Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 559
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 121(39): e2316161121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39298490

ABSTRACT

Uveitis is a vision-threatening disease primarily driven by a dysregulated immune response, with retinal microglia playing a pivotal role in its progression. Although the transcription factor EGR2 is known to be closely associated with uveitis, including Vogt-Koyanagi-Harada disease and Behcet's disease, and is essential for maintaining the dynamic homeostasis of autoimmunity, its exact role in uveitis remains unclear. In this study, diminished EGR2 expression was observed in both retinal microglia from experimental autoimmune uveitis (EAU) mice and inflammation-induced human microglia cell line (HMC3). We constructed a mice model with conditional knockout of EGR2 in microglia and found that EGR2 deficiency resulted in increased intraocular inflammation. Meanwhile, EGR2 overexpression downregulated the expression of inflammatory cytokines as well as cell migration and proliferation in HMC3 cells. Next, RNA sequencing and ChIP-PCR results indicated that EGR2 directly bound to its downstream target growth differentiation factor 15 (GDF15) and further regulated GDF15 transcription. Furthermore, intravitreal injection of GDF15 recombinant protein was shown to ameliorate EAU progression in vivo. Meanwhile, knockdown of GDF15 reversed the phenotype of EGR2 overexpression-induced microglial inflammation in vitro. In summary, this study highlighted the protective role of the transcription factor EGR2 in AU by modulating the microglial phenotype. GFD15 was identified as a downstream target of EGR2, providing a unique target for uveitis treatment.


Subject(s)
Autoimmune Diseases , Early Growth Response Protein 2 , Growth Differentiation Factor 15 , Microglia , Uveitis , Animals , Humans , Mice , Autoimmune Diseases/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/metabolism , Cell Line , Disease Models, Animal , Early Growth Response Protein 2/metabolism , Early Growth Response Protein 2/genetics , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Phenotype , Retina/metabolism , Retina/pathology , Uveitis/immunology , Uveitis/metabolism , Uveitis/pathology , Uveitis/genetics
2.
Mol Ther ; 32(6): 1970-1983, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38627968

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) modulate the immune response through interactions with innate immune cells. We previously demonstrated that MSCs alleviate ocular autoimmune inflammation by directing bone marrow cell differentiation from pro-inflammatory CD11bhiLy6ChiLy6Glo cells into immunosuppressive CD11bmidLy6CmidLy6Glo cells. Herein, we analyzed MSC-induced CD11bmidLy6Cmid cells using single-cell RNA sequencing and compared them with CD11bhiLy6Chi cells. Our investigation revealed seven distinct immune cell types including myeloid-derived suppressor cells (MDSCs) in the CD11bmidLy6Cmid cells, while CD11bhiLy6Chi cells included mostly monocytes/macrophages with a small cluster of neutrophils. These MSC-induced MDSCs highly expressed Retnlg, Cxcl3, Cxcl2, Mmp8, Cd14, and Csf1r as well as Arg1. Comparative analyses of CSF-1RhiCD11bmidLy6Cmid and CSF-1RloCD11bmidLy6Cmid cells demonstrated that the former had a homogeneous monocyte morphology and produced elevated levels of interleukin-10. Functionally, these CSF-1RhiCD11bmidLy6Cmid cells, compared with the CSF-1RloCD11bmidLy6Cmid cells, inhibited CD4+ T cell proliferation and promoted CD4+CD25+Foxp3+ Treg expansion in culture and in a mouse model of experimental autoimmune uveoretinitis. Resistin-like molecule (RELM)-γ encoded by Retnlg, one of the highly upregulated genes in MSC-induced MDSCs, had no direct effects on T cell proliferation, Treg expansion, or splenocyte activation. Together, our study revealed a distinct transcriptional profile of MSC-induced MDSCs and identified CSF-1R as a key cell-surface marker for detection and therapeutic enrichment of MDSCs.


Subject(s)
Mesenchymal Stem Cells , Myeloid-Derived Suppressor Cells , Single-Cell Analysis , Animals , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/immunology , Single-Cell Analysis/methods , Transcriptome , Cell Differentiation/genetics , Gene Expression Profiling , Disease Models, Animal , Uveitis/genetics , Uveitis/immunology , Uveitis/metabolism , Humans
3.
J Neuroinflammation ; 21(1): 112, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684986

ABSTRACT

BACKGROUND: Dimethyl fumarate (DMF) is a fumaric acid ester that exhibits immunoregulatory and anti-inflammatory properties. However, the function of DMF in autoimmune uveitis (AU) is incompletely understood, and studies comprehensively exploring the impact of DMF on immune cells are still lacking. METHODS: To explore the function of DMF in uveitis and its underlying mechanisms, we conducted single-cell RNA sequencing (scRNA-seq) on the cervical draining lymph node (CDLN) cells of normal, experimental autoimmune uveitis (EAU), and DMF-treated EAU mice. Additionally, we integrated scRNA-seq data of the retina and CDLNs to identify the potential impact of DMF on ocular immune cell infiltration. Flow cytometry was conducted to verify the potential target molecules of DMF. RESULTS: Our study showed that DMF treatment effectively ameliorated EAU symptoms. The proportional and transcriptional alterations in each immune cell type during EAU were reversed by DMF treatment. Bioinformatics analysis in our study indicated that the enhanced expression of Pim1 and Cxcr4 in EAU was reversed by DMF treatment. Further experiments demonstrated that DMF restored the balance between effector T (Teff) /regulatory T (Treg) cells through inhibiting the pathway of PIM1-protein kinase B (AKT)-Forkhead box O1 (FOXO1). By incorporating the scRNA-seq data of the retina from EAU mice into analysis, our study identified that T cells highly expressing Pim1 and Cxcr4 were enriched in the retina. DMF repressed the ocular infiltration of Teff cells, and this effect might depend on its inhibition of PIM1 and CXCR4 expression. Additionally, our study indicated that DMF might reduce the proportion of plasma cells by inhibiting PIM1 expression in B cells. CONCLUSIONS: DMF effectively attenuated EAU symptoms. During EAU, DMF reversed the Teff/Treg cell imbalance and suppressed the ocular infiltration of Teff cells by inhibiting PIM1 and CXCR4 expression. Thus, DMF may act as a new drug option for the treatment of AU.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Autoimmune Diseases , Dimethyl Fumarate , Immunosuppressive Agents , Retina , Uveitis , Dimethyl Fumarate/administration & dosage , Dimethyl Fumarate/pharmacology , Uveitis/genetics , Uveitis/immunology , Uveitis/therapy , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Single-Cell Gene Expression Analysis , Disease Models, Animal , Animals , Mice , Female , Mice, Inbred C57BL , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , Transcription, Genetic , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Atlases as Topic , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Retina/drug effects , Retina/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology
4.
Expert Rev Mol Med ; 26: e9, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38618935

ABSTRACT

Glaucoma and uveitis are non-vascular ocular diseases which are among the leading causes of blindness and visual loss. These conditions have distinct characteristics and mechanisms but share a multifactorial and complex nature, making their management challenging and burdensome for patients and clinicians. Furthermore, the lack of symptoms in the early stages of glaucoma and the diverse aetiology of uveitis hinder timely and accurate diagnoses, which are a cause of poor visual outcomes under both conditions. Although current treatment is effective in most cases, it is often associated with low patient adherence and adverse events, which directly impact the overall therapeutic success. Therefore, long-lasting alternatives with improved safety and efficacy are needed. Gene therapy, particularly utilising adeno-associated virus (AAV) vectors, has emerged as a promising approach to address unmet needs in these diseases. Engineered capsids with enhanced tropism and lower immunogenicity have been proposed, along with constructs designed for targeted and controlled expression. Additionally, several pathways implicated in the pathogenesis of these conditions have been targeted with single or multigene expression cassettes, gene editing and silencing approaches. This review discusses strategies employed in AAV-based gene therapies for glaucoma and non-infectious uveitis and provides an overview of current progress and future directions.


Subject(s)
Glaucoma , Uveitis , Humans , Glaucoma/genetics , Glaucoma/therapy , Uveitis/genetics , Uveitis/therapy , Eye , Blindness , Genetic Therapy
5.
FASEB J ; 37(11): e23277, 2023 11.
Article in English | MEDLINE | ID: mdl-37878342

ABSTRACT

Pathogenic Th17 cells are critical drivers of multiple autoimmune diseases, including uveitis and its animal model, experimental autoimmune uveitis (EAU). However, how innate immune signals modulate pathogenic Th17 responses remains largely unknown. Here, we showed that miR-338-3p endowed dendritic cells (DCs) with an increased ability to activate interphotoreceptor retinoid-binding protein (IRBP)1-20 -specific Th17 cells by promoting the production of IL-6, IL-1ß, and IL-23. In vivo administration of LV-miR-338-infected DCs promoted pathogenic Th17 responses and exacerbated EAU development. Mechanistically, dual-specificity phosphatase 16 (Dusp16) was a molecular target of miR-338-3p. miR-338-3p repressed Dusp16 and therefore strengthened the mitogen-activated protein kinase (MAPK) p38 signaling, resulting in increased production of Th17-polarizing cytokines and subsequent pathogenic Th17 responses. In addition, methyltransferase like 3 (Mettl3), a key m6A methyltransferase, mediated the upregulation of miR-338-3p in activated DCs. Together, our findings identify a vital role for Mettl3/miR-338-3p/Dusp16/p38 signaling in DCs-driven pathogenic Th17 responses and suggest a potential therapeutic avenue for uveitis and other Th17 cell-related autoimmune disorders.


Subject(s)
Autoimmune Diseases , MicroRNAs , Uveitis , Animals , Th17 Cells , Autoimmune Diseases/genetics , Methyltransferases , p38 Mitogen-Activated Protein Kinases/genetics , Uveitis/genetics , Dendritic Cells , MicroRNAs/genetics
6.
FASEB J ; 37(3): e22803, 2023 03.
Article in English | MEDLINE | ID: mdl-36753389

ABSTRACT

Methyltransferase like 3 (METTL3), a primary N6-methyladenosine (m6A) methyltransferase, has been implicated in various biological and pathological processes including immune responses. However, the functions and mechanisms of METTL3 in pathogenic T helper (Th)17 cells are poorly understood. Here we found significantly decreased METTL3 expression along with reduced m6A levels in eyeballs and T cells of experimental autoimmune uveitis (EAU). Overexpression of METTL3 ameliorated the development of EAU and suppressed pathogenic Th17 cell responses in vivo and in vitro. Mechanistically, METTL3 promoted the expression of absent, small, or homeotic-like 1 (ASH1L) via enhancing its stability in a YT521-B homology domain containing 2 (YTHDC2)-dependent manner, which further decreased the expression of IL-17 and IL-23 receptor (IL-23R), resulting in reduced pathogenic Th17 responses. Together, our data reveal a pivotal role of METTL3 in regulating pathogenic Th17 responses, which may contribute to human uveitis therapy.


Subject(s)
DNA-Binding Proteins , Histone-Lysine N-Methyltransferase , Methyltransferases , Th17 Cells , Uveitis , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , RNA, Messenger/genetics , Uveitis/genetics , Uveitis/metabolism , Animals , Autoimmune Diseases , Disease Models, Animal
7.
Eur J Pediatr ; 183(1): 1-7, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37735224

ABSTRACT

Blau syndrome is a rare genetic granulomatosis affecting children. It could be responsible for vision-threatening complications and articular deformation. Due to the rarity of this disease, there are no standardized guidelines for its management. This work aimed to provide an updated overview of the different therapeutic options for Blau syndrome. We conducted research in the PubMed database for the different treatments used in Blau syndrome patients, and we proposed a therapeutic algorithm for disease management. High doses of corticosteroids are considered as a bridging therapy in Blau syndrome. Methotrexate should be initiated if the patient has articular or ocular involvement. An anti-tumor necrosis factor α should be added for patients with uveitis or residual arthritis. If the patient remains symptomatic, a switch to another anti-tumor necrosis factor α is the best option. In non-responders to the first- and second-line biotherapies, a switch to an anti-interleukin 1, an anti-interleukin 6, or tofacitinib is necessary. CONCLUSION: This article suggested an algorithm for the treatment of Blau syndrome. Other studies are necessary to confirm the efficacy of these treatments. WHAT IS KNOWN: • Blau syndrome is a rare but severe granulomatosis that could be responsible for vision-threatening complications and articular deformation. • Blau syndrome seems to be refractory to treatments. WHAT IS NEW: • High doses of corticosteroids are usually insufficient and should be considered only as a bridging therapy. • Blau syndrome could be considered as a poor factor for uveitis, thus, an anti-tumor necrosis factor α should be initiated for patients with uveitis or with residual arthritis.


Subject(s)
Arthritis , Sarcoidosis , Synovitis , Uveitis , Child , Humans , Arthritis/drug therapy , Arthritis/genetics , Uveitis/diagnosis , Uveitis/drug therapy , Uveitis/genetics , Adrenal Cortex Hormones/therapeutic use , Necrosis/complications
8.
Am J Dermatopathol ; 46(6): 381-382, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38648024

ABSTRACT

ABSTRACT: Blau syndrome is a rare familial autoinflammatory disorder characterized by the triad of granulomatous dermatitis, polyarthritis, and uveitis. Blau syndrome exhibits an autosomal dominant inheritance pattern and can be caused by a gain-of-function mutation in nucleotide-binding oligomerization domain 2 (NOD2), a member of the NOD-like receptor family of pattern recognition receptors. Mutations in NOD2 cause upregulation of inflammatory cytokines and resultant autoinflammation. Because of the rarity of this condition and early onset of symptoms, Blau syndrome may be misdiagnosed as juvenile idiopathic arthritis. We present a case of a 37-year-old male patient with a long-documented history of juvenile idiopathic arthritis and uveitis, who developed an asymptomatic eruption of pink papules on the trunk and upper extremities. A biopsy demonstrated noncaseating, well-formed dermal granulomas with relatively sparse lymphocytic inflammation and Langerhans-type giant cells. Genetic testing confirmed a mutation in NOD2. Based on the patient's clinical history, histologic findings, genetic testing, the diagnosis of Blau syndrome was made.


Subject(s)
Arthritis , Nod2 Signaling Adaptor Protein , Sarcoidosis , Synovitis , Uveitis , Humans , Male , Uveitis/genetics , Uveitis/diagnosis , Arthritis/genetics , Arthritis/diagnosis , Synovitis/genetics , Synovitis/pathology , Synovitis/diagnosis , Adult , Nod2 Signaling Adaptor Protein/genetics , Sarcoidosis/genetics , Sarcoidosis/diagnosis , Sarcoidosis/pathology , Dermatitis/genetics , Dermatitis/pathology , Dermatitis/diagnosis , Biopsy , Hereditary Autoinflammatory Diseases
9.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125957

ABSTRACT

Blau syndrome (BS) is a rare autoinflammatory granulomatosis characterized by granulomatous arthritis, uveitis, and dermatitis. Ocular complications are particularly severe in BS, significantly contributing to morbidity. This study aims to identify potential biomarkers for BS ocular degeneration through proteomic profiling of tear samples from affected patients. Seven subjects from the same family, including four carriers of the BS-associated NOD2 mutation (p.E383K), were recruited alongside healthy controls. Tear samples were collected using Schirmer strips and analyzed via mass spectrometry. A total of 387 proteins were identified, with significant differences in protein expression between BS patients, healthy familial subjects, and healthy controls. Key findings include the overexpression of alpha-2-macroglobulin (A2M) and immunoglobulin heavy constant gamma 4 (IGHG4) in BS patients. Bioinformatic analysis revealed that differentially expressed proteins are involved in acute-phase response, extracellular exosome formation, and protein binding. Notably, neutrophils' azurophilic granule components, as azurocidin (AZU1), myeloperoxidases (MPO), and defensins (DEFA3), were highly expressed in the most severely affected subject, suggesting a potential role of neutrophils in BS ocular severity. These proteins might be promising biomarkers for ocular involvement in BS, facilitating early detection and tailored treatment strategies.


Subject(s)
Arthritis , Biomarkers , Proteomics , Sarcoidosis , Synovitis , Tears , Uveitis , Humans , Tears/metabolism , Biomarkers/metabolism , Uveitis/metabolism , Uveitis/genetics , Uveitis/diagnosis , Female , Male , Arthritis/genetics , Arthritis/metabolism , Synovitis/metabolism , Synovitis/genetics , Sarcoidosis/genetics , Sarcoidosis/metabolism , Adult , Proteomics/methods , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Middle Aged , Mutation , Proteome/metabolism , Hereditary Autoinflammatory Diseases
10.
Clin Immunol ; 255: 109743, 2023 10.
Article in English | MEDLINE | ID: mdl-37604356

ABSTRACT

OBJECTIVE: Blau syndrome (BS), considered a rare pediatric autoinflammatory disease, is characterised by a triad of granulomatous arthritis, dermatitis and uveitis. Here we present a tale of three families visited in our outpatient department in the last two years (2020-2022) where more than one member was affected with either skin, ophthalmological and joint involvement with either biopsy-proven granuloma or genetic mutation at NOD2 gene suggesting the diagnosis of BS. CASE SERIES: The first family had three affected members where the mother and her two children had skin changes, polyarthritis and a pathogenic mutation in NOD2 gene (exon 4, c.1000C > T, p.Arg334Trp) suggesting BS. The second family had two affected members where both mother and her son had uveitis, skin changes with NOD2 mutation at exon 4 with c.1147G > A (p Glu 383 Lys) variant. The son also had polyarthritis and his skin biopsy was suggestive of granulomatous inflammation. In the third family with two affected members, we found a mutation in NOD2 on exon 4 (c 1324C > T, p.Lys 442 Phe) which was described as pathogenic with only one report published till date. CONCLUSION: These three cases presented to us within the last two years and led to a diagnosis of BS in three other family members with discrete mutations (commonest to rarest) on the NOD2 gene in the three families.


Subject(s)
Arthritis , Sarcoidosis , Uveitis , Child , Female , Humans , Arthritis/genetics , India , Mothers , Mutation , Nod2 Signaling Adaptor Protein/genetics , Sarcoidosis/genetics , Uveitis/genetics , Uveitis/diagnosis , Male
11.
Clin Immunol ; 246: 109210, 2023 01.
Article in English | MEDLINE | ID: mdl-36528252

ABSTRACT

Uveitis with complex pathogenesis is a kind of eye emergency involving refractory and blinding inflammation. Dysregulation of TANK binding kinase 1 (TBK1), which plays an important role in innate immunity, often leads to inflammatory diseases in various organs. However, the role of TBK1 in uveitis remains elusive. In this study, we identified that the mRNA expression level of TBK1 and its phosphorylation level were significantly increased in peripheral blood mononuclear cells (PBMCs) of patients with uveitis. Consistent with this, the expression of Tbk1 was elevated in the ocular tissues of uveitis rats and primary peritoneal macrophages while its phosphorylation levels, which present activation forms, were upregulated as well, accompanied by an increase in the level of nuclear factor-κB (NF-κB) and proinflammatory cytokines. In addition, inhibition of TBK1 may effectively reduce the inflammatory response of uveitis rats by blocking NF-κB entry into the nucleus and impeding the initiation of NLRP3 inflammasome- and caspase-1-mediated pyroptosis pathways.


Subject(s)
NF-kappa B , Uveitis , Animals , Rats , Inflammasomes/metabolism , Inflammation/metabolism , Leukocytes, Mononuclear/metabolism , NF-kappa B/metabolism , Signal Transduction , Uveitis/genetics
12.
Clin Exp Rheumatol ; 41(10): 2105-2114, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37812477

ABSTRACT

OBJECTIVES: Ophthalmologic involvement in monogenic autoinflammatory diseases has been explored mainly in paediatric patients. The aim of this study is to characterise ophthalmologic manifestations, therapeutic management and visual outcomes in a Spanish (UVESAI) cohort of adult/paediatric patients with monogenic autoinflammatory diseases. METHODS: Multicentre and retrospective study of patients with monogenic autoinflammatory diseases and ocular involvement. Eye manifestations, structural complications, treatments used and visual outcomes were analysed, and compared with previous studies. RESULTS: Forty-six patients (44/2 adults/children; 21/25 adult/paediatric-onset) with monogenic autoinflammatory diseases [cryopyrin associated periodic syndromes (n=13/28.3%), mainly Muckle-Wells syndrome (MWS) (n=11/24%); familial Mediterranean fever (FMF) (n=12/26%); TNF receptor-associated periodic syndrome (TRAPS); (n=9/20%); Blau syndrome (n=8/17%); hyperimmunoglobulin D syndrome (HIDS) (n=2/4.3%), deficiency of adenosine deaminase-2 and NLRC4-Autoinflammatory disease] (one each) were included. Conjunctivitis (n=26/56.5%) and uveitis (n=23/50%) were the most frequent ocular manifestations. Twelve (26.1%) patients developed structural complications, being cataracts (n=11/24%) and posterior synechiae (n=10/22%) the most frequent. Conjunctivitis predominated in TRAPS, FMF, MWS and HIDS (mainly in adults), and uveitis, in Blau syndrome. Seven (8%) eyes (all with uveitis) presented with impaired visual acuity. Local and systemic treatment led to good visual outcomes in most patients. Compared with previous studies mainly including paediatric patients, less severe ocular involvement was observed in our adult/paediatric cohort. CONCLUSIONS: Conjunctivitis was the most common ocular manifestation in our TRAPS, FMF, MWS and HIDS patients, and uveitis predominated in Blau syndrome. Severe eye complications and poor visual prognosis were associated with uveitis. Adults with monogenic autoinflammatory diseases seem to exhibit a less severe ophthalmologic presentation than paediatric patients.


Subject(s)
Conjunctivitis , Cryopyrin-Associated Periodic Syndromes , Familial Mediterranean Fever , Hereditary Autoinflammatory Diseases , Uveitis , Humans , Child , Adult , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/genetics , Retrospective Studies , Adenosine Deaminase , Intercellular Signaling Peptides and Proteins , Uveitis/etiology , Uveitis/genetics , Familial Mediterranean Fever/complications , Familial Mediterranean Fever/diagnosis , Familial Mediterranean Fever/genetics , Cryopyrin-Associated Periodic Syndromes/drug therapy , Conjunctivitis/genetics
13.
J Allergy Clin Immunol ; 149(1): 176-188.e7, 2022 01.
Article in English | MEDLINE | ID: mdl-34175136

ABSTRACT

BACKGROUND: Blau syndrome (BS) is an autoinflammatory disease associated with mutations in nucleotide-binding oligomerization domain 2. Although treatments with anti-TNF agents have been reported to be effective, the underlying molecular mechanisms remain unclear. OBJECTIVE: We aimed to elucidate the mechanisms of autoinflammation in patients with BS and to clarify how anti-TNF treatment controls the disease phenotype at the cellular level in clinical samples. METHODS: Macrophages were differentiated from monocytes of 7 BS patients, and global transcriptional profiles of 5 patients were analyzed with or without IFN-γ stimulation. Macrophages were also generated from BS-specific induced pluripotent stem cells (iPSCs), and their transcriptome was examined for comparison. RESULTS: Aberrant inflammatory responses were observed upon IFN-γ stimulation in macrophages from untreated BS patients, but not in those from patients treated with anti-TNF. iPSC-derived macrophages carrying a disease-associated mutation also showed IFN-γ-dependent accelerated inflammatory responses. Comparisons of peripheral blood- and iPSC-derived macrophages revealed the upregulation of nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) targets in unstimulated macrophages as a common feature. CONCLUSIONS: IFN-γ stimulation is one of the key signals driving aberrant inflammatory responses in BS-associated macrophages. However, long-term treatment with anti-TNF agents ameliorates such abnormalities even in the presence of IFN-γ stimulation. Our data thus suggest that preexposure to TNF or functionally similar cytokines inducing NF-κB-driven proinflammatory signaling during macrophage development is a prerequisite for accelerated inflammatory responses upon IFN-γ stimulation in BS.


Subject(s)
Arthritis/immunology , Interferon-gamma/immunology , Macrophages/immunology , Sarcoidosis/immunology , Synovitis/immunology , Tumor Necrosis Factor Inhibitors/pharmacology , Uveitis/immunology , Adult , Arthritis/drug therapy , Arthritis/genetics , Cell Line , Child , Child, Preschool , Female , Humans , Induced Pluripotent Stem Cells/cytology , Male , NF-kappa B/immunology , Sarcoidosis/drug therapy , Sarcoidosis/genetics , Synovitis/drug therapy , Synovitis/genetics , Transcriptome , Tumor Necrosis Factor Inhibitors/therapeutic use , Uveitis/drug therapy , Uveitis/genetics , Young Adult
14.
Int Ophthalmol ; 43(2): 589-596, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35947254

ABSTRACT

PURPOSE: The objective of this article was to examine the potential effect of juvenile idiopathic arthritis-associated uveitis (JIAU) on the risk of major depressive and anxiety disorders through Mendelian randomization (MR) study. METHODS: Genetic instrumental variables from the largest available genome-wide association study for JIAU, major depressive disorder, and anxiety disorder were applied. A set of complementary MR approaches including inverse-variance weighted (IVW) were carried out to verify the estimate association and assess horizontal pleiotropy. RESULTS: Our results indicated that genetically driven JIAU did not causally produce changes in major depressive or anxiety disorders (IVW: OR = 1.001, 95% CI = 0.997-1.006, P = 0.581; IVW: OR = 1.006, 95% CI = 0.980-1.033, P = 0.649, respectively). In addition, the risk of JIAU could not be influenced by genetically predicted major depressive or anxiety disorders (IVW: OR = 1.132, 95% CI = 0.914-1.404, P = 0.256; IVW: OR = 1.019, 95% CI = 0.548-1.896, P = 0.953, respectively). Besides, several sensitivity analyses indicated that our MR results were robust and no horizontal pleiotropy was observed (P > 0.05). CONCLUSIONS: Our MR study does not reveal sufficient evidence to support the causal association of JIAU with the development of major depressive or anxiety disorders in both directions. Further large studies are warranted to validate the undetermined relationship between JIAU and the risk of major depressive or anxiety disorders.


Subject(s)
Arthritis, Juvenile , Depressive Disorder, Major , Uveitis , Humans , Arthritis, Juvenile/complications , Arthritis, Juvenile/genetics , Genome-Wide Association Study , Mendelian Randomization Analysis , Depression , Polymorphism, Single Nucleotide , Uveitis/complications , Uveitis/genetics , Anxiety , Anxiety Disorders/complications , Anxiety Disorders/epidemiology
15.
Genes Immun ; 23(2): 57-65, 2022 04.
Article in English | MEDLINE | ID: mdl-35379982

ABSTRACT

Uveitis is the most common form of intraocular inflammatory disease and is a significant cause of visual impairment worldwide. Aetiologically, uveitis can also be classified into infectious uveitis and non-infectious uveitis. The common non-infectious forms of uveitis include acute anterior uveitis (AAU), Behçet's disease (BD), Vogt-Koyanagi-Harada (VKH) disease, birdshot chorioretinopathy (BSCR), sarcoid uveitis. In addition, a few monogenic autoinflammatory disorders can also cause uveitis, such as Blau Syndrome and haploinsufficiency of A20 (HA20). Although the exact pathogenesis of non-infectious uveitis is still unclear, it is well-recognised that it involves both genetic and environmental risk factors. A hallmark of uveitis is its strong associations with human leucocyte antigens (HLA). For examples, AAU, BD and BSCR are strongly associated with HLA-B27, HLA-B51, and HLA-A29, respectively. In uveitis studies, multiple GWAS have successfully been conducted and led to identification of novel susceptibility loci, for example, IL23R has been identified in BD, VKH and AAU. In this review, we summarize the latest progress on the genetic associations of both HLA and non-HLA genes with major forms of uveitis, including AAU, BD, VKH, BSCR, sarcoid uveitis, Blau Syndrome and HA20, and potential future research directions.


Subject(s)
Arthritis , Behcet Syndrome , Sarcoidosis , Synovitis , Uveitis, Anterior , Uveitis , Behcet Syndrome/genetics , HLA Antigens/genetics , HLA-B27 Antigen , Humans , Sarcoidosis/genetics , Uveitis/genetics , Uveitis, Anterior/genetics
16.
Clin Immunol ; 241: 109080, 2022 08.
Article in English | MEDLINE | ID: mdl-35878734

ABSTRACT

OBJECTIVE: Uveitis is an intraocular inflammatory disease. Epigenetics has been associated with its pathogenesis. However, the role of N6-methyladenosine (m6A) in uveitis has not been reported. We aimed to examine the role of m6A and its regulatory mechanism in experimental autoimmune uveitis (EAU). METHODS: The mRNA expression of m6A-related methylase and demethylase of retinal pigment epithelium (RPE) between mice with EAU and control mice was detected by RT-qPCR. The overall m6A level of ARPE-19 cells was detected by an m6A quantitative detection kit. Cell proliferation was observed by CCK-8 assays, and ELISA was used to test the secretion of inflammatory factors. The expression of tight junction proteins and the target genes of FTO were examined by western blotting and MeRIP-PCR. RESULTS: A decreased expression of FTO in RPE cells was found in mice with EAU. Increased overall m6A%, proliferation of cells and secretion of IL-6, IL-8 and MCP-1 were found after FTO knockdown in ARPE-19 cells. However, ZO-1 and occludin protein expression was decreased. ATF4 protein expression was decreased in the FTO knockdown (shFTO) group as compared with the control (shNC) group. In contrast, the m6A level of ATF4 was elevated, as shown by MeRIP-PCR. Functional analysis showed that p-STAT3 expression was increased in the shFTO group, and the change in occludin expression was reversed in ATF4 rescue experiment. CONCLUSION: FTO may affect the translation of ATF4 by regulating its m6A level, resulting in the increased expression of p-STAT3 and inflammatory factors, and leading to uveitis.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Retinal Pigment Epithelium , Uveitis , Adenosine/analogs & derivatives , Adenosine/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Cytokines/metabolism , Mice , Occludin/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Tight Junctions/metabolism , Uveitis/genetics
17.
Ophthalmology ; 129(7): 821-828, 2022 07.
Article in English | MEDLINE | ID: mdl-35314268

ABSTRACT

PURPOSE: The American College of Medical Genetics and Genomics (ACMG) and the Association for Molecular Pathology call for cautious interpretation of variants as causative of a monogenic disorder by stringent standards. We aimed to reclassify the pathogenicity of nucleotide binding oligomerization domain containing 2 (NOD2) variants according to the ACMG guidelines and to characterize clinical features in patients whose ocular disease might actually be explained by Blau syndrome. DESIGN: Genetic analysis and descriptive study. PARTICIPANTS: A total of 1003 unrelated healthy individuals and 3921 sporadic patients who presented with uveitis. METHODS: Whole-exome sequencing was performed on all healthy participants and 551 patients with uveitis, and targeted NOD2 resequencing was performed on the remaining 3370 patients with uveitis. Pathogenicity for Blau syndrome was classified for NOD2 variants identified by sequencing in study participants according to the ACMG guidelines. Clinical manifestations were compared among NOD2 variants of different levels of classification. MAIN OUTCOME MEASURES: Pathogenicity of variants. RESULTS: Eight NOD2 gain-of-function mutations, p.R334W, p.R334Q, p.E383K, p.G481D, p.W490S, p.M513T, p.R587C, and p.N670K, were classified as pathogenic, and 66 patients (1.7%) with uveitis were diagnosed with Blau syndrome due to these mutations. Of 66 with Blau syndrome, anterior uveitis accounted for 39.4%, posterior uveitis for 9.1%, and panuveitis for 51.5%. A proportion of 21.2% of Blau syndrome presented as multifocal choroiditis, 48.5% had papillitis, and 74.2% showed retinal microvasculitis detected by fundus fluorescein angiography. Six NOD2 variants, p.P268S, p.R311W, p.R471C, p.A612T, p.R702W, and p.V955I, were considered nonpathogenic for Blau syndrome and were identified in 96 patients with uveitis. The incidence of bilateral uveitis (86.4%), secondary glaucoma (47.0%), epiretinal membrane (7.6%), choroidal neovascularization (4.6%), retinal atrophy (10.6%), arthritis (69.7%), joint deformity (51.5%), and skin rash (40.9%) was higher in Blau syndrome than in patients with uveitis carrying non-Blau-causing NOD2 variants. Patients with Blau syndrome permanently experienced overall poorer best-corrected visual acuity. Several rare NOD2 mutations, p.I722L (2 cases), p.T476P (1 case), p.T476del (1 case), and p.R439H (1 case), were newly identified. CONCLUSIONS: Pathogenic NOD2 variants for Blau syndrome were limited to those gain-of-function mutations and were associated with a high risk for arthritis, skin rash, permanent visual loss, and ocular complications in patients with uveitis.


Subject(s)
Arthritis , Exanthema , Sarcoidosis , Uveitis , Arthritis/diagnosis , Arthritis/genetics , China , Exanthema/complications , Humans , Mutation , Nod2 Signaling Adaptor Protein/genetics , Sarcoidosis/complications , Sarcoidosis/diagnosis , Sarcoidosis/genetics , Synovitis , Uveitis/complications , Uveitis/diagnosis , Uveitis/genetics
18.
J Immunol ; 204(8): 2053-2063, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32169850

ABSTRACT

Autoimmune diseases are a physiological state that immune responses are directed against and damage the body's own tissues. Numerous studies have demonstrated promising therapeutic effects in certain autoimmune diseases by targeting IL-23/IL-17 axis, mostly through using Abs against IL-23 or IL-17A. Pyrrole-imidazole polyamides are nuclease-resistant compounds that inhibit gene expression through binding to the minor groove of DNA. To develop a novel gene-silencing agent that targets IL-23/IL-17 axis, we designed polyamide that specifically binds to the transcription factor c-Rel-binding site located in the promoter of IL-23p19 subunit. Our study showed that this polyamide is capable of entering into nucleus with high efficiency in dendritic cells and macrophage. In addition, it prevented the binding of c-Rel to the promoter of IL-23p19 in vivo and specifically inhibited the expression of IL-23. More importantly, we demonstrated that this polyamide is therapeutically effective using both the imiquimod-induced psoriasis and experimental autoimmune uveitis mouse models. Taken together, these results indicate that pyrrole-imidazole polyamide targeting IL-23p19 could be a novel and feasible therapeutic strategy for patients with autoimmune diseases.


Subject(s)
Autoimmune Diseases/drug therapy , Autoimmune Diseases/genetics , Gene Silencing , Interleukin-23 Subunit p19/antagonists & inhibitors , Nylons/pharmacology , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Female , Imidazoles/pharmacology , Imiquimod , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Structure , Psoriasis/chemically induced , Psoriasis/drug therapy , Psoriasis/genetics , Psoriasis/immunology , Pyrroles/pharmacology , Uveitis/chemically induced , Uveitis/drug therapy , Uveitis/genetics , Uveitis/immunology
19.
Anim Genet ; 53(3): 436-440, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35451153

ABSTRACT

Equine recurrent uveitis (ERU) is an autoimmune disease defined by inflammation of the uveal tract of the eye. The cause of ERU is thought to be complex, involving both genetic and environmental factors. The purpose of this study was to investigate potential genetic risk factors for ERU in the Icelandic horse. Fifty-six Icelandic horses (11 affected with ERU and 45 controls) living in Denmark and the USA, eight years or older, were included in the study. A case-control GWAS was performed using the GGP Equine 80K array on the Illumina Infinium HD Beadchip using 40 horses. A mixed linear model analysis identified a single SNP on ECA 11 (BIEC2_141650; NC_009154.3:g.3817009A>G) that reached genome-wide significance (p = 1.79 × 10-7 ). This variant was within an intron of tissue inhibitor of metalloproteinase 2 (TIMP2), a gene previously implicated in ERU. Sanger sequencing identified a single coding variant in this gene; however it was a synonymous mutation (NC_009154.3:g.3858193C>T) and was not perfectly concordant with ERU phenotype (p = 0.68). Further investigation of TIMP2 is warranted. Additional horses and markers are needed to identify other potential loci worthy of further investigation as contributors to ERU risk in Icelandic horses.


Subject(s)
Horse Diseases , Uveitis , Animals , Case-Control Studies , Horse Diseases/genetics , Horses/genetics , Iceland , Tissue Inhibitor of Metalloproteinase-2 , Uveitis/genetics , Uveitis/veterinary
20.
Anim Genet ; 53(6): 872-877, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36210489

ABSTRACT

Equine recurrent uveitis (ERU) is a blinding ocular disorder among horses, and the Appaloosa horse breed is disproportionally affected by a chronic form of this intraocular inflammatory disease known as insidious uveitis. Strong breed predisposition and previous investigations suggest that there is a genetic component to the pathology of insidious uveitis among Appaloosa horses; however, no estimates of the heritability of the disease have previously been determined. This study aimed to characterize the genetic underpinning of the disease by estimating the heritability for insidious uveitis among Appaloosas. After combining two genotyping array datasets from the Illumina Equine SNP70 BeadChip and the Axiom Equine 670 K Genotyping Array, heritability was estimated for 59 affected and 83 unaffected horses using both restricted maximum likelihood (REML) and phenotype correlation - genotype correlation solvers from the linkage disequilibrium adjusted kinship software. Based on previous research, age and sex were used as covariates, and the locus responsible for the characteristic Appaloosa coat pattern (LP), previously associated with ERU risk, was included as a fixed effect ('top predictor'). Using prevalence values from 0.05 to 0.42, the heritability estimate for insidious uveitis ranged from 0.95 (SE = 0.14) to 1.74 (SE = 0.25) with LP contributing 0.16-0.33 to the estimate. This study suggests that insidious uveitis is highly heritable (REML 95% CI, h2  = 0.68-1.0) and additional loci outside of LP are contributing to the genetic risk for insidious uveitis for Appaloosas. Once identified, these other genetic factors may lead to new disease mitigation efforts in veterinary care and breeding practices.


Subject(s)
Horse Diseases , Uveitis , Horses/genetics , Animals , Horse Diseases/genetics , Horse Diseases/epidemiology , Uveitis/genetics , Uveitis/veterinary , Genotype , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL