Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Cell ; 183(3): 752-770.e22, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33125891

ABSTRACT

A greater understanding of hematopoietic stem cell (HSC) regulation is required for dissecting protective versus detrimental immunity to pathogens that cause chronic infections such as Mycobacterium tuberculosis (Mtb). We have shown that systemic administration of Bacille Calmette-Guérin (BCG) or ß-glucan reprograms HSCs in the bone marrow (BM) via a type II interferon (IFN-II) or interleukin-1 (IL1) response, respectively, which confers protective trained immunity against Mtb. Here, we demonstrate that, unlike BCG or ß-glucan, Mtb reprograms HSCs via an IFN-I response that suppresses myelopoiesis and impairs development of protective trained immunity to Mtb. Mechanistically, IFN-I signaling dysregulates iron metabolism, depolarizes mitochondrial membrane potential, and induces cell death specifically in myeloid progenitors. Additionally, activation of the IFN-I/iron axis in HSCs impairs trained immunity to Mtb infection. These results identify an unanticipated immune evasion strategy of Mtb in the BM that controls the magnitude and intrinsic anti-microbial capacity of innate immunity to infection.


Subject(s)
Hematopoietic Stem Cells/microbiology , Immunity , Mycobacterium tuberculosis/physiology , Myelopoiesis , Animals , Bone Marrow Cells/metabolism , Cell Proliferation , Disease Susceptibility , Homeostasis , Interferon Type I/metabolism , Iron/metabolism , Kinetics , Lung/microbiology , Lung/pathology , Macrophages/immunology , Mice, Inbred C57BL , Myeloid Cells/metabolism , Necrosis , Signal Transduction , Transcription, Genetic , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/pathology
2.
Cell ; 172(1-2): 176-190.e19, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29328912

ABSTRACT

The dogma that adaptive immunity is the only arm of the immune response with memory capacity has been recently challenged by several studies demonstrating evidence for memory-like innate immune training. However, the underlying mechanisms and location for generating such innate memory responses in vivo remain unknown. Here, we show that access of Bacillus Calmette-Guérin (BCG) to the bone marrow (BM) changes the transcriptional landscape of hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs), leading to local cell expansion and enhanced myelopoiesis at the expense of lymphopoiesis. Importantly, BCG-educated HSCs generate epigenetically modified macrophages that provide significantly better protection against virulent M. tuberculosis infection than naïve macrophages. By using parabiotic and chimeric mice, as well as adoptive transfer approaches, we demonstrate that training of the monocyte/macrophage lineage via BCG-induced HSC reprogramming is sustainable in vivo. Our results indicate that targeting the HSC compartment provides a novel approach for vaccine development.


Subject(s)
Hematopoietic Stem Cells/immunology , Immunity, Innate , Immunologic Memory , Mycobacterium bovis/immunology , Transcriptome , Animals , Cell Line , Cells, Cultured , Epigenesis, Genetic , Hematopoiesis , Mice , Mice, Inbred C57BL , Tuberculosis/immunology
3.
Immunology ; 172(1): 109-126, 2024 May.
Article in English | MEDLINE | ID: mdl-38316548

ABSTRACT

Dendritic cells (DCs) are the most significant antigen presenting cells of the immune system, critical for the activation of naïve T cells. The pathways controlling DC development, maturation, and effector function therefore require precise regulation to allow for an effective induction of adaptive immune response. MYSM1 is a chromatin binding deubiquitinase (DUB) and an activator of gene expression via its catalytic activity for monoubiquitinated histone H2A (H2A-K119ub), which is a highly abundant repressive epigenetic mark. MYSM1 is an important regulator of haematopoiesis in mouse and human, and a systemic constitutive loss of Mysm1 in mice results in a depletion of many haematopoietic progenitors, including DC precursors, with the downstream loss of most DC lineage cells. However, the roles of MYSM1 at the later checkpoints in DC development, maturation, activation, and effector function at present remain unknown. In the current work, using a range of novel mouse models (Mysm1flCreERT2, Mysm1flCD11c-cre, Mysm1DN), we further the understanding of MYSM1 functions in the DC lineage: assessing the requirement for MYSM1 in DC development independently of other complex developmental phenotypes, exploring its role at the later checkpoints in DC maintenance and activation in response to microbial stimulation, and testing the requirement for the DUB catalytic activity of MYSM1 in these processes. Surprisingly, we demonstrate that MYSM1 expression and catalytic activity in DCs are dispensable for the maintenance of DC numbers in vivo or for DC activation in response to microbial stimulation. In contrast, MYSM1 acts via its DUB catalytic activity specifically in haematopoietic progenitors to allow normal DC lineage development, and its loss results not only in a severe DC depletion but also in the production of functionally altered DCs, with a dysregulation of many housekeeping transcriptional programs and significantly altered responses to microbial stimulation.


Subject(s)
Trans-Activators , Ubiquitin-Specific Proteases , Animals , Humans , Mice , Cell Differentiation , Chromatin/genetics , Dendritic Cells/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , Histones/metabolism , Mice, Knockout , Trans-Activators/genetics , Trans-Activators/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
4.
Immunology ; 170(4): 553-566, 2023 12.
Article in English | MEDLINE | ID: mdl-37688495

ABSTRACT

Rheumatoid arthritis is a chronic and systemic inflammatory disease that affects approximately 1% of the world's population and is characterised by joint inflammation, the destruction of articular cartilage and bone, and many potentially life-threatening extraarticular manifestations. B lymphocytes play a central role in the pathology of rheumatoid arthritis as the precursors of autoantibody secreting plasma cells, as highly potent antigen-presenting cells, and as a source of various inflammatory cytokines, however, the effects of rheumatoid arthritis on B lymphocyte development remain poorly understood. Here, we analyse B lymphocyte development in murine models of rheumatoid arthritis, quantifying all the subsets of B cell precursors in the bone marrow and splenic B cells using flow cytometry. We demonstrate a severe reduction in pre-B cells and immature B cells in the bone marrow of mice with active disease, despite no major effects on the mature naïve B cell numbers. The loss of B cell precursors in the bone marrow of the affected mice was associated with a highly significant reduction in the proportion of Ki67+ cells, indicating impaired cell proliferation, while the viability of the B cell precursors was not significantly affected. We also observed some mobilisation of the B cell precursor cells into the mouse spleen, demonstrated with flow cytometry and pre-B colony forming units assays. In summary, the current work demonstrates a severe dysregulation in B lymphocyte development in murine rheumatoid arthritis, with possible implications for B cell repertoire formation, tolerance induction, and disease mechanisms.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Mice , Animals , Disease Models, Animal , B-Lymphocytes , Immune Tolerance
5.
J Cell Mol Med ; 25(14): 7089-7094, 2021 07.
Article in English | MEDLINE | ID: mdl-34114734

ABSTRACT

MYSM1 is a chromatin-binding protein, widely investigated for its functions in haematopoiesis in human and mouse; however, its role in haematologic malignancies remains unexplored. Here, we investigate the cross-talk between MYSM1 and oncogenic cMYC in the transcriptional regulation of genes encoding ribosomal proteins, and the implications of these mechanisms for cMYC-driven carcinogenesis. We demonstrate that in cMYC-driven B cell lymphoma in mouse models, MYSM1-loss represses ribosomal protein gene expression and protein synthesis. Importantly, the loss of MYSM1 also strongly inhibits cMYC oncogenic activity and protects against B cell lymphoma onset and progression in the mouse models. This advances the understanding of the molecular and transcriptional mechanisms of lymphomagenesis, and suggests MYSM1 as a possible drug target for cMYC-driven malignancies.


Subject(s)
Lymphoma, B-Cell/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Trans-Activators/deficiency , Ubiquitin-Specific Proteases/deficiency , Animals , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Mice , Proto-Oncogene Proteins c-myc/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
6.
Int J Mol Sci ; 21(8)2020 Apr 24.
Article in English | MEDLINE | ID: mdl-32344625

ABSTRACT

MYSM1 has emerged as an important regulator of hematopoietic stem cell function, blood cell production, immune response, and other aspects of mammalian physiology. It is a metalloprotease family protein with deubiquitinase catalytic activity, as well as SANT and SWIRM domains. MYSM1 normally localizes to the nucleus, where it can interact with chromatin and regulate gene expression, through deubiquitination of histone H2A and non-catalytic contacts with other transcriptional regulators. A cytosolic form of MYSM1 protein was also recently described and demonstrated to regulate signal transduction pathways of innate immunity, by promoting the deubiquitination of TRAF3, TRAF6, and RIP2. In this work we review the current knowledge on the molecular mechanisms of action of MYSM1 protein in transcriptional regulation, signal transduction, and potentially other cellular processes. The functions of MYSM1 in different cell types and aspects of mammalian physiology are also reviewed, highlighting the key checkpoints in hematopoiesis, immunity, and beyond regulated by MYSM1. Importantly, mutations in MYSM1 in human were recently linked to a rare hereditary disorder characterized by leukopenia, anemia, and other hematopoietic and developmental abnormalities. Our growing knowledge of MYSM1 functions and mechanisms of actions sheds important insights into its role in mammalian physiology and the etiology of the MYSM1-deficiency disorder in human.


Subject(s)
Deubiquitinating Enzymes/genetics , Hematopoiesis , Trans-Activators/genetics , Ubiquitin-Specific Proteases/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Deubiquitinating Enzymes/metabolism , Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunity, Innate , Signal Transduction , Structure-Activity Relationship , Trans-Activators/chemistry , Trans-Activators/metabolism , Ubiquitin-Specific Proteases/chemistry , Ubiquitin-Specific Proteases/metabolism
7.
Infect Immun ; 88(1)2019 12 17.
Article in English | MEDLINE | ID: mdl-31636138

ABSTRACT

Salmonella is an intracellular bacterium found in the gastrointestinal tract of mammalian, avian, and reptilian hosts. Mouse models have been extensively used to model in vivo distinct aspects of human Salmonella infections and have led to the identification of several host susceptibility genes. We have investigated the susceptibility of Collaborative Cross strains to intravenous infection with Salmonella enterica serovar Typhimurium as a model of human systemic invasive infection. In this model, strain CC042/GeniUnc (CC042) mice displayed extreme susceptibility with very high bacterial loads and mortality. CC042 mice showed lower spleen weights and decreased splenocyte numbers before and after infection, affecting mostly CD8+ T cells, B cells, and all myeloid cell populations, compared with control C57BL/6J mice. CC042 mice also had lower thymus weights with a reduced total number of thymocytes and double-negative and double-positive (CD4+, CD8+) thymocytes compared to C57BL/6J mice. Analysis of bone marrow-resident hematopoietic progenitors showed a strong bias against lymphoid-primed multipotent progenitors. An F2 cross between CC042 and C57BL/6N mice identified two loci on chromosome 7 (Stsl6 and Stsl7) associated with differences in bacterial loads. In the Stsl7 region, CC042 carried a loss-of-function variant, unique to this strain, in the integrin alpha L (Itgal) gene, the causative role of which was confirmed by a quantitative complementation test. Notably, Itgal loss of function increased the susceptibility to S. Typhimurium in a (C57BL/6J × CC042)F1 mouse background but not in a C57BL/6J mouse inbred background. These results further emphasize the utility of the Collaborative Cross to identify new host genetic variants controlling susceptibility to infections and improve our understanding of the function of the Itgal gene.


Subject(s)
Bacteremia/genetics , CD11a Antigen/deficiency , Genetic Predisposition to Disease , Loss of Function Mutation , Salmonella Infections/genetics , Salmonella typhimurium/growth & development , Animals , Bacteremia/immunology , Bacteremia/pathology , Bacterial Load , Bone Marrow/pathology , Disease Models, Animal , Genes , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Salmonella Infections/immunology , Salmonella Infections/pathology , Serogroup , Spleen/pathology , Survival Analysis , Thymus Gland/pathology
8.
Immunology ; 151(1): 110-121, 2017 05.
Article in English | MEDLINE | ID: mdl-28066899

ABSTRACT

Several previous studies outlined the importance of the histone H2A deubiquitinase MYSM1 in the regulation of stem cell quiescence and haematopoiesis. In this study we investigated the role of MYSM1 in T-cell development. Using mouse models that allow conditional Mysm1 ablation at late stages of thymic development, we found that MYSM1 is intricately involved in the maintenance, activation and survival of CD8+ T cells. Mysm1 ablation resulted in a twofold reduction in CD8+ T-cell numbers, and also led to a hyperactivated CD8+ T-cell state accompanied by impaired proliferation and increased pro-inflammatory cytokine production after ex vivo stimulation. These phenotypes coincided with an increased apoptosis and preferential up-regulation of p53 tumour suppressor protein in CD8+ T cells. Lastly, we examined a model of experimental cerebral malaria, in which pathology is critically dependent on CD8+ T cells. In the mice conditionally deleted for Mysm1 in the T-cell compartment, CD8+ T-cell numbers remained reduced following infection, both in the periphery and in the brain, and the mice displayed improved survival after parasite challenge. Collectively, our data identify MYSM1 as a novel factor for CD8+ T cells in the immune system, increasing our understanding of the role of histone H2A deubiquitinases in cytotoxic T-cell biology.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Endopeptidases/metabolism , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic/genetics , Endopeptidases/genetics , Inflammation Mediators/metabolism , Lymphocyte Activation/genetics , Malaria, Cerebral/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Trans-Activators , Tumor Suppressor Protein p53/genetics , Ubiquitin-Specific Proteases
9.
Blood ; 125(15): 2344-8, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25710881

ABSTRACT

MYSM1 is a chromatin-binding transcriptional cofactor that deubiquitinates histone H2A. Studies of Mysm1-deficient mice have shown that it is essential for hematopoietic stem cell (HSC) function and lymphopoiesis. Human carriers of a rare MYSM1-inactivating mutation display similar lymphopoietic deficiencies. However, the mechanism by which MYSM1 regulates hematopoietic homeostasis remains unclear. Here, we show that Mysm1-deficiency results in p53 protein elevation in many hematopoietic cell types. p53 is a central regulator of cellular stress responses and HSC homeostasis. We thus generated double-knockout mice to assess a potential genetic interaction between Mysm1 and p53 in hematopoiesis. Mysm1(-/-)p53(-/-) mouse characterization showed a full rescue of Mysm1(-/-) developmental and hematopoietic defects. This included restoration of lymphopoiesis, and HSC numbers and functions. These results establish p53 activation as the driving mechanism for hematopoietic abnormalities in Mysm1 deficiency. Our findings may advance the understanding of p53 regulation in hematopoiesis and implicate MYSM1 as a potential p53 cofactor.


Subject(s)
Endopeptidases/genetics , Hematopoietic Stem Cells/pathology , Lymphopenia/genetics , Lymphopoiesis , Tumor Suppressor Protein p53/metabolism , Animals , Gene Deletion , Hematopoietic Stem Cells/metabolism , Lymphopenia/metabolism , Lymphopenia/pathology , Mice , Mice, Knockout , Trans-Activators , Tumor Suppressor Protein p53/genetics , Ubiquitin-Specific Proteases
10.
PLoS Genet ; 10(10): e1004688, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25356849

ABSTRACT

Spinster homolog 2 (Spns2) acts as a Sphingosine-1-phosphate (S1P) transporter in zebrafish and mice, regulating heart development and lymphocyte trafficking respectively. S1P is a biologically active lysophospholipid with multiple roles in signalling. The mechanism of action of Spns2 is still elusive in mammals. Here, we report that Spns2-deficient mice rapidly lost auditory sensitivity and endocochlear potential (EP) from 2 to 3 weeks old. We found progressive degeneration of sensory hair cells in the organ of Corti, but the earliest defect was a decline in the EP, suggesting that dysfunction of the lateral wall was the primary lesion. In the lateral wall of adult mutants, we observed structural changes of marginal cell boundaries and of strial capillaries, and reduced expression of several key proteins involved in the generation of the EP (Kcnj10, Kcnq1, Gjb2 and Gjb6), but these changes were likely to be secondary. Permeability of the boundaries of the stria vascularis and of the strial capillaries appeared normal. We also found focal retinal degeneration and anomalies of retinal capillaries together with anterior eye defects in Spns2 mutant mice. Targeted inactivation of Spns2 in red blood cells, platelets, or lymphatic or vascular endothelial cells did not affect hearing, but targeted ablation of Spns2 in the cochlea using a Sox10-Cre allele produced a similar auditory phenotype to the original mutation, suggesting that local Spns2 expression is critical for hearing in mammals. These findings indicate that Spns2 is required for normal maintenance of the EP and hence for normal auditory function, and support a role for S1P signalling in hearing.


Subject(s)
Anion Transport Proteins/genetics , Cochlea/pathology , Ear, Inner/pathology , Hearing Loss/genetics , Age of Onset , Animals , Anion Transport Proteins/deficiency , Anion Transport Proteins/metabolism , Anterior Eye Segment/metabolism , Anterior Eye Segment/pathology , Cochlea/metabolism , Connexin 26 , Connexins , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hearing Loss/metabolism , Hearing Loss/pathology , Lysophospholipids/metabolism , Mice , Organogenesis/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Stria Vascularis/pathology , Zebrafish
11.
Blood ; 119(6): 1370-9, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22184403

ABSTRACT

Stem cell differentiation and lineage specification depend on coordinated programs of gene expression, but our knowledge of the chromatin-modifying factors regulating these events remains incomplete. Ubiquitination of histone H2A (H2A-K119u) is a common chromatin modification associated with gene silencing, and controlled by the ubiquitin-ligase polycomb repressor complex 1 (PRC1) and H2A-deubiquitinating enzymes (H2A-DUBs). The roles of H2A-DUBs in mammalian development, stem cells, and hematopoiesis have not been addressed. Here we characterized an H2A-DUB targeted mouse line Mysm1(tm1a/tm1a) and demonstrated defects in BM hematopoiesis, resulting in lymphopenia, anemia, and thrombocytosis. Development of lymphocytes was impaired from the earliest stages of their differentiation, and there was also a depletion of erythroid cells and a defect in erythroid progenitor function. These phenotypes resulted from a cell-intrinsic requirement for Mysm1 in the BM. Importantly, Mysm1(tm1a/tm1a) HSCs were functionally impaired, and this was associated with elevated levels of reactive oxygen species, γH2AX DNA damage marker, and p53 protein in the hematopoietic progenitors. Overall, these data establish a role for Mysm1 in the maintenance of BM stem cell function, in the control of oxidative stress and genetic stability in hematopoietic progenitors, and in the development of lymphoid and erythroid lineages.


Subject(s)
Cell Differentiation/genetics , Endopeptidases/genetics , Hematopoiesis/genetics , Lymphocytes/metabolism , Animals , Blood Cell Count , Blotting, Western , Endopeptidases/metabolism , Female , Flow Cytometry , Gene Expression Profiling , Genotype , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Specific Proteases
12.
J Immunol ; 189(1): 102-11, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22664872

ABSTRACT

Sphingosine-1-phosphate (S1P) is lipid messenger involved in the regulation of embryonic development, immune system functions, and many other physiological processes. However, the mechanisms of S1P transport across cellular membranes remain poorly understood, with several ATP-binding cassette family members and the spinster 2 (Spns2) member of the major facilitator superfamily known to mediate S1P transport in cell culture. Spns2 was also shown to control S1P activities in zebrafish in vivo and to play a critical role in zebrafish cardiovascular development. However, the in vivo roles of Spns2 in mammals and its involvement in the different S1P-dependent physiological processes have not been investigated. In this study, we characterized Spns2-null mouse line carrying the Spns2(tm1a(KOMP)Wtsi) allele (Spns2(tm1a)). The Spns2(tm1a/tm1a) animals were viable, indicating a divergence in Spns2 function from its zebrafish ortholog. However, the immunological phenotype of the Spns2(tm1a/tm1a) mice closely mimicked the phenotypes of partial S1P deficiency and impaired S1P-dependent lymphocyte trafficking, with a depletion of lymphocytes in circulation, an increase in mature single-positive T cells in the thymus, and a selective reduction in mature B cells in the spleen and bone marrow. Spns2 activity in the nonhematopoietic cells was critical for normal lymphocyte development and localization. Overall, Spns2(tm1a/tm1a) resulted in impaired humoral immune responses to immunization. This study thus demonstrated a physiological role for Spns2 in mammalian immune system functions but not in cardiovascular development. Other components of the S1P signaling network are investigated as drug targets for immunosuppressive therapy, but the selective action of Spns2 may present an advantage in this regard.


Subject(s)
Anion Transport Proteins/physiology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/pathology , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Anion Transport Proteins/deficiency , Anion Transport Proteins/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Crosses, Genetic , Gene Targeting , Immunophenotyping , Lymphocyte Subsets/metabolism , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/pathology , Lysophospholipids/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Insertional/immunology , Protein Transport/genetics , Protein Transport/immunology , Sphingosine/genetics , Sphingosine/metabolism
13.
Front Immunol ; 15: 1353138, 2024.
Article in English | MEDLINE | ID: mdl-38529289

ABSTRACT

Introduction: BAP1 is a deubiquitinase (DUB) of the Ubiquitin C-terminal Hydrolase (UCH) family that regulates gene expression and other cellular processes, through its direct catalytic activity on the repressive epigenetic mark histone H2AK119ub, as well as on several other substrates. BAP1 is also a highly important tumor suppressor, expressed and functional across many cell types and tissues. In recent work, we demonstrated a cell intrinsic role of BAP1 in the B cell lineage development in murine bone marrow, however the role of BAP1 in the regulation of B cell mediated humoral immune response has not been previously explored. Methods and results: In the current study, we demonstrate that a B-cell intrinsic loss of BAP1 in activated B cells in the Bap1 fl/fl Cγ1-cre murine model results in a severe defect in antibody production, with altered dynamics of germinal centre B cell, memory B cell, and plasma cell numbers. At the cellular and molecular level, BAP1 was dispensable for B cell immunoglobulin class switching but resulted in an impaired proliferation of activated B cells, with genome-wide dysregulation in histone H2AK119ub levels and gene expression. Conclusion and discussion: In summary, our study establishes the B-cell intrinsic role of BAP1 in antibody mediated immune response and indicates its central role in the regulation of the genome-wide landscapes of histone H2AK119ub and downstream transcriptional programs of B cell activation and humoral immunity.


Subject(s)
B-Lymphocytes , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Animals , Mice , Antibodies/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Histones/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
14.
Nature ; 447(7145): 686-90, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17554302

ABSTRACT

Accumulation of DNA damage leading to adult stem cell exhaustion has been proposed to be a principal mechanism of ageing. Here we address this question by taking advantage of the highly specific role of DNA ligase IV in the repair of DNA double-strand breaks by non-homologous end-joining, and by the discovery of a unique mouse strain with a hypomorphic Lig4(Y288C) mutation. The Lig4(Y288C) mouse, identified by means of a mutagenesis screening programme, is a mouse model for human LIG4 syndrome, showing immunodeficiency and growth retardation. Diminished DNA double-strand break repair in the Lig4(Y288C) strain causes a progressive loss of haematopoietic stem cells and bone marrow cellularity during ageing, and severely impairs stem cell function in tissue culture and transplantation. The sensitivity of haematopoietic stem cells to non-homologous end-joining deficiency is therefore a key determinant of their ability to maintain themselves against physiological stress over time and to withstand culture and transplantation.


Subject(s)
Aging/physiology , DNA Repair , Hematopoietic Stem Cells/cytology , Animals , Cell Proliferation , Cellular Senescence/physiology , DNA Breaks, Double-Stranded , DNA Damage , DNA Ligase ATP , DNA Ligases/deficiency , DNA Ligases/genetics , DNA Ligases/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Mutation, Missense/drug effects , Mutation, Missense/genetics , Syndrome
15.
Int J Biochem Cell Biol ; 157: 106384, 2023 04.
Article in English | MEDLINE | ID: mdl-36738766

ABSTRACT

Monoubiquitinated lysine 119 of histone H2A (H2AK119ub) is a highly abundant epigenetic mark, associated with gene repression and deposited on chromatin by the polycomb repressor complex 1 (PRC1), which is an essential regulator of diverse transcriptional programs in mammalian development and tissue homeostasis. While multiple deubiquitinases (DUBs) with catalytic activity for H2AK119ub (H2A-DUBs) have been identified, we lack systematic analyses of their roles and cross-talk in transcriptional regulation. Here, we address H2A-DUB functions in epigenetic regulation of mammalian development and tissue maintenance by conducting a meta-analysis of 248 genomics datasets from 32 independent studies, focusing on the mouse model and covering embryonic stem cells (ESCs), hematopoietic, and immune cell lineages. This covers all the publicly available datasets that map genomic H2A-DUB binding and H2AK119ub distributions (ChIP-Seq), and all datasets assessing dysregulation in gene expression in the relevant H2A-DUB knockout models (RNA-Seq). Many accessory datasets for PRC1-2 and DUB-interacting proteins are also analyzed and interpreted, as well as further data assessing chromatin accessibility (ATAC-Seq) and transcriptional activity (RNA-seq). We report co-localization in the binding of H2A-DUBs BAP1, USP16, and to a lesser extent others that is conserved across different cell-types, and also the enrichment of antagonistic PRC1-2 protein complexes at the same genomic locations. Such conserved sites enriched for the H2A-DUBs and PRC1-2 are proximal to transcriptionally active genes that engage in housekeeping cellular functions. Nevertheless, they exhibit H2AK119ub levels significantly above the genomic average that can undergo further increase with H2A-DUB knockout. This indicates a cooperation between H2A-DUBs and PRC1-2 in the modulation of housekeeping transcriptional programs, conserved across many cell types, likely operating through their antagonistic effects on H2AK119ub and the regulation of local H2AK119ub turnover. Our study further highlights existing knowledge gaps and discusses important directions for future work.


Subject(s)
Drosophila Proteins , Histones , Animals , Mice , Histones/genetics , Histones/metabolism , Epigenesis, Genetic , Chromatin/genetics , Polycomb-Group Proteins/genetics , Hematopoiesis/genetics , Ubiquitination , Drosophila Proteins/genetics , Deubiquitinating Enzymes/genetics , Deubiquitinating Enzymes/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Mammals/genetics , Mammals/metabolism
16.
Sci Rep ; 13(1): 338, 2023 01 07.
Article in English | MEDLINE | ID: mdl-36611064

ABSTRACT

Myb-like SWIRM and MPN domains 1 (MYSM1) is a chromatin binding protein with deubiquitinase (DUB) catalytic activity. Rare MYSM1 mutations in human patients result in an inherited bone marrow failure syndrome, highlighting the biomedical significance of MYSM1 in the hematopoietic system. We and others characterized Mysm1-knockout mice as a model of this disorder and established that MYSM1 regulates hematopoietic function and leukocyte development in such models through different mechanisms. It is, however, unknown whether the DUB catalytic activity of MYSM1 is universally required for its many functions and for the maintenance of hematopoiesis in vivo. To test this, here we generated a new mouse strain carrying a Mysm1D660N point mutation (Mysm1DN) and demonstrated that the mutation renders MYSM1 protein catalytically inactive. We characterized Mysm1DN/DN and Mysm1fl/DN CreERT2 mice, against appropriate controls, for constitutive and inducible loss of MYSM1 catalytic function. We report a profound similarity in the developmental, hematopoietic, and immune phenotypes resulting from the loss of MYSM1 catalytic function and the full loss of MYSM1 protein. Overall, our work for the first time establishes the critical role of MYSM1 DUB catalytic activity in vivo in hematopoiesis, leukocyte development, and other aspects of mammalian physiology.


Subject(s)
Endopeptidases , Ubiquitin-Specific Proteases , Humans , Mice , Animals , Endopeptidases/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Cell Differentiation , Hematopoiesis/genetics , Mutation , Hematopoietic Stem Cells/metabolism , Mice, Knockout , Mammals/metabolism , Trans-Activators/metabolism
17.
J Immunol ; 184(5): 2539-50, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20107187

ABSTRACT

With the rapid rise in the incidence of multidrug resistant infections, there is substantial interest in host defense peptides as templates for production of new antimicrobial therapeutics. Natural peptides are multifunctional mediators of the innate immune response, with some direct antimicrobial activity and diverse immunomodulatory properties. We have previously developed an innate defense regulator (IDR) 1, with protective activity against bacterial infection mediated entirely through its effects on the immunity of the host, as a novel approach to anti-infective therapy. In this study, an immunomodulatory peptide IDR-1002 was selected from a library of bactenecin derivatives based on its substantially more potent ability to induce chemokines in human PBMCs. The enhanced chemokine induction activity of the peptide in vitro correlated with stronger protective activity in vivo in the Staphylococcus aureus-invasive infection model, with a >5-fold reduction in the protective dose in direct comparison with IDR-1. IDR-1002 also afforded protection against the Gram-negative bacterial pathogen Escherichia coli. Chemokine induction by IDR-1002 was found to be mediated through a Gi-coupled receptor and the PI3K, NF-kappaB, and MAPK signaling pathways. The protective activity of the peptide was associated with in vivo augmentation of chemokine production and recruitment of neutrophils and monocytes to the site of infection. These results highlight the importance of the chemokine induction activity of host defense peptides and demonstrate that the optimization of the ex vivo chemokine-induction properties of peptides is a promising method for the rational development of immunomodulatory IDR peptides with enhanced anti-infective activity.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Bacterial Infections/metabolism , Chemokines/metabolism , Leukocytes/metabolism , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/chemical synthesis , Bacterial Infections/microbiology , Bacterial Infections/prevention & control , Cell Line , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CCL7/metabolism , Chemokine CXCL1/metabolism , Female , Humans , Interleukin-8/metabolism , Leukocytes/cytology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Front Immunol ; 13: 1041010, 2022.
Article in English | MEDLINE | ID: mdl-36561751

ABSTRACT

With the major advances in cancer immunology and immunotherapy, it is critical to consider that most immune cells are short-lived and need to be continuously replenished from hematopoietic stem and progenitor cells. Hematologic abnormalities are prevalent in cancer patients, and many ground-breaking studies over the past decade provide insights into their underlying cellular and molecular mechanisms. Such studies demonstrate that the dysfunction of hematopoiesis is more than a side-effect of cancer pathology, but an important systemic feature of cancer disease. Here we review these many advances, covering the cancer-associated phenotypes of hematopoietic stem and progenitor cells, the dysfunction of myelopoiesis and erythropoiesis, the importance of extramedullary hematopoiesis in cancer disease, and the developmental origins of tumor associated macrophages. We address the roles of many secreted mediators, signaling pathways, and transcriptional and epigenetic mechanisms that mediate such hematopoietic dysfunction. Furthermore, we discuss the important contribution of the hematopoietic dysfunction to cancer immunosuppression, the possible avenues for therapeutic intervention, and highlight the unanswered questions and directions for future work. Overall, hematopoietic dysfunction is established as an active component of the cancer disease mechanisms and an important target for therapeutic intervention.


Subject(s)
Hematologic Diseases , Neoplasms , Humans , Hematopoietic Stem Cells/metabolism , Hematopoiesis/genetics , Neoplasms/metabolism , Myelopoiesis , Hematologic Diseases/metabolism , Disease Progression
19.
Pain ; 163(7): e821-e836, 2022 07 01.
Article in English | MEDLINE | ID: mdl-34913882

ABSTRACT

ABSTRACT: The pathophysiology of fibromyalgia syndrome (FMS) remains elusive, leading to a lack of objective diagnostic criteria and targeted treatment. We globally evaluated immune system changes in FMS by conducting multiparametric flow cytometry analyses of peripheral blood mononuclear cells and identified a natural killer (NK) cell decrease in patients with FMS. Circulating NK cells in FMS were exhausted yet activated, evidenced by lower surface expression of CD16, CD96, and CD226 and more CD107a and TIGIT. These NK cells were hyperresponsive, with increased CCL4 production and expression of CD107a when co-cultured with human leukocyte antigen null target cells. Genetic and transcriptomic pathway analyses identified significant enrichment of cell activation pathways in FMS driven by NK cells. Skin biopsies showed increased expression of NK activation ligand, unique long 16-binding protein, on subepidermal nerves of patients FMS and the presence of NK cells near peripheral nerves. Collectively, our results suggest that chronic activation and redistribution of circulating NK cells to the peripheral nerves contribute to the immunopathology associated with FMS.


Subject(s)
Fibromyalgia , Fibromyalgia/metabolism , Flow Cytometry , Humans , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear , Peripheral Nerves
20.
J Immunol ; 183(9): 5788-98, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19812202

ABSTRACT

The human cathelicidin peptide LL-37 is a multifunctional immunomodulatory and antimicrobial host defense peptide of the human immune system. LL-37 modulates host cell responses to microbial stimuli and also affects the action of other endogenous immune mediators such as IL-1beta and GM-CSF. This activity of LL-37 is known to be complex, with the functional outcomes being dependent on the cell type and activation status, timing of exposure, and other immune mediators present. It was demonstrated in this study that LL-37 inhibited cellular responses to IFN-gamma, the key cytokine of Th1-polarized immunity. The inhibitory activity of LL-37 on IFN-gamma responses was characterized in monocytes, macrophages, dendritic cells, and B lymphocytes, showing suppression of cell activation, proliferation, and production of proinflammatory and Th1-polarizing cytokines, and Abs. It was further shown that in monocytes the suppressive effects of LL-37 were mediated through inhibition of STAT1-independent signaling events, involving both the p65 subunit of NF-kappaB and p38 MAPK. This study suggests that LL-37 modulates IFN-gamma responses during both the innate and adaptive phases of immune responses, indicating a new immunomodulatory role for this endogenous peptide. These effects on IFN-gamma activity should be taken into consideration in the development of cathelicidin-based peptides for therapeutic applications as immunomodulatory or microbicidal agents.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antimicrobial Cationic Peptides/physiology , Interferon-gamma/antagonists & inhibitors , Adaptive Immunity , Amino Acid Sequence , Animals , Cathelicidins , Cell Line , Cells, Cultured , Humans , Immunity, Innate , Immunomodulation/immunology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interferon-gamma/physiology , Mice , Molecular Sequence Data , Monocytes/immunology , Monocytes/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/pharmacology , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/physiology , Th1 Cells/immunology , Th1 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL