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










Publication year range
1.
J Exp Med ; 220(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-36820829

ABSTRACT

In innate immune cells, intracellular sensors such as cGAS-STING stimulate type I/III interferon (IFN) expression, which promotes antiviral defense and immune activation. However, how IFN-I/III expression is controlled in adaptive cells is poorly understood. Here, we identify a transcriptional rheostat orchestrated by RELA that confers human T cells with innate-like abilities to produce IFN-I/III. Despite intact cGAS-STING signaling, IFN-I/III responses are stunted in CD4+ T cells compared with dendritic cells or macrophages. We find that lysine residues in RELA tune the IFN-I/III response at baseline and in response to STING stimulation in CD4+ T cells. This response requires positive feedback driven by cGAS and IRF7 expression. By combining RELA with IRF3 and DNA demethylation, IFN-I/III production in CD4+ T cells reaches levels observed in dendritic cells. IFN-I/III production provides self-protection of CD4+ T cells against HIV infection and enhances the elimination of tumor cells by CAR T cells. Therefore, innate-like functions can be tuned and leveraged in human T cells.


Subject(s)
HIV Infections , Interferon Type I , Humans , Immunity, Innate/genetics , Nucleotidyltransferases/metabolism , Interferon Type I/metabolism , T-Lymphocytes/metabolism , Transcription Factor RelA
2.
J Allergy Clin Immunol Pract ; 9(2): 803-818.e11, 2021 02.
Article in English | MEDLINE | ID: mdl-33217613

ABSTRACT

BACKGROUND: Gain-of-function mutations in STING1 underlie a type I interferonopathy termed SAVI (STING-associated vasculopathy with onset in infancy). This severe disease is variably characterized by early-onset systemic inflammation, skin vasculopathy, and interstitial lung disease (ILD). OBJECTIVE: To describe a cohort of patients with SAVI. METHODS: Assessment of clinical, radiological and immunological data from 21 patients (17 families) was carried out. RESULTS: Patients carried heterozygous substitutions in STING1 previously described in SAVI, mainly the p.V155M. Most were symptomatic from infancy, but late onset in adulthood occurred in 1 patient. Systemic inflammation, skin vasculopathy, and ILD were observed in 19, 18, and 21 patients, respectively. Extensive tissue loss occurred in 4 patients. Severity of ILD was highly variable with insidious progression up to end-stage respiratory failure reached at teenage in 6 patients. Lung imaging revealed early fibrotic lesions. Failure to thrive was almost constant, with severe growth failure seen in 4 patients. Seven patients presented polyarthritis, and the phenotype in 1 infant mimicked a combined immunodeficiency. Extended features reminiscent of other interferonopathies were also found, including intracranial calcification, glaucoma and glomerular nephropathy. Increased expression of interferon-stimulated genes and interferon α protein was constant. Autoantibodies were frequently found, in particular rheumatoid factor. Most patients presented with a T-cell defect, with low counts of memory CD8+ cells and impaired T-cell proliferation in response to antigens. Long-term follow-up described in 8 children confirmed the clinical benefit of ruxolitinib in SAVI where the treatment was started early in the disease course, underlying the need for early diagnosis. Tolerance was reasonably good. CONCLUSION: The largest worldwide cohort of SAVI patients yet described, illustrates the core features of the disease and extends the clinical and immunological phenotype to include overlap with other monogenic interferonopathies.


Subject(s)
Lung Diseases, Interstitial , Membrane Proteins/genetics , Vascular Diseases , Adolescent , Adult , Child , Humans , Infant , Inflammation , Mutation
3.
J Allergy Clin Immunol ; 143(2): 712-725.e5, 2019 02.
Article in English | MEDLINE | ID: mdl-29800647

ABSTRACT

BACKGROUND: Autosomal dominant gain-of-function mutations in human stimulator of interferon genes (STING) lead to a severe autoinflammatory disease called STING-associated vasculopathy with onset in infancy that is associated with enhanced expression of interferon-stimulated gene transcripts. OBJECTIVE: The goal of this study was to analyze the phenotype of a new mouse model of STING hyperactivation and the role of type I interferons in this system. METHODS: We generated a knock-in model carrying an amino acid substitution (V154M) in mouse STING, corresponding to a recurrent mutation seen in human patients with STING-associated vasculopathy with onset in infancy. Hematopoietic development and tissue histology were analyzed. Lymphocyte activation and proliferation were assessed in vitro. STING V154M/wild-type (WT) mice were crossed to IFN-α/ß receptor (IFNAR) knockout mice to evaluate the type I interferon dependence of the mutant Sting phenotype recorded. RESULTS: In STING V154M/WT mice we detected variable expression of inflammatory infiltrates in the lungs and kidneys. These mice showed a marked decrease in survival and developed a severe combined immunodeficiency disease (SCID) affecting B, T, and natural killer cells, with an almost complete lack of antibodies and a significant expansion of monocytes and granulocytes. The blockade in B- and T-cell development was present from early immature stages in bone marrow and thymus. In addition, in vitro experiments revealed an intrinsic proliferative defect of mature T cells. Although the V154M/WT mutant demonstrated increased expression of interferon-stimulated genes, the SCID phenotype was not reversed in STING V154M/WT IFNAR knockout mice. However, the antiproliferative defect in T cells was rescued partially by IFNAR deficiency. CONCLUSIONS: STING gain-of-function mice developed an interferon-independent SCID phenotype with a T-cell, B-cell, and natural killer cell developmental defect and hypogammaglobulinemia that is associated with signs of inflammation in lungs and kidneys. Only the intrinsic proliferative defect of T cells was partially interferon dependent.


Subject(s)
B-Lymphocytes/physiology , Inflammation/genetics , Killer Cells, Natural/immunology , Membrane Proteins/genetics , Mutation/genetics , Severe Combined Immunodeficiency/genetics , T-Lymphocytes/physiology , Agammaglobulinemia , Animals , Cell Differentiation/genetics , Disease Models, Animal , Humans , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/genetics
4.
J Exp Med ; 214(6): 1769-1785, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28484079

ABSTRACT

Activation of the cyclic dinucleotide sensor stimulator of interferon (IFN) genes (STING) is critical for IFN and inflammatory gene expression during innate immune responses. However, the role of STING in adaptive immunity is still unknown. In this study, we show that STING activation reduces the proliferation of T lymphocytes. This activity was independent of TBK1 and IRF3 recruitment and of type I IFN but required a distinct C-terminal domain of STING that activates NF-κB. Inhibition of cell proliferation by STING required its relocalization to the Golgi apparatus and caused mitotic errors. T lymphocytes from patients carrying constitutive active mutations in TMEM173 encoding STING showed impaired proliferation and reduced numbers of memory cells. Endogenous STING inhibited proliferation of mouse T lymphocytes. Therefore, STING, a critical innate sensor, also functions intrinsically in cells of the adaptive immune system to inhibit proliferation.


Subject(s)
Immunity, Innate , Membrane Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Binding Sites , Cell Proliferation , Dendritic Cells/metabolism , Humans , Interferon Regulatory Factor-3/metabolism , Membrane Proteins/chemistry , Mitosis , Mutant Proteins/metabolism , Protein Domains , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
5.
J Allergy Clin Immunol ; 140(2): 543-552.e5, 2017 08.
Article in English | MEDLINE | ID: mdl-28087229

ABSTRACT

BACKGROUND: Gain-of-function mutations in transmembrane protein 173 (TMEM173) encoding stimulator of interferon genes (STING) underlie a recently described type I interferonopathy called STING-associated vasculopathy with onset in infancy (SAVI). OBJECTIVES: We sought to define the molecular and cellular pathology relating to 3 individuals variably exhibiting the core features of the SAVI phenotype including systemic inflammation, destructive skin lesions, and interstitial lung disease. METHODS: Genetic analysis, conformational studies, in vitro assays and ex vivo flow-cytometry were performed. RESULTS: Molecular and in vitro data demonstrate that the pathology in these patients is due to amino acid substitutions at positions 206, 281, and 284 of the human STING protein. These mutations confer cGAMP-independent constitutive activation of type I interferon signaling through TBK1 (TANK-binding kinase), independent from the alternative STING pathway triggered by membrane fusion of enveloped RNA viruses. This constitutive activation was abrogated by ex vivo treatment with the janus kinase 1/2 inhibitor ruxolitinib. CONCLUSIONS: Structural analysis indicates that the 3 disease-associated mutations at positions 206, 281, and 284 of the STING protein define a novel cluster of amino acids with functional importance in the regulation of type I interferon signaling.


Subject(s)
Inflammation/genetics , Interferon Type I/genetics , Membrane Proteins/genetics , Adolescent , Adult , Amino Acid Substitution , Child , Female , HEK293 Cells , Humans , Interferon Type I/metabolism , Male , Mutation , STAT1 Transcription Factor/metabolism , Signal Transduction
6.
Chest ; 150(3): e65-71, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27613991

ABSTRACT

We report three cases of pulmonary disease suggesting fibrosis in two familial and one sporadic case. Pulmonary symptoms were associated with various clinical features of systemic inflammation and vasculitis involving the skin, and appeared at different ages. A strong interferon signature was found in all three cases. Disease was not responsive to corticosteroids, and lung transplantation was considered for all three subjects at an early age. One of them underwent double-lung transplantation, but she immediately experienced a primary graft dysfunction and died soon after. Recognized causes of familial interstitial lung disease were all excluded. All three subjects had a mutation in the previously described autoinflammatory disease called SAVI (stimulator of interferon genes [STING]-associated vasculopathy with onset in infancy). These cases emphasize the need to consider this possibility in children and young adults with lung fibrosis after common causes have been ruled out.


Subject(s)
Autoimmune Diseases/genetics , Inflammation/genetics , Membrane Proteins/genetics , Mutation , Pulmonary Fibrosis/genetics , Adult , Autoimmune Diseases/diagnostic imaging , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Chilblains/immunology , Child , Fatal Outcome , Female , Humans , Infant , Inflammation/diagnostic imaging , Inflammation/immunology , Inflammation/pathology , Male , Pulmonary Fibrosis/diagnostic imaging , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Severity of Illness Index , Tomography, X-Ray Computed
8.
Blood ; 126(2): 133-43, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-25947942

ABSTRACT

Arthrogryposis, renal dysfunction, and cholestasis (ARC) syndrome is caused by deficiencies in the trafficking proteins VPS33B or VIPAR, and is associated with a bleeding diathesis and a marked reduction in platelet α-granules. We generated a tamoxifen-inducible mouse model of VPS33B deficiency, Vps33b(fl/fl)-ER(T2), and studied the platelet phenotype and α-granule biogenesis. Ultrastructural analysis of Vps33b(fl/fl)-ER(T2) platelets identified a marked reduction in α-granule count and the presence of small granule-like structures in agreement with the platelet phenotype observed in ARC patients. A reduction of ∼65% to 75% was observed in the α-granule proteins von Willebrand factor and P-selectin. Although platelet aggregation responses were not affected, a defect in δ-granule secretion was observed. Under arteriolar shear conditions, Vps33b(fl/fl)-ER(T2) platelets were unable to form stable aggregates, and tail-bleeding measurement revealed a bleeding diathesis. Analysis of bone marrow-derived megakaryocytes (MKs) by conventional and immuno-electron microscopy from Vps33b(fl/fl)-ER(T2) mice revealed a reduction in mature type-II multivesicular bodies (MVB II) and an accumulation of large vacuoles. Proteins that are normally stored in α-granules were underrepresented in MVB II and proplatelet extensions. These results demonstrate that abnormal protein trafficking and impairment in MVB maturation in MKs underlie the α-granule deficiency in Vps33b(fl/fl)-ER(T2) mouse and ARC patients.


Subject(s)
Cytoplasmic Granules/metabolism , Megakaryocytes/metabolism , Vesicular Transport Proteins/physiology , Animals , Arthrogryposis/genetics , Cells, Cultured , Cholestasis/genetics , Gray Platelet Syndrome/genetics , Gray Platelet Syndrome/metabolism , Humans , Megakaryocytes/cytology , Megakaryocytes/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organelles/metabolism , Platelet Count , Polymorphism, Single Nucleotide , Protein Transport/genetics , Renal Insufficiency/genetics , Vesicular Transport Proteins/genetics
9.
JAMA Dermatol ; 151(8): 872-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25992765

ABSTRACT

IMPORTANCE: The type I interferonopathies comprise a recently recognized group of mendelian diseases characterized by an upregulation of type I interferon signaling. These monogenic phenotypes include classic Aicardi-Goutières syndrome and syndromic forms of systemic lupus erythematosus, including familial chilblain lupus and spondyloenchondrodysplasia. Dermatologic features provide a major diagnostic clue to this disease grouping, as exemplified by the recently described stimulator of interferon genes-associated vasculopathy with onset in infancy (SAVI) caused by gain-of-function mutations in TMEM173. OBSERVATIONS: We describe a male child who, from the age of 2 months, had significant cutaneous disease that manifested as red violaceous plaques of the cheeks, nose, ears, fingers, and toes that progressed to gangrenous necrosis. In addition to his severe cutaneous vasculopathy, he experienced recurrent fevers, interstitial lung disease, and failure to thrive. His clinical syndrome was refractory to multiple immunosuppressive therapies. Evidence of marked upregulation of type I interferon signaling was observed in peripheral blood, and genetic testing identified a de novo germline mutation in TMEM173, confirming a diagnosis of SAVI 7 years after the onset of his disease. CONCLUSIONS AND RELEVANCE: This observational report describes a new case of SAVI, a recently defined monogenic inflammatory phenotype, that exemplifies an emerging group of disorders related to primary upregulation of type I interferon signaling.


Subject(s)
Granulomatosis with Polyangiitis/diagnosis , Interferon Type I/genetics , Membrane Proteins/genetics , Skin Diseases, Vascular/diagnosis , Humans , Infant , Male , Mutation , Phenotype , Skin Diseases, Vascular/genetics , Skin Diseases, Vascular/pathology , Up-Regulation
10.
J Clin Invest ; 124(12): 5516-20, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25401470

ABSTRACT

Innate immunity to viral infection involves induction of the type I IFN response; however, dysfunctional regulation of this pathway leads to inappropriate inflammation. Here, we evaluated a nonconsanguineous family of mixed European descent, with 4 members affected by systemic inflammatory and autoimmune conditions, including lupus, with variable clinical expression. We identified a germline dominant gain-of-function mutation in TMEM173, which encodes stimulator of type I IFN gene (STING), in the affected individuals. STING is a key signaling molecule in cytosolic DNA-sensing pathways, and STING activation normally requires dimerization, which is induced by 2'3' cyclic GMP-AMP (cGAMP) produced by the cGAMP synthase in response to cytosolic DNA. Structural modeling supported constitutive activation of the mutant STING protein based on stabilized dimerization. In agreement with the model predictions, we found that the STING mutant spontaneously localizes in the Golgi of patient fibroblasts and is constitutively active in the absence of exogenous 2'3'-cGAMP in vitro. Accordingly, we observed elevated serum IFN activity and a type I IFN signature in peripheral blood from affected family members. These findings highlight the key role of STING in activating both the innate and adaptive immune responses and implicate aberrant STING activation in features of human lupus.


Subject(s)
Genetic Diseases, Inborn/immunology , Lupus Erythematosus, Systemic/immunology , Mutation , Protein Multimerization/immunology , Signal Transduction/immunology , Adaptive Immunity/genetics , Adult , Aged , Child, Preschool , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Humans , Immunity, Innate/genetics , Interferon Type I/genetics , Interferon Type I/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Male , Membrane Proteins , Nucleotides, Cyclic/genetics , Nucleotides, Cyclic/immunology , Protein Multimerization/genetics , Signal Transduction/genetics , Syndrome
12.
Semin Arthritis Rheum ; 43(2): 217-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23786871

ABSTRACT

OBJECTIVE: RASopathies (Noonan syndrome (NS) and Noonan-related syndromes) are neurodevelopmental syndromes resulting from germline mutations in genes that participate in the rat sarcoma/mitogen-activated protein kinases (RAS/MAPK) pathway (PTPN11, SOS1, RAF, KRAS or NRAS, and SHOC2). Some monogenic conditions are associated with the development of systemic lupus erythematosus (SLE), and a few reports described the association of SLE with NS. We aim to search for a relationship between RASopathy and the development of SLE. METHODS: We reported for the first time a case of 13-year-old boy with NS with loose anagen hair (NSLAH) resulting from mutation in SHOC2 who developed an autoimmune disorder that fulfilled four American College of Rheumatology (ACR) criteria for the classification of SLE (polyarthritis, pericarditis, antinuclear antibodies, and anti-DNA antibodies). The case report then prompted a literature review by a systematic search for English and French articles on the subjects of RASopathies and SLE that had English abstracts in PubMed from 1966 to 2012. RESULTS: We identified seven additional patients with RASopathy and SLE. The male-to-female ratio was 1:1 and age at onset of SLE ranged from 5 to 32 years. The most common features were polyarthritis (7/8 patients), autoimmune cytopenia (4/8 patients), and pericarditis (4/8 patients) while only one patient presented with skin involvement. CONCLUSION: The association of two rare diseases in eight patients suggests that RASopathies may be associated with the development of SLE, which is characterized by a higher male-to-female ratio, a lower rate of skin involvement, and a higher rate of pericarditis than "classic" SLE.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Lupus Erythematosus, Systemic/genetics , Mutation , Noonan Syndrome/genetics , Adolescent , Humans , Lupus Erythematosus, Systemic/etiology , Male , Noonan Syndrome/complications
13.
Methods Mol Biol ; 707: 105-18, 2011.
Article in English | MEDLINE | ID: mdl-21287332

ABSTRACT

The discovery of Foxp3 as a reliable marker for murine regulatory T cells has led to an explosion in the development of genetic tools for investigating the biology of regulatory T cells. More than 25 Foxp3-based mouse strains have been published with a variety of characteristics. The effects of Foxp3 expression can be analyzed using null, hypomorphic, conditional, altered control, and over-expression strains. Reporter strains are available to efficiently isolate Foxp3+ cells, with various reporter designs in terms of construct (fusion, replacement, and bicistronic positioning), and reporter system (GFP, YFP, RFP, Luciferase, Thy1.1). Multifunction strain fusion, replacement, and bicistronic positionings add functional proteins under the control of the Foxp3 promoter allowing induced apoptosis or lineage-specific Cre recombinase activity. In this chapter, we discuss the uses of the cornucopia of genetic tools, in isolation and in combination, for research on Foxp3(+) regulatory T cells.


Subject(s)
Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...