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










Publication year range
1.
Nat Commun ; 15(1): 4696, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824133

ABSTRACT

Age-related microangiopathy, also known as small vessel disease (SVD), causes damage to the brain, retina, liver, and kidney. Based on the DNA damage theory of aging, we reasoned that genomic instability may underlie an SVD caused by dominant C-terminal variants in TREX1, the most abundant 3'-5' DNA exonuclease in mammals. C-terminal TREX1 variants cause an adult-onset SVD known as retinal vasculopathy with cerebral leukoencephalopathy (RVCL or RVCL-S). In RVCL, an aberrant, C-terminally truncated TREX1 mislocalizes to the nucleus due to deletion of its ER-anchoring domain. Since RVCL pathology mimics that of radiation injury, we reasoned that nuclear TREX1 would cause DNA damage. Here, we show that RVCL-associated TREX1 variants trigger DNA damage in humans, mice, and Drosophila, and that cells expressing RVCL mutant TREX1 are more vulnerable to DNA damage induced by chemotherapy and cytokines that up-regulate TREX1, leading to depletion of TREX1-high cells in RVCL mice. RVCL-associated TREX1 mutants inhibit homology-directed repair (HDR), causing DNA deletions and vulnerablility to PARP inhibitors. In women with RVCL, we observe early-onset breast cancer, similar to patients with BRCA1/2 variants. Our results provide a mechanistic basis linking aberrant TREX1 activity to the DNA damage theory of aging, premature senescence, and microvascular disease.


Subject(s)
DNA Damage , Exodeoxyribonucleases , Phosphoproteins , Animals , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Humans , Phosphoproteins/genetics , Phosphoproteins/metabolism , Mice , Recombinational DNA Repair , Phenotype , Mutation , Drosophila/genetics , Aging/genetics , Aging/metabolism , Female , Drosophila melanogaster/genetics , Male , Retinal Diseases , Vascular Diseases , Hereditary Central Nervous System Demyelinating Diseases
2.
JCI Insight ; 7(17)2022 09 08.
Article in English | MEDLINE | ID: mdl-36073546

ABSTRACT

STING gain-of-function mutations cause STING-associated vasculopathy with onset in infancy (SAVI) in humans, a disease characterized by spontaneous lung inflammation and fibrosis. Mice with STING gain-of-function mutations (SAVI mice) develop αß T cell-dependent lung disease and also lack lymph nodes. Although SAVI has been regarded as a type I interferonopathy, the relative contributions of the three interferon receptors are incompletely understood. Here, we show that STING gain of function led to upregulation of IFN-γ-induced chemokines in the lungs of SAVI mice and that deletion of the type II IFN receptor (IFNGR1), but not the type I IFN receptor (IFNAR1) or type III IFN receptor (IFNλR1), ameliorated lung disease and restored lymph node development in SAVI mice. Furthermore, deletion of IFNGR1, but not IFNAR1 or IFNλR1, corrected the ratio of effector to Tregs in SAVI mice and in mixed bone marrow chimeric mice. Finally, cultured SAVI mouse macrophages were hyperresponsive to IFN-γ, but not IFN-ß, in terms of Cxcl9 upregulation and cell activation. These results demonstrate that IFNGR1 plays a major role in autoinflammation and immune dysregulation mediated by STING gain of function.


Subject(s)
Lung Diseases , Vascular Diseases , Animals , Gain of Function Mutation , Humans , Lung , Membrane Proteins/genetics , Mice , T-Lymphocytes , Vascular Diseases/genetics
3.
Cell Rep ; 35(6): 109113, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33979608

ABSTRACT

STING modulates immunity by responding to bacterial and endogenous cyclic dinucleotides (CDNs). Humans and mice with STING gain-of-function mutations develop a syndrome known as STING-associated vasculopathy with onset in infancy (SAVI), which is characterized by inflammatory or fibrosing lung disease. We hypothesized that hyperresponsiveness of gain-of-function STING to bacterial CDNs might explain autoinflammatory lung disease in SAVI mice. We report that depletion of gut microbes with oral antibiotics (vancomycin, neomycin, and ampicillin [VNA]) nearly eliminates lung disease in SAVI mice, implying that gut microbes might promote STING-associated autoinflammation. However, we show that germ-free SAVI mice still develop severe autoinflammatory disease and that transferring gut microbiota from antibiotics-treated mice to germ-free animals eliminates lung inflammation. Depletion of anaerobes with metronidazole abolishes the protective effect of the VNA antibiotics cocktail, and recolonization with the metronidazole-sensitive anaerobe Bacteroides thetaiotaomicron prevents disease, confirming a protective role of a metronidazole-sensitive microbe in a model of SAVI.


Subject(s)
Gastrointestinal Microbiome/physiology , Lung Diseases/physiopathology , Animals , Humans , Mice , Mutation , Signal Transduction
4.
Cell Rep ; 31(11): 107771, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32553167

ABSTRACT

STING gain-of-function causes autoimmunity and immunodeficiency in mice and STING-associated vasculopathy with onset in infancy (SAVI) in humans. Here, we report that STING gain-of-function in mice prevents development of lymph nodes and Peyer's patches. We show that the absence of secondary lymphoid organs is associated with diminished numbers of innate lymphoid cells (ILCs), including lymphoid tissue inducer (LTi) cells. Although wild-type (WT) α4ß7+ progenitors differentiate efficiently into LTi cells, STING gain-of-function progenitors do not. Furthermore, STING gain-of-function impairs development of all types of ILCs. Patients with STING gain-of-function mutations have fewer ILCs, although they still have lymph nodes. In mice, expression of the STING mutant in RORγT-positive lineages prevents development of lymph nodes and reduces numbers of LTi cells. RORγT lineage-specific expression of STING gain-of-function also causes lung disease. Since RORγT is expressed exclusively in LTi cells during fetal development, our findings suggest that STING gain-of-function prevents lymph node organogenesis by reducing LTi cell numbers in mice.


Subject(s)
Cell Differentiation/immunology , Immunity, Innate/immunology , Lymph Nodes/immunology , Lymphocytes/cytology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Gain of Function Mutation/immunology , Lymphoid Tissue/immunology , Mice , Organogenesis/immunology
5.
J Virol ; 93(19)2019 10 01.
Article in English | MEDLINE | ID: mdl-31315996

ABSTRACT

Autosomal dominant STAT1 mutations in humans have been associated with chronic mucocutaneous candidiasis (CMC), as well as with increased susceptibility to herpesvirus infections. Prior studies have focused on mucosal and Th17-mediated immunity against Candida, but mechanisms of impaired antiviral immunity have not previously been examined. To begin to explore the mechanisms of STAT1-associated immunodeficiency against herpesviruses, we generated heterozygous STAT1 R274W knock-in mice that have a frequently reported STAT1 mutation associated in humans with susceptibility to herpesvirus infections. In primary macrophages and fibroblasts, we found that STAT1 R274W had no appreciable effect on cell-intrinsic immunity against herpes simplex virus 1 (HSV-1) or gammaherpesvirus 68 (γHV68) infection. However, intraperitoneal inoculation of mice with γHV68 was associated with impaired control of infection at day 14 in STAT1 R274W mice compared with that in wild-type (WT) littermate control animals. Infection of STAT1 R274W mice was associated with paradoxically decreased expression of IFN-stimulated genes (ISGs) and gamma interferon (IFN-γ), likely secondary to defective CD4+ and CD8+ T cell responses, including diminished numbers of antigen-specific CD8+ T cells. Viral pathogenesis studies in WT and STAT1 R274W mixed bone marrow chimeric mice revealed that the presence of WT leukocytes was sufficient to limit infection and that antigen-specific STAT1 R274W CD8+ T cell responses were impaired even in the presence of WT leukocytes. Thus, in addition to regulating Th17 responses against Candida, a STAT1 gain-of-function mutant impedes antigen-specific T cell responses against a common gammaherpesvirus in mice.IMPORTANCE Mechanisms of immunodeficiency related to STAT1 gain of function have not been previously studied in an animal model of viral pathogenesis. Using virological and immunological techniques, we examined the immune response to γHV68 in heterozygous mice that have an autosomal dominant mutation in the STAT1 coiled-coil domain (STAT1 R274W). We observed impaired control of infection, which was associated with diminished production of gamma interferon (IFN-γ), fewer effector CD4+ and CD8+ T cells, and a reduction in the number of antigen-specific CD8+ T cells. These findings indicate that a STAT1 gain-of-function mutation limits production of antiviral T cells, likely contributing to immunodeficiency against herpesviruses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gain of Function Mutation , Herpesviridae Infections/immunology , Mutation, Missense , Rhadinovirus/immunology , STAT1 Transcription Factor/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Fibroblasts/immunology , Fibroblasts/virology , Gene Knock-In Techniques , Interferon-gamma/metabolism , Macrophages/immunology , Macrophages/virology , Mice , STAT1 Transcription Factor/genetics
6.
Leukemia ; 33(8): 1978-1995, 2019 08.
Article in English | MEDLINE | ID: mdl-30718771

ABSTRACT

The distinct clinical features of myelofibrosis (MF) have been attributed in part to dysregulated inflammatory cytokine production. Circulating cytokine levels are elevated in MF patients; a subset of which have been shown to be poor prognostic indicators. In this study, cytokine overproduction was examined in MF patient plasma and in MF blood cells ex vivo using mass cytometry. Plasma cytokines measured following treatment with ruxolitinib remained markedly abnormal, indicating that aberrant cytokine production persists despite therapeutic JAK2 inhibition. In MF patient samples, 14/15 cytokines measured by mass cytometry were found to be constitutively overproduced, with the principal cellular source for most cytokines being monocytes, implicating a non-cell-autonomous role for monocyte-derived cytokines impacting disease-propagating stem/progenitor cells in MF. The majority of cytokines elevated in MF exhibited ex vivo hypersensitivity to thrombopoietin (TPO), toll-like receptor (TLR) ligands, and/or tumor necrosis factor (TNF). A subset of this group (including TNF, IL-6, IL-8, IL-10) was minimally sensitive to ruxolitinib. All TPO/TLR/TNF-sensitive cytokines, however, were sensitive to pharmacologic inhibition of NFκB and/or MAP kinase signaling. These results indicate that NFκB and MAP kinase signaling maintain cytokine overproduction in MF, and that inhibition of these pathways may provide optimal control of inflammatory pathophysiology in MF.


Subject(s)
Cytokines/biosynthesis , Janus Kinases/physiology , MAP Kinase Signaling System/physiology , NF-kappa B/physiology , Primary Myelofibrosis/immunology , STAT Transcription Factors/physiology , Signal Transduction/physiology , Humans , MAP Kinase Signaling System/drug effects , Monocytes/immunology , NF-kappa B/antagonists & inhibitors , Nitriles , Primary Myelofibrosis/drug therapy , Pyrazoles/therapeutic use , Pyrimidines , Thrombopoietin/pharmacology , Toll-Like Receptors/physiology
7.
J Allergy Clin Immunol ; 144(1): 254-266.e8, 2019 07.
Article in English | MEDLINE | ID: mdl-30772497

ABSTRACT

BACKGROUND: Monogenic interferonopathies are thought to be mediated by type I interferon. For example, a gain-of-function mutation in stimulator of interferon genes (STING; N153S) upregulates type I interferon-stimulated genes and causes perivascular inflammatory lung disease in mice. The equivalent mutation in human subjects also causes lung disease, which is thought to require signaling through the cyclic GMP-AMP synthase (cGAS)-STING pathway and subsequent activation of interferon regulatory factors (IRFs) 3 and 7, type I interferon, and interferon-stimulated genes. OBJECTIVE: We set out to define the roles of cGAS, IRF3, IRF7, the type I interferon receptor (IFN-α and IFN-ß receptor subunit 1 [IFNAR1]), T cells, and B cells in spontaneous lung disease in STING N153S mice. METHODS: STING N153S mice were crossed to animals lacking cGAS, IRF3/IRF7, IFNAR1, adaptive immunity, αß T cells, and mature B cells. Mice were evaluated for spontaneous lung disease. Additionally, bone marrow chimeric mice were assessed for lung disease severity and survival. RESULTS: Lung disease in STING N153S mice developed independently of cGAS, IRF3/IRF7, and IFNAR1. Bone marrow transplantation revealed that certain features of STING N153S-associated disease are intrinsic to the hematopoietic compartment. Recombination-activating gene 1 (Rag1)-/- STING N153S mice that lack adaptive immunity had no lung disease, and T-cell receptor ß chain (Tcrb)-/- STING N153S animals only had mild disease. STING N153S led to a reduction in percentages and numbers of naive and regulatory T cells, as well as an increased frequency of cytokine-producing effector T cells. CONCLUSION: Spontaneous lung disease in STING N153S mice develops independently of type I interferon signaling and cGAS. STING N153S relies primarily on T cells to promote lung disease in mice.


Subject(s)
Lung Diseases/immunology , Membrane Proteins/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/immunology , Bone Marrow Transplantation , Female , Gain of Function Mutation , Interferon Type I/immunology , Lung/immunology , Male , Membrane Proteins/genetics , Mice, Transgenic , Nucleotidyltransferases/immunology , Spleen/immunology
8.
J Virol ; 93(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30463976

ABSTRACT

We previously generated STING N153S knock-in mice that have a human disease-associated gain-of-function mutation in STING. Patients with this mutation (STING N154S in humans) develop STING-associated vasculopathy with onset in infancy (SAVI), a severe pediatric autoinflammatory disease characterized by pulmonary fibrosis. Since this mutation promotes the upregulation of antiviral type I interferon-stimulated genes (ISGs), we hypothesized that STING N153S knock-in mice may develop more severe autoinflammatory disease in response to a virus challenge. To test this hypothesis, we infected heterozygous STING N153S mice with murine gammaherpesvirus 68 (γHV68). STING N153S mice were highly vulnerable to infection and developed pulmonary fibrosis after infection. In addition to impairing CD8+ T cell responses and humoral immunity, STING N153S also promoted the replication of γHV68 in cultured macrophages. In further support of a combined innate and adaptive immunodeficiency, γHV68 infection was more severe in Rag1-/- STING N153S mice than in Rag1-/- littermate mice, which completely lack adaptive immunity. Thus, a gain-of-function STING mutation creates a combined innate and adaptive immunodeficiency that leads to virus-induced pulmonary fibrosis.IMPORTANCE A variety of human rheumatologic disease-causing mutations have recently been identified. Some of these mutations are found in viral nucleic acid-sensing proteins, but whether viruses can influence the onset or progression of these human diseases is less well understood. One such autoinflammatory disease, called STING-associated vasculopathy with onset in infancy (SAVI), affects children and leads to severe lung disease. We generated mice with a SAVI-associated STING mutation and infected them with γHV68, a common DNA virus that is related to human Epstein-Barr virus. Mice with the human disease-causing STING mutation were more vulnerable to infection than wild-type littermate control animals. Furthermore, the STING mutant mice developed lung fibrosis similar to that of patients with SAVI. These findings reveal that a human STING mutation creates severe immunodeficiency, leading to virus-induced lung disease in mice.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/physiology , Pulmonary Fibrosis/genetics , Adaptive Immunity/genetics , Animals , Gain of Function Mutation/genetics , Gammaherpesvirinae/metabolism , Gammaherpesvirinae/physiology , Immunologic Deficiency Syndromes , Inflammation/genetics , Lung/virology , Macrophages/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mutation , Pulmonary Fibrosis/metabolism , Signal Transduction , T-Lymphocytes/metabolism
9.
Proc Natl Acad Sci U S A ; 115(33): E7768-E7775, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061387

ABSTRACT

The adaptor molecule stimulator of IFN genes (STING) is central to production of type I IFNs in response to infection with DNA viruses and to presence of host DNA in the cytosol. Excessive release of type I IFNs through STING-dependent mechanisms has emerged as a central driver of several interferonopathies, including systemic lupus erythematosus (SLE), Aicardi-Goutières syndrome (AGS), and stimulator of IFN genes-associated vasculopathy with onset in infancy (SAVI). The involvement of STING in these diseases points to an unmet need for the development of agents that inhibit STING signaling. Here, we report that endogenously formed nitro-fatty acids can covalently modify STING by nitro-alkylation. These nitro-alkylations inhibit STING palmitoylation, STING signaling, and subsequently, the release of type I IFN in both human and murine cells. Furthermore, treatment with nitro-fatty acids was sufficient to inhibit production of type I IFN in fibroblasts derived from SAVI patients with a gain-of-function mutation in STING. In conclusion, we have identified nitro-fatty acids as endogenously formed inhibitors of STING signaling and propose for these lipids to be considered in the treatment of STING-dependent inflammatory diseases.


Subject(s)
Fatty Acids/metabolism , Herpes Simplex/metabolism , Herpesvirus 2, Human/metabolism , Membrane Proteins/metabolism , Signal Transduction , Animals , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/metabolism , Autoimmune Diseases of the Nervous System/pathology , Herpes Simplex/genetics , Herpes Simplex/pathology , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Lipoylation , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Nervous System Malformations/pathology , RAW 264.7 Cells
10.
J Exp Med ; 214(11): 3279-3292, 2017 Nov 06.
Article in English | MEDLINE | ID: mdl-28951494

ABSTRACT

Patients with stimulator of interferon genes (STING)-associated vasculopathy with onset in infancy (SAVI) develop systemic inflammation characterized by vasculopathy, interstitial lung disease, ulcerative skin lesions, and premature death. Autosomal dominant mutations in STING are thought to trigger activation of IRF3 and subsequent up-regulation of interferon (IFN)-stimulated genes (ISGs) in patients with SAVI. We generated heterozygous STING N153S knock-in mice as a model of SAVI. These mice spontaneously developed inflammation within the lung, hypercytokinemia, T cell cytopenia, skin ulcerations, and premature death. Cytometry by time-of-flight (CyTOF) analysis revealed that the STING N153S mutation caused myeloid cell expansion, T cell cytopenia, and dysregulation of immune cell signaling. Unexpectedly, we observed only mild up-regulation of ISGs in STING N153S fibroblasts and splenocytes and STING N154S SAVI patient fibroblasts. STING N153S mice lacking IRF3 also developed lung disease, myeloid cell expansion, and T cell cytopenia. Thus, the SAVI-associated STING N153S mutation triggers IRF3-independent immune cell dysregulation and lung disease in mice.


Subject(s)
Inflammation/metabolism , Interferon Regulatory Factor-3/metabolism , Membrane Proteins/metabolism , Vascular Diseases/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Fibroblasts/metabolism , Humans , Inflammation/genetics , Interferon Regulatory Factor-3/genetics , Lung/metabolism , Lung/pathology , Membrane Proteins/genetics , Mice, Knockout , Mice, Transgenic , Mutation , Skin/metabolism , Skin/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Vascular Diseases/genetics
11.
J Immunol ; 198(4): 1417-1422, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28093522

ABSTRACT

Most knowledge on NK cells is based on studies of what are now known as conventional NK cells in the mouse spleen or human peripheral blood. However, recent studies in mice indicate the presence of tissue-resident NK cells in certain organs, such as the liver, that display different markers and transcription factor dependencies as compared with conventional NK cells. In this study, we provide evidence from cytometry by time-of-flight analysis and humanized mice indicating that human CD49e- NK cells are tissue resident in the liver. Thus, these studies indicate that tissue-resident NK cells are evolutionarily conserved in humans and mice, providing a foundation to explore their role in human disease.


Subject(s)
Integrin alpha5/immunology , Killer Cells, Natural/physiology , Liver/cytology , Liver/immunology , Animals , Capillaries/immunology , Flow Cytometry , Humans , Integrin alpha5/genetics , Killer Cells, Natural/immunology , Liver/blood supply , Mice , Mice, Transgenic , Phenotype , Transcription Factors
12.
J Immunol ; 194(4): 1945-53, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25582853

ABSTRACT

Because NK cells secrete cytotoxic granules and cytokines that can destroy surrounding cells and help shape the subsequent immune response, they must be kept under tight control. Several mechanisms, at different levels, are in place to control NK cell function. In this study, we describe a novel mechanism regulating NK cell function in which NK cells acquire ligands for activating receptors from target cells by trogocytosis, rendering the NK cells hyporesponsive. In this model, murine NK cells acquire m157, the murine CMV-encoded ligand for the Ly49H-activating receptor, from target cells both in vitro and in vivo. Although acquisition of m157 requires cell-to-cell contact, it does not require the expression of the Ly49H receptor by the NK cell. Acquired m157 protein is expressed on the NK cell surface with a glycosylphosphatidylinisotol linkage and interacts with the Ly49H receptor expressed on the NK cell. This interaction results in blocking the Ly49H receptor that prevents the NK cells from recognizing m157-expressing targets and continuous engagement of the Ly49H-activating receptor, which results in the hyporesponsiveness of the Ly49H(+) NK cell to stimulation through other activating receptors. Thus, NK cell acquisition of a ligand for an activation receptor by trogocytosis renders them hyporesponsive. This mechanism, by which mature NK cell function can be altered, has important implications in regard to how NK cells respond to tumors in specific microenvironments as well as the use of expanded NK cells in treating various malignancies.


Subject(s)
Antigens, Viral/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Adoptive Transfer , Animals , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/immunology
13.
Stem Cell Reports ; 2(1): 26-35, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24511468

ABSTRACT

We show here that singular loss of the Bright/Arid3A transcription factor leads to reprograming of mouse embryonic fibroblasts (MEFs) and enhancement of standard four-factor (4F) reprogramming. Bright-deficient MEFs bypass senescence and, under standard embryonic stem cell (ESC) culture conditions, spontaneously form clones that in vitro express pluripotency markers, differentiate to all germ lineages, and in vivo form teratomas and chimeric mice. We demonstrate that BRIGHT binds directly to the promoter/enhancer regions of Oct4, Sox2, and Nanog to contribute to their repression in both MEFs and ESCs. Thus, elimination of the BRIGHT barrier may provide an approach for somatic cell reprogramming.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Cellular Reprogramming , Cellular Senescence , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Lewis X Antigen/metabolism , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , SOXB1 Transcription Factors/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptome
14.
Elife ; 2: e00802, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23878724

ABSTRACT

Store-operated calcium entry (SOCE) by calcium release activated calcium (CRAC) channels constitutes a primary route of calcium entry in most cells. Orai1 forms the pore subunit of CRAC channels and Stim1 is the endoplasmic reticulum (ER) resident Ca(2+) sensor. Upon store-depletion, Stim1 translocates to domains of ER adjacent to the plasma membrane where it interacts with and clusters Orai1 hexamers to form the CRAC channel complex. Molecular steps enabling activation of SOCE via CRAC channel clusters remain incompletely defined. Here we identify an essential role of α-SNAP in mediating functional coupling of Stim1 and Orai1 molecules to activate SOCE. This role for α-SNAP is direct and independent of its known activity in NSF dependent SNARE complex disassembly. Importantly, Stim1-Orai1 clustering still occurs in the absence of α-SNAP but its inability to support SOCE reveals that a previously unsuspected molecular re-arrangement within CRAC channel clusters is necessary for SOCE. DOI:http://dx.doi.org/10.7554/eLife.00802.001.


Subject(s)
Calcium/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/physiology , Animals , Drosophila , Humans , Ion Transport , NFATC Transcription Factors/metabolism
15.
Mol Immunol ; 49(1-2): 367-79, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21963220

ABSTRACT

Previous data suggested that constitutive expression of the transcription factor Bright (B cell regulator of immunoglobulin heavy chain transcription), normally tightly regulated during B cell differentiation, was associated with autoantibody production. Here we show that constitutive Bright expression results in skewing of mature B lineage subpopulations toward marginal zone cells at the expense of the follicular subpopulation. C57Bl/6 transgenic mice constitutively expressing Bright in B lineage cells generated autoantibodies that were not the result of global increases in immunoglobulin or of breaches in key tolerance checkpoints typically defective in other autoimmune mouse models. Rather, autoimmunity correlated with increased numbers of marginal zone B cells and alterations in the phenotype and gene expression profiles of lymphocytes within the follicular B cell compartment. These data suggest a novel role for Bright in the normal development of mature B cell subsets and in autoantibody production.


Subject(s)
Autoantibodies/biosynthesis , B-Lymphocytes/immunology , Cell Differentiation/immunology , DNA-Binding Proteins/immunology , Transcription Factors/immunology , Animals , Antibody Formation/immunology , Autoantibodies/immunology , Autoimmunity/immunology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Blotting, Western , Cell Separation , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Transcription Factors/metabolism
16.
Mol Cell Biol ; 31(5): 1041-53, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21199920

ABSTRACT

Bright/Arid3a has been characterized both as an activator of immunoglobulin heavy-chain transcription and as a proto-oncogene. Although Bright expression is highly B lineage stage restricted in adult mice, its expression in the earliest identifiable hematopoietic stem cell (HSC) population suggests that Bright might have additional functions. We showed that >99% of Bright(-/-) embryos die at midgestation from failed hematopoiesis. Bright(-/-) embryonic day 12.5 (E12.5) fetal livers showed an increase in the expression of immature markers. Colony-forming assays indicated that the hematopoietic potential of Bright(-/-) mice is markedly reduced. Rare survivors of lethality, which were not compensated by the closely related paralogue Bright-derived protein (Bdp)/Arid3b, suffered HSC deficits in their bone marrow as well as B lineage-intrinsic developmental and functional deficiencies in their peripheries. These include a reduction in a natural antibody, B-1 responses to phosphocholine, and selective T-dependent impairment of IgG1 class switching. Our results place Bright/Arid3a on a select list of transcriptional regulators required to program both HSC and lineage-specific differentiation.


Subject(s)
B-Lymphocytes/cytology , DNA-Binding Proteins/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Lymphopoiesis/genetics , Transcription Factors/metabolism , Animals , Antibodies/blood , B-Lymphocytes/metabolism , Cell Lineage , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Developmental , Genes, Lethal , Hematopoietic Stem Cells/metabolism , Immunoglobulin Class Switching/genetics , Male , Mice , Mice, Inbred C57BL , Phosphorylcholine/immunology , Phosphorylcholine/metabolism , Transcription Factors/genetics
17.
Stem Cells ; 28(9): 1560-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20680960

ABSTRACT

B-cell regulator of immunoglobulin heavy chain transcription (Bright)/ARID3a, an A+T-rich interaction domain protein, was originally discovered in B lymphocyte lineage cells. However, expression patterns and high lethality levels in knockout mice suggested that it had additional functions. Three independent lines of evidence show that functional inhibition of Bright results in increased developmental plasticity. Bright-deficient cells from two mouse models expressed a number of pluripotency-associated gene products, expanded indefinitely, and spontaneously differentiated into cells of multiple lineages. Furthermore, direct knockdown of human Bright resulted in colonies capable of expressing multiple lineage markers. These data suggest that repression of this single molecule confers adult somatic cells with new developmental options.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Proliferation , DNA-Binding Proteins/deficiency , Pluripotent Stem Cells/metabolism , Transcription Factors/deficiency , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genes, Dominant , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , RNA Interference , Teratoma/genetics , Teratoma/metabolism , Transcription Factors/genetics
18.
J Immunol ; 181(10): 6913-22, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981111

ABSTRACT

The transcription factor Bright up-regulates Ig H chain production from select V region promoters and requires Bright dimerization, Bruton's tyrosine kinase (Btk), and the Btk substrate, TFII-I, for this activity. Defects in Btk cause X-linked immunodeficiency disease in mice and humans. Btk-deficient mice exhibit decreased serum IgM production, B cell developmental blocks, absence of peritoneal B1 cells, and subnormal immune responses against Ags, including phosphorylcholine, which confer protection against Streptococcus pneumoniae. Transgenic mice expressing dominant-negative Bright share similarities with Btk-deficient mice, including decreased serum IgM, poor anti-phosphorylcholine responses, and slightly reduced numbers of mature B cells. Although dominant-negative Bright mice developed B1 B cells, these were functionally deficient in Ig secretion. These data suggest a mechanistic explanation for the abnormal responses to phosphorylcholine observed in Btk-deficient mice, and indicate that Bright functions in a subset of Btk-dependent pathways in vivo, particularly those responses dominated by B1 B cells.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Oncogenes/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Agammaglobulinaemia Tyrosine Kinase , Amino Acid Sequence , Animals , Antibodies/blood , Antigens, CD19/genetics , Blotting, Western , DNA-Binding Proteins , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Lymphocyte Activation/immunology , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Promoter Regions, Genetic , Protein-Tyrosine Kinases , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Transcription Factors
19.
Biol Psychiatry ; 62(10): 1070-9, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17826749

ABSTRACT

BACKGROUND: Early life adverse experience alters adult emotional and cognitive development. Here we assess early life learning about adverse experience and its consequences on adult fear conditioning and amygdala activity. METHODS: Neonatal rats were conditioned daily from 8-12 days-old with paired odor (conditioned stimulus, CS) .5mA shock, unpaired, odor-only, or naive (no infant conditioning). In adulthood, each infant training group was divided into three adult training groups: paired, unpaired or odor-only, using either the same infant CS odor, or a novel adult CS odor without or with the infant CS present as context. Adults were cue tested for freezing (odor in novel environment), with amygdala (14)C 2-DG autoradiography and electrophysiology assessment. RESULTS: Infant paired odor-shock conditioning attenuated adult fear conditioning, but only if the same infant CS odor was used. The (14)C 2-DG activity correlated with infant paired odor-shock conditioning produced attenuated amygdala but heightened olfactory bulb activity. Electrophysiological amygdala assessment further suggests early experience causes changes in amygdala processing as revealed by increased paired-pulse facilitation in adulthood. CONCLUSIONS: This suggests some enduring effects of early life adversity (shock) are under CS control and dependent upon learning for their impact on later adult fear learning.


Subject(s)
Aging , Amygdala/physiology , Conditioning, Classical/physiology , Fear , Memory/physiology , Odorants , Amygdala/diagnostic imaging , Analysis of Variance , Animals , Animals, Newborn , Autoradiography/methods , Avoidance Learning , Behavior, Animal , Deoxyglucose/metabolism , Electric Stimulation/methods , Evoked Potentials/radiation effects , Freezing Reaction, Cataleptic/physiology , Male , Maze Learning , Olfactory Bulb/diagnostic imaging , Radiography , Rats , Rats, Long-Evans , Reaction Time/physiology , Reaction Time/radiation effects
20.
Learn Mem ; 13(6): 801-8, 2006.
Article in English | MEDLINE | ID: mdl-17101877

ABSTRACT

Fetal and infant rats can learn to avoid odors paired with illness before development of brain areas supporting this learning in adults, suggesting an alternate learning circuit. Here we begin to document the transition from the infant to adult neural circuit underlying odor-malaise avoidance learning using LiCl (0.3 M; 1% of body weight, ip) and a 30-min peppermint-odor exposure. Conditioning groups included: Paired odor-LiCl, Paired odor-LiCl-Nursing, LiCl, and odor-saline. Results showed that Paired LiCl-odor conditioning induced a learned odor aversion in postnatal day (PN) 7, 12, and 23 pups. Odor-LiCl Paired Nursing induced a learned odor preference in PN7 and PN12 pups but blocked learning in PN23 pups. 14C 2-deoxyglucose (2-DG) autoradiography indicated enhanced olfactory bulb activity in PN7 and PN12 pups with odor preference and avoidance learning. The odor aversion in weanling aged (PN23) pups resulted in enhanced amygdala activity in Paired odor-LiCl pups, but not if they were nursing. Thus, the neural circuit supporting malaise-induced aversions changes over development, indicating that similar infant and adult-learned behaviors may have distinct neural circuits.


Subject(s)
Association Learning/physiology , Avoidance Learning/physiology , Brain/growth & development , Olfactory Pathways/growth & development , Smell/physiology , Age Factors , Amygdala/growth & development , Amygdala/physiology , Animals , Brain/physiology , Conditioning, Classical/physiology , Female , Male , Olfactory Bulb/growth & development , Olfactory Bulb/physiology , Olfactory Pathways/physiology , Rats , Rats, Long-Evans
SELECTION OF CITATIONS
SEARCH DETAIL
...