanding the mechanisms promoting chromosomal translocations of the rearranging receptor loci in leukemia and lymphoma remains incomplete. Here we show that leukemias induced by aberrant activation of ß-catenin in thymocytes, which bear recurrent Tcra/Myc-Pvt1 translocations, depend on Tcf-1. The DNA double strand breaks (DSBs) in the Tcra site of the translocation are Rag-generated, whereas the Myc-Pvt1 DSBs are not. Aberrantly activated ß-catenin redirects Tcf-1 binding to novel DNA sites to alter chromatin accessibility and down-regulate genome-stability pathways. Impaired homologous recombination (HR) DNA repair and replication checkpoints lead to retention of DSBs that promote translocations and transformation of double-positive (DP) thymocytes. The resulting lymphomas, which resemble human T cell acute lymphoblastic leukemia (T-ALL), are sensitive to PARP inhibitors (PARPis). Our findings indicate that aberrant ß-catenin signaling contributes to translocations in thymocytes by guiding Tcf-1 to promote the generation and retention of replication-induced DSBs allowing their coexistence with Rag-generated DSBs. Thus, PARPis could offer therapeutic options in hematologic malignancies with active Wnt/ß-catenin signaling.


Subject(s)
Cell Transformation, Neoplastic , Genomic Instability , Hepatocyte Nuclear Factor 1-alpha , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Thymocytes , Translocation, Genetic , beta Catenin , Animals , Cell Transformation, Neoplastic/genetics , DNA Breaks, Double-Stranded , Genomic Instability/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-myc/genetics , RNA, Long Noncoding/genetics , Thymocytes/pathology , Translocation, Genetic/genetics , beta Catenin/genetics , beta Catenin/metabolism
6.
Phytomedicine ; 100: 154076, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35378414

ABSTRACT

BACKGROUND: Colorectal cancer was the second leading cause of mortality in 2019 and the number of new colorectal cancer cases was the highest in 2018 and 2019 in Japan. PURPOSE: The present study investigated the inhibitory effects of 2(S)-2',5,6',7-tetrahydroxyflavanone and 2 (R), 3(R)-2',3,5,6'-7-pentahydroxyflavanone on the incidence and growth of tumors in azoxymethane (AOM) plus dextran sulfate sodium (DSS)-treated mice. METHODS: The intraperitoneal administration of AOM (10 mg/kg) on day 0 induced colorectal carcinogenesis. Mice were given free and unlimited access to drinking water containing 1.5% (w/v) DSS on days 5 - 8, 30 - 33, and 56 - 57. They were orally administered tetra- and penta-hydroxyflavanones (10 and 30 mg/kg) for 10, 11, and 14 days followed by discontinuation intervals of 20 and 15 days. Cytokine, chemokine, programmed cell death-1 (PD-1), cyclooxygenase (COX)-2, and thymocyte selection-associated high mobility group box protein (TOX)/TOX2 expression levels were measured using their respective ELISA kits and an immunohistochemical analysis. RESULTS: The number and area of tumors decreased by 60.6 and 72.9% in mice administered 10 mg/kg tetra- and pentahydroxyflavanones, respectively, with reductions of 95.0 and 87.0% in Ki-67-positive cells, 91.7 and 92.7% in COX-2-postive cells, and 83.1 and 93.8% in TOX/TOX2-positive cells, respectively, in the colon. On the other hand, two tera- and pentahydroxyflavanone had no effect on p53 (a tumor suppressor by cell cycle arrest and apoptosis)-positive cells. The administration of 10 mg/kg tetra- and pentahydroxyflavanones to AOM/DSS-treated mice also resulted in decreases of 59.5 and 42.5% in IL-10 levels and 58.1 and 93.9% in PD-1 levels, respectively, in the colon. CONCLUSION: The inhibitory effects of tetra- and pentahydroxyflavanones on the growth of colon tumors in AOM/DSS-treated mice appear to be associated with decreases in the colon levels of IL-10 and PD-1 through the down-regulated expression of COX-2 and CD8+ T-cell exhaustion by TOX/TOX2 in the tumor microenvironment.


Subject(s)
Colitis-Associated Neoplasms , Colitis , Colonic Neoplasms , Animals , Apoptosis , Azoxymethane/toxicity , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colon/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/drug therapy , Colonic Neoplasms/prevention & control , Cyclooxygenase 2/metabolism , Dextran Sulfate/adverse effects , HMGB Proteins/metabolism , HMGB Proteins/pharmacology , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism , Scutellaria baicalensis , Thymocytes/metabolism , Thymocytes/pathology , Tumor Microenvironment
7.
Front Immunol ; 13: 797244, 2022.
Article in English | MEDLINE | ID: mdl-35185889

ABSTRACT

PTEN (Phosphatase and TENsin homolog) is a well-known tumor suppressor involved in numerous types of cancer, including T-cell acute lymphoblastic leukemia (T-ALL). In human, loss-of-function mutations of PTEN are correlated to mature T-ALL expressing a T-cell receptor (TCR) at their cell surface. In accordance with human T-ALL, inactivation of Pten gene in mouse thymocytes induces TCRαß+ T-ALL development. Herein, we explored the functional interaction between TCRαß signaling and PTEN. First, we performed single-cell RNA sequencing (scRNAseq) of PTEN-deficient and PTEN-proficient thymocytes. Bioinformatic analysis of our scRNAseq data showed that pathological Ptendel thymocytes express, as expected, Myc transcript, whereas inference of pathway activity revealed that these Ptendel thymocytes display a lower calcium pathway activity score compared to their physiological counterparts. We confirmed this result using ex vivo calcium flux assay and showed that upon TCR activation tumor Ptendel blasts were unable to release calcium ions (Ca2+) from the endoplasmic reticulum to the cytosol. In order to understand such phenomena, we constructed a mathematical model centered on the mechanisms controlling the calcium flux, integrating TCR signal strength and PTEN interactions. This qualitative model displays a dynamical behavior coherent with the dynamics reported in the literature, it also predicts that PTEN affects positively IP3 (inositol 1,4,5-trisphosphate) receptors (ITPR). Hence, we analyzed Itpr expression and unraveled that ITPR proteins levels are reduced in PTEN-deficient tumor cells compared to physiological and leukemic PTEN-proficient cells. However, calcium flux and ITPR proteins expression are not defective in non-leukemic PTEN-deficient T cells indicating that beyond PTEN loss an additional alteration is required. Altogether, our study shows that ITPR/Calcium flux is a part of the oncogenic landscape shaped by PTEN loss and pinpoints a putative role of PTEN in the regulation of ITPR proteins in thymocytes, which remains to be characterized.


Subject(s)
Calcium Signaling/genetics , PTEN Phosphohydrolase/deficiency , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Thymocytes/metabolism , Animals , Cell Proliferation/genetics , Mice , Mice, Transgenic , PTEN Phosphohydrolase/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Thymocytes/pathology
8.
Virology ; 567: 77-86, 2022 02.
Article in English | MEDLINE | ID: mdl-35032866

ABSTRACT

Type-I interferon (IFN-I) signals exert a critical role in disease progression during viral infections. However, the immunomodulatory mechanisms by which IFN-I dictates disease outcomes remain to be fully defined. Here we report that IFN-I signals mediate thymic atrophy in viral infections, with more severe and prolonged loss of thymic output and unique kinetics and subtypes of IFN-α/ß expression in chronic infection compared to acute infection. Loss of thymic output was linked to inhibition of early stages of thymopoiesis (DN1-DN2 transition, and DN3 proliferation) and pronounced apoptosis during the late DP stage. Notably, infection-associated thymic defects were largely abrogated upon ablation of IFNαßR and partially mitigated in the absence of CD8 T cells, thus implicating direct as well as indirect effects of IFN-I on thymocytes. These findings provide mechanistic underpinnings for immunotherapeutic strategies targeting IFN-1 signals to manipulate disease outcomes during chronic infections and cancers.


Subject(s)
Atrophy/virology , Interferon-alpha/immunology , Interferon-beta/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Thymocytes/virology , Thymus Gland/virology , Animals , Atrophy/genetics , Atrophy/immunology , Atrophy/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Chronic Disease , Female , Gene Expression Regulation , Humans , Immunologic Memory , Interferon-alpha/genetics , Interferon-beta/genetics , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/virology , Lymphocyte Depletion , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction/immunology , Single-Cell Analysis , Thymocytes/immunology , Thymocytes/pathology , Thymus Gland/immunology , Thymus Gland/pathology
9.
Sci Rep ; 11(1): 23221, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34853379

ABSTRACT

Deficiency of adenosine deaminase (ADA, EC3.5.4.4), a housekeeping enzyme intrinsic to the purine salvage pathway, leads to severe combined immunodeficiency (SCID) both in humans and mice. Lack of ADA results in the intracellular accumulation of toxic metabolites which have effects on T cell development and function. While untreated ADA-SCID is a fatal disorder, there are different therapeutic options available to restore ADA activity and reconstitute a functioning immune system, including enzyme replacement therapy (ERT). Administration of ERT in the form of pegylated bovine ADA (PEG-ADA) has proved a life-saving though non-curative treatment for ADA-SCID patients. However, in many patients treated with PEG-ADA, there is suboptimal immune recovery with low T and B cell numbers. Here, we show reduced thymus cellularity in ADA-SCID mice despite weekly PEG-ADA treatment. This was associated with lack of effective adenosine (Ado) detoxification in the thymus. We also show that thymocyte development in ADA-deficient thymi is arrested at the DN3-to-DN4 stage transition with thymocytes undergoing dATP-induced apoptosis rather than defective TCRß rearrangement or ß-selection. Our studies demonstrate at a detailed level that exogenous once-a-week enzyme replacement does not fully correct intra-thymic metabolic or immunological abnormalities associated with ADA deficiency.


Subject(s)
Adenosine Deaminase/therapeutic use , Agammaglobulinemia/drug therapy , Severe Combined Immunodeficiency/drug therapy , Thymocytes/pathology , Adenosine Deaminase/deficiency , Agammaglobulinemia/pathology , Animals , Cattle , Enzyme Replacement Therapy , Mice, SCID , Severe Combined Immunodeficiency/pathology , Thymocytes/drug effects , Thymocytes/metabolism
10.
Cancer Res ; 81(19): 5033-5046, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34321240

ABSTRACT

Mutations in the isocitrate dehydrogenase 1 (IDH1) and IDH2 genes are frequently observed in a wide variety of hematologic malignancies, including myeloid and T-cell leukemias. In this study, we generated Idh2R140Q transgenic mice to examine the role of the Idh2R140Q mutation in leukemia. No leukemia developed in Idh2R140Q transgenic mice, suggesting a need for additional genetic events for leukemia development. Because myeloid cells from NUP98-HOXD13 fusion (NHD13) transgenic mice frequently acquire somatic Idh mutations when they transform to acute myeloid leukemia, we generated Idh2R140Q/NHD13 double transgenic mice. Idh2R140Q/NHD13 transgenic mice developed an immature T-cell leukemia with an immunophenotype similar to double-negative 1 (DN1) or DN2 thymocytes. Idh2R140Q/NHD13 leukemic cells were enriched for an early thymic precursor transcriptional signature, and the gene expression profile for Idh2R140Q/NHD13 DN1/DN2 T-ALL closely matched that of human early/immature T-cell precursor (EITP) acute lymphoblastic leukemia (ALL). Moreover, recurrent mutations found in patients with EITP ALL, including KRAS, PTPN11, JAK3, SH2B3, and EZH2 were also found in Idh2R140Q/NHD13 DN1/DN2 T-ALL. In vitro treatment of Idh2R140Q/NHD13 thymocytes with enasidenib, a selective inhibitor of mutant IDH2, led to a marked decrease in leukemic cell proliferation. These findings demonstrate that Idh2R140Q/NHD13 mice can serve as a useful in vivo model for the study of early/immature thymocyte precursor acute lymphoblastic leukemia development and therapy. SIGNIFICANCE: T-cell leukemia induced in Idh2R140Q/NUP98-HOXD13 mice is immunophenotypically, transcriptionally, and genetically similar to human EITP ALL, providing a model for studying disease development and treatment.


Subject(s)
Homeodomain Proteins/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , Nuclear Pore Complex Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Thymocytes/metabolism , Animals , Biomarkers, Tumor , Cell Differentiation/genetics , Cell Line, Tumor , Computational Biology/methods , DNA Methylation , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Heterografts , Humans , Immunophenotyping , Mice , Mice, Transgenic , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Thymocytes/pathology , Transcriptome
11.
Front Immunol ; 12: 669893, 2021.
Article in English | MEDLINE | ID: mdl-34140950

ABSTRACT

Down syndrome (DS) patients prematurely show clinical manifestations usually associated with aging. Their immune system declines earlier than healthy individuals, leading to increased susceptibility to infections and higher incidence of autoimmune phenomena. Clinical features of accelerated aging indicate that trisomy 21 increases the biological age of tissues. Based on previous studies suggesting immune senescence in DS, we hypothesized that induction of cellular senescence may contribute to early thymic involution and immune dysregulation. Immunohistochemical analysis of thymic tissue showed signs of accelerated thymic aging in DS patients, normally seen in older healthy subjects. Moreover, our whole transcriptomic analysis on human Epcam-enriched thymic epithelial cells (hTEC), isolated from three DS children, which revealed disease-specific transcriptomic alterations. Gene set enrichment analysis (GSEA) of DS TEC revealed an enrichment in genes involved in cellular response to stress, epigenetic histone DNA modifications and senescence. Analysis of senescent markers and oxidative stress in hTEC and thymocytes confirmed these findings. We detected senescence features in DS TEC, thymocytes and peripheral T cells, such as increased ß-galactosidase activity, increased levels of the cell cycle inhibitor p16, telomere length and integrity markers and increased levels of reactive oxygen species (ROS), all factors contributing to cellular damage. In conclusion, our findings support the key role of cellular senescence in the pathogenesis of immune defect in DS while adding new players, such as epigenetic regulation and increased oxidative stress, to the pathogenesis of immune dysregulation.


Subject(s)
Cell Proliferation , Cellular Senescence , Down Syndrome/metabolism , Epithelial Cells/metabolism , Immunosenescence , Oxidative Stress , Thymocytes/metabolism , Thymus Gland/metabolism , Age Factors , Case-Control Studies , Cell Proliferation/genetics , Cellular Senescence/genetics , Child , Child, Preschool , Down Syndrome/genetics , Down Syndrome/immunology , Down Syndrome/pathology , Epigenesis, Genetic , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Gene Expression Profiling , Humans , Immunosenescence/genetics , Infant , Male , Oxidative Stress/genetics , Thymocytes/immunology , Thymocytes/pathology , Thymus Gland/immunology , Thymus Gland/pathology , Transcriptome
12.
Cell Death Dis ; 12(6): 526, 2021 05 22.
Article in English | MEDLINE | ID: mdl-34023853

ABSTRACT

Thymic atrophy in sepsis is a critical disadvantage because it induces immunosuppression and increases the mortality rate as the disease progresses. However, the exact mechanism of thymic atrophy has not been fully elucidated. In this study, we discovered a novel role for VSIG4-positive peritoneal macrophages (V4(+) cells) as the principal cells that induce thymic atrophy and thymocyte apoptosis. In CLP-induced mice, V4(+) cells were activated after ingestion of invading microbes, and the majority of these cells migrated into the thymus. Furthermore, these cells underwent a phenotypic shift from V4(+) to V4(-) and from MHC II(low) to MHC II(+). In coculture with thymocytes, V4(+) cells mainly induced apoptosis in DP thymocytes via the secretion of TNF-α. However, there was little effect on CD4 or CD8 SP and DN thymocytes. V4(-) cells showed low levels of activity compared to V4(+) cells. Thymic atrophy in CLP-induced V4(KO) mice was much less severe than that in CLP-induced wild-type mice. In addition, V4(KO) peritoneal macrophages also showed similar activity to V4(-) cells. Taken together, the current study demonstrates that V4(+) cells play important roles in inducing immunosuppression via thymic atrophy in the context of severe infection. These data also suggest that controlling the function of V4(+) cells may play a crucial role in the development of new therapies to prevent thymocyte apoptosis in sepsis.


Subject(s)
Macrophages, Peritoneal/physiology , Receptors, Complement/metabolism , Sepsis/pathology , Thymocytes/physiology , Animals , Apoptosis/genetics , Cecum/pathology , Cecum/surgery , Disease Models, Animal , Female , Ligation , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Punctures , Receptors, Complement/genetics , Sepsis/genetics , Sepsis/metabolism , Thymocytes/metabolism , Thymocytes/pathology , Tumor Necrosis Factor-alpha/metabolism
13.
Histochem Cell Biol ; 156(2): 133-146, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33993340

ABSTRACT

Immunosuppressive drugs such as cyclosporine A (CSA) can disrupt thymic structure and functions, ultimately inducing syngeneic/autologous graft-versus-host disease together with involuted medullas. To elucidate the effects of CSA on the thymus more precisely, we analyzed the effects of CSA on the thymus and T cell system using rats. In addition to confirming the phenomena already reported, we newly found that the proportion of recent thymic emigrants also greatly decreased, suggesting impaired supply. Immunohistologically, the medullary thymic epithelial cells (mTECs) presented with a relative decrease in the subset with a competent phenotype and downregulation of class II major histocompatibility complex molecules. In control rats, thymic dendritic cells (DCs) comprised two subsets, XCR1+SIRP1α-CD4- and XCR1-SIRP1α+CD4+. The former had a tendency to selectively localize in the previously-reported epithelium-containing areas of the rat medullas, and the number was significantly reduced by CSA treatment. The epithelium-free areas, another unique domains in the rat medullas, contained significantly more Foxp3+ thymic Tregs. With CSA treatment, the epithelium-free areas presented strong involution, and the number and distribution of Tregs in the medulla were greatly reduced. These results suggest that CSA inhibits the production of single-positive thymocytes, including Tregs, and disturbs the microenvironment of the thymic medulla, with a decrease of the competent mTECs and disorganization of epithelium-free areas and DC subsets, leading to a generation of autoreactive T cells with selective medullary involution.


Subject(s)
Cyclosporine/pharmacology , Epithelial Cells/drug effects , Forkhead Transcription Factors/analysis , Immunosuppressive Agents/pharmacology , T-Lymphocytes, Regulatory/drug effects , Thymus Gland/drug effects , Animals , Cyclosporine/administration & dosage , Dendritic Cells/drug effects , Dendritic Cells/pathology , Epithelial Cells/pathology , Forkhead Transcription Factors/metabolism , Immunohistochemistry , Immunosuppressive Agents/administration & dosage , Injections, Subcutaneous , Male , Optical Imaging , Rats , Rats, Inbred Lew , Receptors, Chemokine/analysis , Receptors, Chemokine/deficiency , Receptors, Chemokine/metabolism , T-Lymphocytes, Regulatory/pathology , Thymocytes/drug effects , Thymocytes/pathology , Thymus Gland/pathology
14.
Cell Rep ; 35(2): 108967, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852867

ABSTRACT

T lymphocyte differentiation in the steady state is characterized by high cellular turnover whereby thymocytes do not self-renew. However, if deprived of competent progenitors, the thymus can temporarily maintain thymopoiesis autonomously. This bears a heavy cost, because prolongation of thymus autonomy causes leukemia. Here, we show that, at an early stage, thymus autonomy relies on double-negative 3 early (DN3e) thymocytes that acquire stem-cell-like properties. Following competent progenitor deprivation, DN3e thymocytes become long lived, are required for thymus autonomy, differentiate in vivo, and include DNA-label-retaining cells. At the single-cell level, the transcriptional programs of thymopoiesis in autonomy and the steady state are similar. However, a new cell population emerges in autonomy that expresses an aberrant Notch target gene signature and bypasses the ß-selection checkpoint. In summary, DN3e thymocytes have the potential to self-renew and differentiate in vivo if cell competition is impaired, but this generates atypical cells, probably the precursors of leukemia.


Subject(s)
Hematopoiesis/genetics , Leukemia/genetics , Receptors, Notch/genetics , Thymocytes/immunology , Thymus Gland/immunology , Transcription Factors/genetics , Animals , Cell Differentiation , Cell Proliferation , EGF Family of Proteins/genetics , EGF Family of Proteins/immunology , Gene Expression Profiling , Gene Expression Regulation , Hematopoiesis/immunology , Humans , Immunophenotyping , Kidney , Leukemia/immunology , Leukemia/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Notch/immunology , Signal Transduction , Single-Cell Analysis , Thymocytes/classification , Thymocytes/pathology , Thymus Gland/pathology , Thymus Gland/transplantation , Transcription Factors/immunology , Transplantation, Heterotopic , Transplantation, Homologous
15.
Front Immunol ; 12: 696415, 2021.
Article in English | MEDLINE | ID: mdl-34987496

ABSTRACT

Disseminated infection with the high virulence strain of Mycobacterium avium 25291 leads to progressive thymic atrophy. We previously showed that M. avium-induced thymic atrophy results from increased glucocorticoid levels that synergize with nitric oxide (NO) produced by interferon gamma (IFNγ) activated macrophages. Where and how these mediators act is not understood. We hypothesized that IFNγ and NO promote thymic atrophy through their effects on bone marrow (BM) T cell precursors and T cell differentiation in the thymus. We show that M. avium infection cause a reduction in the percentage and number of common lymphoid progenitors (CLP). Additionally, BM precursors from infected mice show an overall impaired ability to reconstitute thymi of RAGKO mice, in part due to IFNγ. Thymi from infected mice present an IFNγ and NO-driven inflammation. When transplanted under the kidney capsule of uninfected mice, thymi from infected mice are unable to sustain T cell differentiation. Finally, we observed increased thymocyte death via apoptosis after infection, independent of both IFNγ and iNOS; and a decrease on active caspase-3 positive thymocytes, which is not observed in the absence of iNOS expression. Together our data suggests that M. avium-induced thymic atrophy results from a combination of defects mediated by IFNγ and NO, including alterations in the BM T cell precursors, the thymic structure and the thymocyte differentiation.


Subject(s)
Bone Marrow/pathology , Interferon-gamma/physiology , Lymphoid Progenitor Cells/pathology , Nitric Oxide Synthase Type II/physiology , Thymus Gland/pathology , Tuberculosis/pathology , Animals , Apoptosis , Atrophy , Bone Marrow Transplantation , Cell Differentiation , DNA-Binding Proteins/deficiency , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mycobacterium avium , Nitric Oxide/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Thymocytes/pathology , Thymus Gland/transplantation , Tuberculosis/immunology
16.
J Cell Physiol ; 236(2): 1375-1390, 2021 02.
Article in English | MEDLINE | ID: mdl-33128793

ABSTRACT

Myeloid-related protein 8/14 (MRP8/14) participates in various inflammatory responses, however, its effect on macrophage efferocytosis remains unclear. Here, we demonstrate that MRP8/14 significantly inhibits the efferocytosis of apoptotic thymocytes by mouse bone marrow-derived macrophages (BMDMs), which later proves to be associated with the receptor for advanced glycation end products (RAGE) or for reducing the expression of growth arrest-specific protein 6 and milk fat globule epidermal growth factor 8, independent of RAGE. Furthermore, MRP8/14 promotes polarization of BMDMs from the M2 - to M1 -like phenotype by upregulating expression of M1 -related surface receptor proteins and signature M1 -marker genes and by downregulating signature M2 -marker gene expression, which depends on Toll-like receptor 4 and p38 mitogen-activated protein kinase/nuclear factor κB pathways. Thus, we report a significant inhibitory effect of MRP8/14 on macrophage efferocytosis and MRP8/14-mediated phenotypic polarization, which may be helpful in developing novel therapeutic strategies leading to inflammation resolution.


Subject(s)
Antigens, Surface/genetics , Calgranulin A/genetics , Inflammation/genetics , Intercellular Signaling Peptides and Proteins/genetics , Milk Proteins/genetics , Toll-Like Receptor 4/genetics , Animals , Apoptosis/genetics , Cell Polarity/genetics , Humans , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , NF-kappa B/genetics , Phagocytosis/genetics , Receptor for Advanced Glycation End Products/genetics , Signal Transduction , Thymocytes/metabolism , Thymocytes/pathology , p38 Mitogen-Activated Protein Kinases/genetics
17.
J Cell Physiol ; 236(6): 4725-4737, 2021 06.
Article in English | MEDLINE | ID: mdl-33269476

ABSTRACT

The differentiation of mature medullary thymic epithelial cells (mTECs) is critical for the induction of central immune tolerance. Although the critical effect of mechanistic target of rapamycin complex 1 (mTORC1) in shaping mTEC differentiation has been studied, the regulatory role of mTORC2 in the differentiation and maturation of mTECs is poorly understood. We herein reported that TEC-specific ablation of a rapamycin-insensitive companion of mTOR (RICTOR), a key component of mTORC2, significantly decreased the thymus size and weight, the total cell number of TECs, and the cell number of mTECs with a smaller degree of reduced cortical thymic epithelial cells. Interestingly, RICTOR deficiency significantly accelerated the mTEC maturation process, as indicated by the increased ratios of mature mTECs (MHCIIhi , CD80+ , and Aire+ ) to immature mTECs (MHCIIlo , CD80- , and Aire- ) in Rictor-deficient mice. The RNA-sequencing assays showed that the upregulated nuclear factor-κB (NF-κB) signaling pathway in Rictor-deficient mTECs was one of the obviously altered pathways compared with wild-type mTECs. Our studies further showed that Rictor-deficient mTECs exhibited upregulated expression of receptor activator of NF-κB (RANK) and lymphotoxin ß receptor (LTßR), as well as increased activity of canonical and noncanonical NF-κB signaling pathways as determined by ImageStream and Simple Western. Finally, our results showed that inhibition of NF-κB signaling pathways could partially reverse the accelerated maturation of mTECs in Rictor conditional KO mice. Thus, mTORC2 negatively controls the kinetics of the mTEC maturation process by inhibiting the LTßR/RANK-NF-κB signal axis.


Subject(s)
Cell Differentiation , Epithelial Cells/enzymology , Lymphotoxin beta Receptor/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , NF-kappa B/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Thymus Gland/enzymology , Animals , Epithelial Cells/pathology , Gene Expression Regulation , Kinetics , Lymphotoxin beta Receptor/genetics , Mechanistic Target of Rapamycin Complex 2/genetics , Mice, Knockout , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Signal Transduction , Thymocytes/enzymology , Thymocytes/pathology , Thymus Gland/pathology
18.
Front Immunol ; 11: 575154, 2020.
Article in English | MEDLINE | ID: mdl-33178202

ABSTRACT

Natural IgM antibodies (NAbs) have been shown to recognize injury-associated neoepitopes and to initiate pathogenic complement activation. The NAb termed C2 binds to a subset of phospholipids displayed on injured cells, and its role(s) in arthritis, as well as the potential therapeutic benefit of a C2 NAb-derived ScFv-containing protein fused to a complement inhibitor, complement receptor-related y (Crry), on joint inflammation are unknown. Our first objective was to functionally test mAb C2 binding to apoptotic cells from the joint and also evaluate its inflammation enhancing capacity in collagen antibody-induced arthritis (CAIA). The second objective was to generate and test the complement inhibitory capacity of C2-Crry fusion protein in the collagen-induced arthritis (CIA) model. The third objective was to demonstrate in vivo targeting of C2-Crry to damaged joints in mice with arthritis. The effect of C2-NAb on CAIA in C57BL/6 mice was examined by inducing a suboptimal disease. The inhibitory effect of C2-Crry in DBA/1J mice with CIA was determined by injecting 2x per week with a single dose of 0.250 mg/mouse. Clinical disease activity (CDA) was examined, and knee joints were fixed for analysis of histopathology, C3 deposition, and macrophage infiltration. In mice with suboptimal CAIA, at day 10 there was a significant (p < 0.017) 74% increase in the CDA in mice treated with C2 NAb, compared to mice treated with F632 control NAb. In mice with CIA, at day 35 there was a significant 39% (p < 0.042) decrease in the CDA in mice treated with C2-Crry. Total scores for histopathology were also 50% decreased (p < 0.0005) in CIA mice treated with C2-Crry. C3 deposition was significantly decreased in the synovium (44%; p < 0.026) and on the surface of cartilage (42%; p < 0.008) in mice treated with C2-Crry compared with PBS treated CIA mice. Furthermore, C2-Crry specifically bound to apoptotic fibroblast-like synoviocytes in vitro, and also localized in the knee joints of arthritic mice as analyzed by in vivo imaging. In summary, NAb C2 enhanced arthritis-related injury, and targeted delivery of C2-Crry to inflamed joints demonstrated disease modifying activity in a mouse model of human inflammatory arthritis.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Experimental/drug therapy , Complement Activation/drug effects , Immunoglobulin M/pharmacology , Joints/drug effects , Receptors, Complement 3b/metabolism , Single-Chain Antibodies/pharmacology , Synoviocytes/drug effects , Animals , Apoptosis/drug effects , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Cells, Cultured , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Joints/immunology , Joints/metabolism , Joints/pathology , Mice, Inbred C57BL , Mice, Inbred DBA , Recombinant Fusion Proteins/pharmacology , Synoviocytes/immunology , Synoviocytes/metabolism , Synoviocytes/pathology , Thymocytes/drug effects , Thymocytes/immunology , Thymocytes/metabolism , Thymocytes/pathology
19.
Front Immunol ; 11: 1745, 2020.
Article in English | MEDLINE | ID: mdl-32903477

ABSTRACT

Even though the thymus is exquisitely sensitive to acute insults like infection, shock, or common cancer therapies such as cytoreductive chemo- or radiation-therapy, it also has a remarkable capacity for repair. This phenomenon of endogenous thymic regeneration has been known for longer even than its primary function to generate T cells, however, the underlying mechanisms controlling the process have been largely unstudied. Although there is likely continual thymic involution and regeneration in response to stress and infection in otherwise healthy people, acute and profound thymic damage such as that caused by common cancer cytoreductive therapies or the conditioning regimes as part of hematopoietic cell transplantation (HCT), leads to prolonged T cell deficiency; precipitating high morbidity and mortality from opportunistic infections and may even facilitate cancer relapse. Furthermore, this capacity for regeneration declines with age as a function of thymic involution; which even at steady state leads to reduced capacity to respond to new pathogens, vaccines, and immunotherapy. Consequently, there is a real clinical need for strategies that can boost thymic function and enhance T cell immunity. One approach to the development of such therapies is to exploit the processes of endogenous thymic regeneration into novel pharmacologic strategies to boost T cell reconstitution in clinical settings of immune depletion such as HCT. In this review, we will highlight recent work that has revealed the mechanisms by which the thymus is capable of repairing itself and how this knowledge is being used to develop novel therapies to boost immune function.


Subject(s)
Cell Proliferation , Epithelial Cells/pathology , Regeneration , Thymocytes/pathology , Thymus Gland/physiopathology , Animals , Cell Communication , Cell Proliferation/drug effects , Cellular Microenvironment , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Immunologic Factors/therapeutic use , Regeneration/drug effects , Signal Transduction , Thymocytes/drug effects , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/drug effects , Thymus Gland/immunology , Thymus Gland/pathology
20.
Neuroimmunomodulation ; 27(1): 28-37, 2020.
Article in English | MEDLINE | ID: mdl-32155637

ABSTRACT

BACKGROUND: Previous work revealed the existence of a severe thymic atrophy with massive loss of immature CD4+CD8+ thymocytes in animals developing insulin-dependent diabetes, chemically induced by alloxan. Furthermore, the intrathymic expression of chemokines, such as CXCL12, is changed in these animals, suggesting that cell migration-related patterns may be altered. One molecular interaction involved in normal thymocyte migration is that mediated by soluble semaphorin-3A and its cognate receptor neuropilin-1. OBJECTIVES: We investigated herein the expression and role of semaphorin-3A in the migratory responses of thymocytes from alloxan-induced diabetic mice. We characterized semaphorin-3A and its receptor, neuropilin-1, in thymuses from control and diabetic mice as well as semaphorin-3A-dependent migration of developing thymocytes in both control and diabetic animals. METHODS: Diabetes was chemically induced after a single injection of alloxan in young adult BALB/c mice. Thymocytes were excised from control and diabetic individuals and subjected to cytofluorometry for simultaneous detection of semaphorin-3A or neuropilin-1 in CD4/CD8-defined subsets. Cell migration in response to semaphorin-3A was performed using cell migration transwell chambers. RESULTS: Confirming previous data, we observed a severe decrease in the total numbers of thymocytes in diabetic mice, which comprised alterations in both immature (double-negative subpopulations) and mature CD4/CD8-defined thymocyte subsets. These were accompanied by a decrease in the absolute numbers of semaphorin-3A-bearing thymocytes, comprising CD4-CD8-, CD4+CD8+, and CD4-CD8+ cells. Additionally, immature CD4-CD8- and CD4+CD8+ developing T cells exhibited a decrease in the membrane density of semaphorin-3A. The relative and absolute numbers of neuropilin-1-positive thymocytes were also decreased in diabetic mouse thymocytes compared to controls, as seen in CD4-CD8-, CD4+CD8+, and CD4-CD8+ cell subpopulations. Functionally, we observed a decrease in the chemorepulsive role of semaphorin-3A, as revealed by transwell migration chambers. Such an effect was seen in all immature and mature thymocyte subsets. CONCLUSIONS: Taken together, our data clearly unravel a disruption in the normal cell migration pattern of developing thymocytes following chemically induced insulin-dependent diabetes, as ascertained by the altered migratory response to sempahorin-3A. In conceptual terms, it is plausible to think that such disturbances in the migration pattern of thymocytes from these diabetic animals may exert an impact in the cell-mediated immune response of these mice.


Subject(s)
Cell Movement/immunology , Diabetes Mellitus, Experimental/immunology , Semaphorin-3A/metabolism , Thymocytes/pathology , Animals , Diabetes Mellitus, Type 1/immunology , Male , Mice , Mice, Inbred BALB C , Thymocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL