Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
Int J Mol Sci ; 25(15)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39125957

RESUMEN

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.


Asunto(s)
Artritis , Biomarcadores , Proteómica , Sarcoidosis , Sinovitis , Lágrimas , Uveítis , Humanos , Lágrimas/metabolismo , Biomarcadores/metabolismo , Uveítis/metabolismo , Uveítis/genética , Uveítis/diagnóstico , Femenino , Masculino , Artritis/genética , Artritis/metabolismo , Sinovitis/metabolismo , Sinovitis/genética , Sarcoidosis/genética , Sarcoidosis/metabolismo , Adulto , Proteómica/métodos , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , Persona de Mediana Edad , Mutación , Proteoma/metabolismo , Enfermedades Autoinflamatorias Hereditarias
2.
Arthritis Rheumatol ; 76(9): 1439-1445, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38782587

RESUMEN

OBJECTIVE: The objective is to characterize transcriptomic profiles and immune cell composition and distribution in juvenile idiopathic arthritis (JIA) synovial biopsies, assess for associations of these features with clinical parameters, and compare JIA and rheumatoid arthritis (RA) synovial features. METHODS: RNA sequencing (RNASeq) was performed on 24 samples, with pathway analysis and inference of relative abundance of immune cell subsets based on gene expression data. Two multiplex fluorescence immunohistochemistry (IHC) panels were performed on 28 samples (including 13 on which RNASeq was performed), staining for CD206- classical and CD206+ nonclassical macrophages, and CD8+ and CD4+ T and B lymphocytes. Data were compared to a published series of early RA synovial biopsies. RESULTS: Pathway analysis of the most variably expressed genes (n = 339) identified a B and plasma cell signature as the main driver of heterogeneity in JIA synovia, with strong overlap between JIA and RA synovitis. Multiplex IHC confirmed heterogeneity of immune cell infiltration. M1-like macrophage-rich synovial lining was associated with greater lining hypertrophy and higher (CD45+) pan-immune cell and CD8+ T cell infiltration. CONCLUSION: Our study indicates significant similarities between JIA and RA synovitis. Similar to RA, JIA synovia may be broadly categorized into two groups: (1) those with an inflammatory/adaptive immune transcriptomic signature, M1-like macrophage and CD8+ T cell infiltration, and thicker, M1-like macrophage-rich synovial lining, and (2) those with an M2-like macrophage transcriptomic signature, greater M2/M1-like macrophage ratios, and thinner, M2-like macrophage-rich synovial lining. Synovial features were not significantly associated with clinical parameters, likely because of group size and heterogeneity.


Asunto(s)
Artritis Juvenil , Artritis Reumatoide , Linfocitos B , Macrófagos , Membrana Sinovial , Humanos , Artritis Juvenil/patología , Artritis Juvenil/inmunología , Membrana Sinovial/patología , Membrana Sinovial/inmunología , Artritis Reumatoide/patología , Artritis Reumatoide/inmunología , Artritis Reumatoide/genética , Macrófagos/patología , Macrófagos/inmunología , Biopsia , Masculino , Femenino , Niño , Linfocitos B/patología , Linfocitos B/inmunología , Transcriptoma , Adolescente , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/inmunología , Sinovitis/patología , Sinovitis/inmunología , Sinovitis/genética , Células Plasmáticas/patología , Células Plasmáticas/inmunología , Inmunohistoquímica , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/inmunología
3.
Am J Dermatopathol ; 46(6): 381-382, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38648024

RESUMEN

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.


Asunto(s)
Artritis , Proteína Adaptadora de Señalización NOD2 , Sarcoidosis , Sinovitis , Uveítis , Humanos , Masculino , Uveítis/genética , Uveítis/diagnóstico , Artritis/genética , Artritis/diagnóstico , Sinovitis/genética , Sinovitis/patología , Sinovitis/diagnóstico , Adulto , Proteína Adaptadora de Señalización NOD2/genética , Sarcoidosis/genética , Sarcoidosis/diagnóstico , Sarcoidosis/patología , Dermatitis/genética , Dermatitis/patología , Dermatitis/diagnóstico , Biopsia , Enfermedades Autoinflamatorias Hereditarias
4.
Ocul Immunol Inflamm ; 32(8): 1728-1732, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38180755

RESUMEN

PURPOSE: To document the long-term visual outcomes in patients with Blau syndrome. METHODS: A retrospective institutional cohort study was conducted, and 13 patients with genetically confirmed Blau syndrome were included. Demographic and clinical data were collected from standardised medical charts. Baseline was defined as the first detected uveitis and data were recorded onwards at intervals of 1, 3, 5, 10, 15 and 20 years. RESULTS: Anterior uveitis was the most common classification at baseline (57.1%). Among patients with documented uveitis lasting 10 years or more, all of them developed panuveitis. Median logMAR visual acuity at baseline was 0 (range -0.5; 0.7), 0.19 (range 0; 1.5) at year 5, and 0.7 (range 0.1 - no perception of light) at year 20, as recorded in 13, 16, and 10 eyes, respectively. All patients received treatment with topical and oral steroids, and multiple systemic immunosuppressants including biologics. Disease control, defined as having cells <1+ in both eyes and using topical steroid eye drops less than twice daily, was achieved in 14.3% to 37.5% of patients at the different time points. Cataract surgery was performed in 12 eyes of 8 patients, 3 eyes of 3 patients necessitated glaucoma surgery, and 4 eyes of 4 patients required surgery for retinal detachment. CONCLUSION: Uveitis associated with Blau syndrome commonly leads to severe, chronic panuveitis, requiring long-term systemic immunosuppression. Early diagnosis and timely initiation of biologics may prevent significant visual impairment.


Asunto(s)
Artritis , Sarcoidosis , Sinovitis , Uveítis , Agudeza Visual , Humanos , Estudios Retrospectivos , Masculino , Femenino , Agudeza Visual/fisiología , Sinovitis/diagnóstico , Sinovitis/genética , Sinovitis/tratamiento farmacológico , Sinovitis/fisiopatología , Artritis/diagnóstico , Artritis/fisiopatología , Artritis/genética , Sarcoidosis/diagnóstico , Sarcoidosis/complicaciones , Sarcoidosis/fisiopatología , Adulto , Uveítis/diagnóstico , Uveítis/fisiopatología , Uveítis/tratamiento farmacológico , Uveítis/complicaciones , Adolescente , Estudios de Seguimiento , Adulto Joven , Niño , Persona de Mediana Edad , Linfadenitis/diagnóstico , Inmunosupresores/uso terapéutico , Proteína Adaptadora de Señalización NOD2/genética , Glucocorticoides/uso terapéutico , Enfermedades de los Nervios Craneales/diagnóstico , Enfermedades de los Nervios Craneales/genética , Enfermedades de los Nervios Craneales/fisiopatología , Enfermedades Autoinflamatorias Hereditarias
5.
Front Immunol ; 14: 1149686, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37545537

RESUMEN

Introduction: Osteoarthritis (OA) is a chronic disease with high morbidity and disability rates whose molecular mechanism remains unclear. This study sought to identify OA markers associated with synovitis and cartilage apoptosis by bioinformatics analysis. Methods: A total of five gene-expression profiles were selected from the Gene Expression Omnibus database. We combined the GEO with the GeneCards database and performed Gene Ontology and Kyoto Encyclopedia of Genes and Genome analyses; then, the least absolute shrinkage and selection operator (LASSO) algorithm was used to identify the characteristic genes, and a predictive risk score was established. We used the uniform manifold approximation and projection (UMAP) method to identify subtypes of OA patients, while the CytoHubba algorithm and GOSemSim R package were used to screen out hub genes. Next, an immunological assessment was performed using single-sample gene set enrichment analysis and CIBERSORTx. Results: A total of 56OA-related differential genes were selected, and 10 characteristic genes were identified by the LASSO algorithm. OA samples were classified into cluster 1 and cluster 2 subtypes byUMAP, and the clustering results showed that the characteristic genes were significantly different between these groups. MYOC, CYP4B1, P2RY14, ADIPOQ, PLIN1, MFAP5, and LYVE1 were highly expressed in cluster 2, and ANKHLRC15, CEMIP, GPR88, CSN1S1, TAC1, and SPP1 were highly expressed in cluster 1. Protein-protein interaction network analysis showed that MMP9, COL1A, and IGF1 were high nodes, and the differential genes affected the IL-17 pathway and tumor necrosis factor pathway. The GOSemSim R package showed that ADIPOQ, COL1A, and SPP1 are closely related to the function of 31 hub genes. In addition, it was determined that mmp9 and Fos interact with multiple transcription factors, and the ssGSEA and CIBERSORTx algorithms revealed significant differences in immune infiltration between the two OA subtypes. Finally, a qPCR experiment was performed to explore the important genes in rat cartilage and synovium tissues; the qPCR results showed that COL1A and IL-17A were both highly expressed in synovitis tissues and cartilage tissues of OA rats, which is consistent with the predicted results. Discussion: In the future, common therapeutic targets might be found forsimultaneous remissions of both phenotypes of OA.


Asunto(s)
Condrocitos , Sinovitis , Animales , Ratas , Metaloproteinasa 9 de la Matriz , Biomarcadores , Sinovitis/genética , Apoptosis/genética
6.
J Orthop Surg Res ; 18(1): 535, 2023 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-37496062

RESUMEN

BACKGROUND: N6-methyladenosine (m6A) is a universal RNA modification pattern regulated by multiple m6A regulators. In osteoarthritis (OA), m6A regulators influence disease progression by regulating cartilage degradation. However, the function of m6A regulators in synovial tissue remains unclear. In this work, we investigated the biological significance of m6A regulators in osteoarthritic synovitis. METHODS: Datasets were acquired from Gene Expression Omnibus. Differential analysis of merged data identified the differentially expressed m6A regulators. Machine learning models were used to evaluate genetic importance. To predict disease risk, a nomogram was constructed based on above m6A regulators. Cluster analysis divided the OA sample into different subgroups. Immune infiltration revealed the immune m6A regulators, which were validated using clinical samples. Eventually, a competing endogenous RNA (ceRNA) network was constructed. RESULTS: We acquired five differentially expressed m6A regulators and a random forest model. The nomogram accurately predicted disease risk. We identified 122 differentially expressed genes between two m6A subgroups. The analysis of immune infiltration showed that YTHDF2 was an immune-related m6A regulator closely related with macrophages. In clinical samples, the protein and mRNA contents of YTHDF2 were consistent with the results of bioinformatic analysis. The ceRNA network based on YTHDF2 revealed 75 lncRNA nodes and 19 miRNA nodes. CONCLUSION: YTHDF2 has a high diagnostic value in the synovitis of OA and significantly influences the immune status of patients. Hence, YTHDF2, a critical m6A regulator, may provide a biomarker for diagnosis and immune therapy of osteoarthritic synovitis.


Asunto(s)
MicroARNs , Sinovitis , Humanos , Factores de Transcripción , Sinovitis/genética , Membrana Sinovial , Adenosina , Proteínas de Unión al ARN
7.
Pediatr Rheumatol Online J ; 20(1): 86, 2022 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-36192768

RESUMEN

BACKGROUND: Blau syndrome (BS) is a rare autoinflammatory disorder with NOD2 gain-of-function mutation and characterized by autoactivation of the NFκB pathway. Classically considered a disease of high penetrance, reports on NOD2 mutations underlining BS with incomplete penetrance is limited. CASE PRESENTATION: The proband is a 9-year-old girl presented with brownish annular infiltrative plaques and symmetric boggy polyarthritis over bilateral wrists and ankles. Her skin biopsy revealed noncaseating granulomas inflammation with multinucleated giant cells. A novel C483W NOD2 mutation was identify in the proband and her asymptomatic father. Functional examinations including autoactivation of the NFκB pathway demonstrated by in vitro HEK293T NOD2 overexpression test as well as intracellular staining of phosphorylated-NFκB in patient's CD11b+ cells were consistent with BS. CONCLUSIONS: We reported a novel C483W NOD2 mutation underlining BS with incomplete penetrance. Moreover, a phosphorylated-NFκB intracellular staining assay of CD11b+ was proposed to assist functional evaluation of NFκB autoactivation in patient with BS.


Asunto(s)
Artritis , Sarcoidosis , Sinovitis , Uveítis , Artritis/genética , Niño , Femenino , Células HEK293 , Humanos , Mutación , Proteína Adaptadora de Señalización NOD2/genética , Penetrancia , Sarcoidosis/diagnóstico , Sarcoidosis/genética , Sinovitis/genética
8.
Cell Death Dis ; 13(6): 567, 2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35739102

RESUMEN

Emerging evidence has shown an imbalance in M1/M2 macrophage polarization to play an essential role in osteoarthritis (OA) progression. However, the underlying mechanistic basis for this polarization is unknown. RNA sequencing of OA M1-polarized macrophages found highly expressed levels of pentraxin 3 (PTX3), suggesting a role for PTX3 in OA occurrence and development. Herein, PTX3 was found to be increased in the synovium and articular cartilage of OA patients and OA mice. Intra-articular injection of PTX3 aggravated, while PTX3 neutralization reversed synovitis and cartilage degeneration. No metabolic disorder or proteoglycan loss were observed in cartilage explants when treated with PTX3 alone. However, cartilage explants exhibited an OA phenotype when treated with culture supernatants of macrophages stimulated with PTX3, suggesting that PTX3 did not have a direct effect on chondrocytes. Therefore, the OA anti-chondrogenic effects of PTX3 are primarily mediated through macrophages. Mechanistically, PTX3 was upregulated by miR-224-5p deficiency, which activated the p65/NF-κB pathway to promote M1 macrophage polarization by targeting CD32. CD32 was expressed by macrophages, that when stimulated with PTX3, secreted abundant pro-inflammation cytokines that induced severe articular cartilage damage. The paracrine interaction between macrophages and chondrocytes produced a feedback loop that enhanced synovitis and cartilage damage. The findings of this study identified a functional pathway important to OA development. Blockade of this pathway and PTX3 may prevent and treat OA.


Asunto(s)
Proteína C-Reactiva , MicroARNs , Osteoartritis , Componente Amiloide P Sérico , Sinovitis , Animales , Condrocitos/metabolismo , Humanos , Macrófagos , Ratones , MicroARNs/metabolismo , Osteoartritis/genética , Osteoartritis/metabolismo , Componente Amiloide P Sérico/genética , Componente Amiloide P Sérico/metabolismo , Sinovitis/genética , Sinovitis/metabolismo
9.
Front Immunol ; 13: 895765, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35711422

RESUMEN

Blau syndrome is a systemic autoinflammatory granulomatous disease caused by mutations in the nucleotide-binding oligomerization domain 2 (NOD2) gene. NOD2 is an intracellular pathogen recognition receptor. Upon binding to muramyl dipeptide (MDP), NOD2 activates the NF-κB pathway, leading to the upregulation of proinflammatory cytokines. Clinical manifestations of Blau syndrome appear in patients before the age of four. Skin manifestations resolve spontaneously in some cases; however, joint and eye manifestations are progressive, and lead to serious complications, such as joint contracture and blindness. Currently, there is no specific curative treatment for the disease. Administration of high-dose oral steroids can improve clinical manifestations; however, treatments is difficult to maintain due to the severity of the side effects, especially in children. While several new therapies have been reported, including JAK inhibitors, anti-IL-6 and anti-IL-1 therapies, anti-TNF therapy plays a central role in the treatment of Blau syndrome. We recently performed an ex vivo study, using peripheral blood and induced pluripotent stem cells from patients. This study demonstrated that abnormal cytokine expression in macrophages from untreated patients requires IFNγ stimulation, and that anti-TNF treatment corrects the abnormalities associated with Blau syndrome, even in the presence of IFNγ. Therefore, although the molecular mechanisms by which the genetic mutations in NOD2 lead to granuloma formation remain unclear, it is possible that prior exposure to TNFα combined with IFNγ stimulation may provide the impetus for the clinical manifestations of Blau syndrome.


Asunto(s)
Sinovitis , Uveítis , Artritis , Niño , Citocinas/metabolismo , Humanos , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , Sarcoidosis , Sinovitis/tratamiento farmacológico , Sinovitis/genética , Sinovitis/metabolismo , Inhibidores del Factor de Necrosis Tumoral , Uveítis/tratamiento farmacológico , Uveítis/genética , Uveítis/metabolismo
10.
Proc Natl Acad Sci U S A ; 119(21): e2116855119, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35594394

RESUMEN

Cartilage mineralization is a tightly controlled process, imperative for skeletal growth and fracture repair. However, in osteoarthritis (OA), cartilage mineralization may impact the joint range of motion, inflict pain, and increase chances for joint effusion. Here we attempt to understand the link between inflammation and cartilage mineralization by targeting Sirtuin 1 (SIRT1) and lymphoid enhancer binding factor 1 (LEF1), both reported to have contrasting effects on cartilage. We find that inflammatory-dependent cleavage of SIRT1 or its cartilage-specific genetic ablation, directly enhanced LEF1 expression accompanied by a catabolic response. Applying a posttraumatic OA (PTOA) model to cartilage-specific Sirt1 nulls displayed severe OA, which was accompanied by synovitis, meniscal mineralization, and osteophyte formation of the lateral joint compartment. Alternatively, cartilage-specific Lef1 nulls presented reduced lateral mineralization, OA severity, and local pain. Differential gene expression analysis revealed that Lef1 ablation reduced nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and Toll-like receptor (Tlr) pathways, while enhancing SRY-Box transcription factor 9 (Sox9) and cartilaginous extracellular matrix genes. The results support a link between inflammation and Lef1-dependent cartilage mineralization, mediated by the inactivation of Sirt1. By ablating Lef1 in a PTOA model, the structural and pain-related phenotypes of OA were reduced, in part, by preventing cartilage mineralization of the lateral joint compartment, partially manifested by meniscal tissue mineralization. Overall, these data provide a molecular axis to link between inflammation and cartilage in a PTOA model.


Asunto(s)
Calcinosis , Cartílago Articular , Osteoartritis , Sinovitis , Calcinosis/metabolismo , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Humanos , Inflamación , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Osteoartritis/genética , Osteoartritis/metabolismo , Dolor , Sinovitis/genética , Sinovitis/patología
11.
Rheumatology (Oxford) ; 61(12): 4952-4961, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-35157043

RESUMEN

OBJECTIVES: To define imaging sub-phenotypes in patients with PsA; determine their association with whole blood gene expression and identify biological pathways characterizing the sub-phenotypes. METHODS: Fifty-five patients with PsA ready to initiate treatment for active disease were prospectively recruited. We performed musculoskeletal ultrasound assessment of the extent of inflammation in the following domains: synovitis, peritenonitis, tenosynovitis and enthesitis. Peripheral whole blood was profiled with RNAseq, and gene expression data were obtained. First, unsupervised cluster analysis was performed to define imaging sub-phenotypes that reflected the predominant tissue involved. Subsequently, principal component analysis was used to determine the association between imaging-defined sub-phenotypes and peripheral blood gene expression profile. Pathway enrichment analysis was performed to identify underlying mechanisms that characterize individual sub-phenotypes. RESULTS: Cluster analysis revealed three imaging sub-phenotypes: (i) synovitis predominant [n = 31 (56%)]; (ii) enthesitis predominant [n = 13 (24%)]; (iii) peritenonitis predominant [n = 11 (20%)]. The peritenonitis-predominant sub-phenotype had the most severe clinical joint involvement, whereas the enthesitis-predominant sub-phenotype had the highest tender entheseal count. Unsupervised clustering of gene expression data identified three sub-phenotypes that partially overlapped with the imaging sub-phenotypes suggesting biological and clinical relevance of these sub-phenotypes. We therefore characterized enriched differential pathways, which included: immune system (innate system, B cells and neutrophil degranulation), complement system, platelet activation and coagulation function. CONCLUSIONS: We identified three sub-phenotypes based on the predominant tissue involved in patients with active PsA. Distinct biological pathways may underlie these imaging sub-phenotypes seen in PsA, suggesting their biological and clinical importance.


Asunto(s)
Artritis Psoriásica , Entesopatía , Sinovitis , Tenosinovitis , Humanos , Artritis Psoriásica/diagnóstico por imagen , Artritis Psoriásica/genética , Artritis Psoriásica/complicaciones , Entesopatía/complicaciones , Tenosinovitis/complicaciones , Sinovitis/diagnóstico por imagen , Sinovitis/genética , Sinovitis/complicaciones , Fenotipo , Expresión Génica
12.
Mod Rheumatol ; 32(6): 1054-1063, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34850088

RESUMEN

OBJECTIVES: To explore the effect of miR-106b on synovial inflammation and damage in rheumatoid arthritis (RA) patients and further to investigate its possible mechanism. METHODS: Quantitative real-time polymerase chain reaction, immunofluorescence, in situ hybridization, and immunohistochemistry assay were used to verify the levels of miR-106b and cytokines. Pearson's correlation analysis was conducted to examine bivariate relationship between miR-106b and cytokines or receptor activator of nuclear factor-κ B ligand (RANKL). Following the isolation of fibroblast-like synoviocytes (FLS), the cultured cells were separately transfected with or without miR-106b mimic. Thereafter, cell proliferation, invasion and migration were measured by Cell Counting Kit-8 assay and Transwell assay, respectively. Furthermore, concentration and expression of cytokines were separately detected by enzyme-linked immunosorbent assay and Western blot. RESULTS: Compared with osteoarthritis, RA patients had a lower level of miR-106b and higher levels of RANKL, tumour necrosis factor-a (TNF-a), and interleukin-6 (IL-6). The relative transcription of miR-106b level was negatively correlated to TNF-a, IL-6, and RNKAL levels in both patients (all P < 0.05). Furthermore, miR-106b overexpression suppressed cell proliferation, migration, and invasion capacity of RA-FLS. CONCLUSIONS: miR-106b overexpression suppresses synovial inflammation and alleviates synovial damage; thus, it may be served as a potential therapeutic target for RA patients.


Asunto(s)
Artritis Reumatoide , MicroARNs , Sinovitis , Artritis Reumatoide/patología , Proliferación Celular/genética , Células Cultivadas , Citocinas/metabolismo , Fibroblastos/metabolismo , Humanos , Inflamación/patología , Interleucina-6/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Sinovitis/genética , Sinovitis/metabolismo
13.
J Allergy Clin Immunol ; 149(1): 176-188.e7, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34175136

RESUMEN

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.


Asunto(s)
Artritis/inmunología , Interferón gamma/inmunología , Macrófagos/inmunología , Sarcoidosis/inmunología , Sinovitis/inmunología , Inhibidores del Factor de Necrosis Tumoral/farmacología , Uveítis/inmunología , Adulto , Artritis/tratamiento farmacológico , Artritis/genética , Línea Celular , Niño , Preescolar , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , FN-kappa B/inmunología , Sarcoidosis/tratamiento farmacológico , Sarcoidosis/genética , Sinovitis/tratamiento farmacológico , Sinovitis/genética , Transcriptoma , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , Uveítis/tratamiento farmacológico , Uveítis/genética , Adulto Joven
14.
FEBS J ; 289(1): 121-139, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34270864

RESUMEN

Matrix metalloproteinase-13 (MMP-13) is a uniquely important collagenase that promotes the irreversible destruction of cartilage collagen in osteoarthritis (OA). Collagenase activation is a key control point for cartilage breakdown to occur, yet our understanding of the proteinases involved in this process is limited. Neutrophil elastase (NE) is a well-described proteoglycan-degrading enzyme which is historically associated with inflammatory arthritis, but more recent evidence suggests a potential role in OA. In this study, we investigated the effect of neutrophil elastase on OA cartilage collagen destruction and collagenase activation. Neutrophil elastase induced significant collagen destruction from human OA cartilage ex vivo, in an MMP-dependent manner. In vitro, neutrophil elastase directly and robustly activated pro-MMP-13, and N-terminal sequencing identified cleavage close to the cysteine switch at 72 MKKPR, ultimately resulting in the fully active form with the neo-N terminus of 85 YNVFP. Mole-per-mole, activation was more potent than by MMP-3, a classical collagenase activator. Elastase was detectable in human OA synovial fluid and OA synovia which displayed histologically graded evidence of synovitis. Bioinformatic analyses demonstrated that, compared with other tissues, control cartilage exhibited remarkably high transcript levels of the major elastase inhibitor, (AAT) alpha-1 antitrypsin (gene name SERPINA1), but these were reduced in OA. AAT was located predominantly in superficial cartilage zones, and staining enhanced in regions of cartilage damage. Finally, active MMP-13 specifically inactivated AAT by removal of the serine proteinase cleavage/inhibition site. Taken together, this study identifies elastase as a novel activator of pro-MMP-13 that has relevance for cartilage collagen destruction in OA patients with synovitis.


Asunto(s)
Inflamación/genética , Elastasa de Leucocito/genética , Metaloproteinasa 13 de la Matriz/genética , Osteoartritis/genética , alfa 1-Antitripsina/genética , Cisteína/genética , Humanos , Inflamación/metabolismo , Inflamación/patología , Metaloproteinasa 3 de la Matriz/genética , Neutrófilos/enzimología , Osteoartritis/metabolismo , Osteoartritis/patología , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Sinovitis/genética , Sinovitis/metabolismo , Sinovitis/patología , Deficiencia de alfa 1-Antitripsina/genética , Deficiencia de alfa 1-Antitripsina/patología
15.
Front Immunol ; 12: 734322, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956173

RESUMEN

Osteoarthritis (OA) may result from impaired ability of synovial macrophages to resolve joint inflammation. Increasing macrophage counts in inflamed joints through injection with bone marrow mononuclear cells (BMNC) induces lasting resolution of synovial inflammation. To uncover mechanisms by which BMNC may affect resolution, in this study, differential transcriptional signatures of BMNC in response to normal (SF) and inflamed synovial fluid (ISF) were analyzed. We demonstrate the temporal behavior of co-expressed gene networks associated with traits from related in vivo and in vitro studies. We also identified activated and inhibited signaling pathways and upstream regulators, further determining their protein expression in the synovium of inflamed joints treated with BMNC or DPBS controls. BMNC responded to ISF with an early pro-inflammatory response characterized by a short spike in the expression of a NF-ƙB- and mitogen-related gene network. This response was associated with sustained increased expression of two gene networks comprising known drivers of resolution (IL-10, IGF-1, PPARG, isoprenoid biosynthesis). These networks were common to SF and ISF, but more highly expressed in ISF. Most highly activated pathways in ISF included the mevalonate pathway and PPAR-γ signaling, with pro-resolving functional annotations that improve mitochondrial metabolism and deactivate NF-ƙB signaling. Lower expression of mevalonate kinase and phospho-PPARγ in synovium from inflamed joints treated with BMNC, and equivalent IL-1ß staining between BMNC- and DPBS-treated joints, associates with accomplished resolution in BMNC-treated joints and emphasize the intricate balance of pro- and anti-inflammatory mechanisms required for resolution. Combined, our data suggest that BMNC-mediated resolution is characterized by constitutively expressed homeostatic mechanisms, whose expression are enhanced following inflammatory stimulus. These mechanisms translate into macrophage proliferation optimizing their capacity to counteract inflammatory damage and improving their general and mitochondrial metabolism to endure oxidative stress while driving tissue repair. Such effect is largely achieved through the synthesis of several lipids that mediate recovery of homeostasis. Our study reveals candidate mechanisms by which BMNC provide lasting improvement in patients with OA and suggests further investigation on the effects of PPAR-γ signaling enhancement for the treatment of arthritic conditions.


Asunto(s)
Células de la Médula Ósea/inmunología , Leucocitos Mononucleares/inmunología , Osteoartritis/complicaciones , Osteoartritis/inmunología , Sinovitis/complicaciones , Sinovitis/inmunología , Transcriptoma/genética , Animales , Articulaciones del Carpo/inmunología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Genómica/métodos , Caballos , Lipopolisacáridos/efectos adversos , Macrófagos/inmunología , Masculino , Osteoartritis/genética , Líquido Sinovial/inmunología , Sinovitis/inducido químicamente , Sinovitis/genética
16.
Arthritis Res Ther ; 23(1): 294, 2021 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-34863246

RESUMEN

BACKGROUND: The use of interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitor as a treatment for the inflammatory joint disease is a promising method. However, its underlying mechanism in osteoarthritis (OA) remains unclear. The purpose of this study is to look into the effects of adenovirus-mediated knockdown of IRAK4 on synovitis in the OA rabbit model. METHODS: Ad-shIRAK4 was injected two weeks after anterior cruciate ligament resection. Six weeks later, the rabbits were killed. The expression of IRAK4, TNFR-associated factor 6(TRAF6), TGF-activated kinase 1(TAK1), p-IKB kinase (p-IKK), p-nuclear factor kappa-B (p-NFκB), p38, and p-p38 in the synovial membrane was detected by western blot, qRT-PCR, and immunohistochemistry analysis. Immunohistochemistry was to detect the expression of IRAK4 proteins in articular cartilage. H&E staining was to assess the pathological changes of synovium and cartilage. The levels of interleukin (IL)-1ß, tumor necrosis factor-α(TNF-α), and MMP-13 in the synovial fluid were measured by ELISA. X-ray and micro-computerized tomography (µCT) scans were used to assess knee joint conditions and microstructure of subchondral bone. RESULTS: IRAK4 expression levels in synovial tissues of the OA model group exhibited a significant upward trend. Ad-shIRAK4 significantly reduced IRAK4 mRNA expression in synovium tissues. Notably, Ad-shIRAK4 suppressed the Toll-like receptor/interleukin-1 receptor (TLR/IL-1R) signaling. In addition, in the Ad-shIRAK4 treatment group, we can see less inflammatory cell infiltration and reduced hyperplasia and angiogenesis. The levels of IL-1ß, TNF-α, and MMP-13 in the synovial fluid in the OA model group were significantly higher than that in the control group, which were reduced by Ad-shIRAK4 treatment. Finally, Results of HE stains, immunohistochemistry, and µCT showed that Ad-shIRAK4 treatment has a protective effect on cartilage damage. CONCLUSIONS: IRAK4 is significantly upregulated in the synovium from the osteoarthritis rabbit model. In addition, Ad-shIRAK4 reduced the expression of IRAK4 and suppressed TLR/IL-1R signaling in the synovium from the osteoarthritis rabbit model. Ad-shIRAK4 could alleviate synovitis and cartilage degradation in the osteoarthritis rabbit model, and thus alleviate the symptoms of OA and prevent the progression of OA.


Asunto(s)
Cartílago Articular , Quinasas Asociadas a Receptores de Interleucina-1 , Osteoartritis , Sinovitis , Adenoviridae/genética , Animales , Técnicas de Silenciamiento del Gen , Quinasas Asociadas a Receptores de Interleucina-1/genética , Osteoartritis/genética , Conejos , Membrana Sinovial , Sinovitis/genética
17.
Ophthalmic Genet ; 42(6): 753-764, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34251956

RESUMEN

BACKGROUND: Blau syndrome (BS) is a rare dominantly-inherited autoinflammatory disorder characterized by the triad of arthritis, uveitis and dermatitis that is consequence of gain-of-function NOD2 mutations. We describe the clinical features and genetic basis of a family with two affected members in consecutive generations affected with childhood onset arthritis and uveitis. MATERIALS AND METHODS: Clinical features were retrospectively collected from clinical records. Genetic studies were performed using the Sanger method of DNA sequencing. RESULTS: The proband is a 44 years-old female, who was diagnosed with juvenile onset arthritis at the age of 9 years. She subsequently developed uveitis at age 12 and since then she was managed between the uveitis and rheumatology services. The proband's daughter developed episcleritis at the age of 7 years, and arthritis with bilateral intermediate uveitis two years later. NOD2 analyses revealed in both patients the heterozygous c.1494A>C transversion, predicted to lead the novel, missense p.E498D variant in the NOD2 protein. Additional studies including databases searches and in silico bioinformatic predictions strongly support the "likely pathogenic" classification for this novel variant. CONCLUSIONS: We report a novel pathogenic NOD2 variant in a multiplex family with clinical features compatible with the BS diagnosis. This condition is inherited as a dominant trait in its familial form and should be considered in patients with granulomatous uveitis in association with arthritis and/or dermatitis. Further insight into NOD2 variants and their downstream effects may have implications in the treatment of BS and other inflammatory granulomatous diseases.


Asunto(s)
Artritis/genética , Mutación Missense/genética , Proteína Adaptadora de Señalización NOD2/genética , Polimorfismo de Nucleótido Simple , Sarcoidosis/genética , Sinovitis/genética , Uveítis/genética , Adulto , Artritis/diagnóstico , Niño , Femenino , Angiografía con Fluoresceína , Humanos , Relaciones Madre-Hijo , Madres , Núcleo Familiar , Linaje , Estudios Retrospectivos , Sarcoidosis/diagnóstico , Análisis de Secuencia de ADN , Sinovitis/diagnóstico , Tomografía de Coherencia Óptica , Uveítis/diagnóstico
18.
Sci Rep ; 11(1): 12455, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-34127696

RESUMEN

Adult onset Still's disease (AOSD) is a rare systemic autoinflammatory disease, characterised by fever, arthritis, and skin rash, and joint involvement is one of its clinical manifestations. The aims of this work were to assess joint involvement, to describe main patterns of involvement, and associated clinical characteristics. In this work, we aimed at assessing the joint involvement in AOSD by using MRI, to describe main patterns and associated clinical characteristics. In addition, we aimed at assessing the global transcriptomic profile of synovial tissues in AOSD to elucidate possible pathogenic pathways involved. We also evaluated the global transcriptomic profile of synovial tissues to elucidate possible pathogenic pathways involved in the disease. Thus, AOSD patients, who underwent to MRI exam on joints, were assessed to describe patterns of joint involvement and associated clinical characteristics. Some synovial tissues were collected for RNA-sequencing purposes. The most common MRI finding was the presence of synovitis on 60.5%, mainly in peripheral affected joints, with low to intermediate signal intensity on T1-weighted images and intermediate to high signal intensity on T2-fat-saturated weighted and STIR images. Bone oedema and MRI-bone erosions were reported on 34.9% and 25.6% MRI exams, respectively. Patients with MRI-bone erosions showed a higher prevalence of splenomegaly, a more frequent chronic disease course, lower levels of erythrocyte sedimentation rate, and ferritin. In AOSD synovial tissues, a hyper-expression of interleukin (IL)-1, IL-6, and TNF pathways was shown together with ferritin genes. In conclusion, in AOSD patients, the most common MRI-finding was the presence of synovitis, characterised by intermediate to high signal intensity on T2-fat-saturated weighted and STIR images. MRI-bone erosions and bone oedema were also observed. In AOSD synovial tissues, IL-1, IL-6, and TNF pathways together with ferritin genes resulted to be hyper-expressed.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Enfermedad de Still del Adulto/complicaciones , Membrana Sinovial/diagnóstico por imagen , Sinovitis/inmunología , Adulto , Femenino , Humanos , Interleucina-1/genética , Interleucina-1/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , RNA-Seq , Transducción de Señal/genética , Transducción de Señal/inmunología , Enfermedad de Still del Adulto/genética , Enfermedad de Still del Adulto/inmunología , Enfermedad de Still del Adulto/patología , Membrana Sinovial/inmunología , Membrana Sinovial/patología , Sinovitis/genética , Sinovitis/patología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
19.
J Cell Mol Med ; 25(14): 6664-6678, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34117708

RESUMEN

Synovial macrophage polarization and inflammation are essential for osteoarthritis (OA) development, yet the molecular mechanisms and regulation responsible for the pathogenesis are still poorly understood. Here, we report that pseudolaric acid B (PAB) attenuated articular cartilage degeneration and synovitis during OA. PAB, a diterpene acid, specifically inhibited NF-κB signalling and reduced the production of pro-inflammatory cytokines, which further decreased M1 polarization and vessel formation. We further provide in vivo and in vitro evidences that PAB suppressed NF-κB signalling by stabilizing PPARγ. Using PPARγ antagonist could abolish anti-inflammatory effect of PAB and rescue the activation of NF-κB signalling during OA. Our findings identify a previously unrecognized role of PAB in the regulation of OA and provide mechanisms by which PAB regulates NF-κB signalling through PPARγ, which further suggest targeting synovial inflammation or inhibiting vessel formation at early stage could be an effective preventive strategy for OA.


Asunto(s)
Diterpenos/farmacología , Osteoartritis/tratamiento farmacológico , PPAR gamma/genética , Sinovitis/tratamiento farmacológico , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/crecimiento & desarrollo , Cartílago Articular/efectos de los fármacos , Cartílago Articular/crecimiento & desarrollo , Cartílago Articular/patología , Condrocitos/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/parasitología , Ratones , FN-kappa B/genética , Osteoartritis/genética , Osteoartritis/patología , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Sinovitis/genética , Sinovitis/patología , Factor de Transcripción ReIA/genética
20.
Immunol Res ; 69(3): 225-238, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33983569

RESUMEN

P53 is a transcription factor that regulates many signaling pathways like apoptosis, cell cycle, DNA repair, and cellular stress responses. P53 is involved in inflammatory responses through the regulation of inflammatory signaling pathways, induction of cytokines, and matrix metalloproteinase expression. Also, p53 regulates immune responses through modulating Toll-like receptors expression and innate and adaptive immune cell differentiation and maturation. P53 is a modulator of the apoptosis and proliferation processes through regulating multiple anti and pro-apoptotic genes. Rheumatoid arthritis (RA) is categorized as an invasive inflammatory autoimmune disease with irreversible deformity of joints and bone resorption. Different immune and non-immune cells contribute to RA pathogenesis. Fibroblast-like synoviocytes (FLSs) have been recently introduced as a key player in the pathogenesis of RA. These cells in RA synovium produce inflammatory cytokines and matrix metalloproteinases which results in synovitis and joint destruction. Besides, hyper proliferation and apoptosis resistance of FLSs lead to synovial hyperplasia and bone and cartilage destruction. Given the critical role of p53 in inflammation, apoptosis, and cell proliferation, lack of p53 function (due to mutation or low expression) exerts a prominent role for this gene in the pathogenesis of RA. This review focuses on the role of p53 in different mechanisms and cells (specially FLSs) that involved in RA pathogenesis.


Asunto(s)
Artritis Reumatoide/genética , Epigénesis Genética/inmunología , Sinoviocitos/patología , Sinovitis/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/genética , Apoptosis/inmunología , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Cartílago Articular/inmunología , Cartílago Articular/metabolismo , Cartílago Articular/patología , Proliferación Celular/genética , Citocinas/metabolismo , Metilación de ADN , Humanos , Hiperplasia/genética , Hiperplasia/inmunología , Hiperplasia/patología , Mutación con Pérdida de Función , Metaloproteinasas de la Matriz/metabolismo , Ratones , Transducción de Señal/genética , Transducción de Señal/inmunología , Membrana Sinovial/citología , Membrana Sinovial/inmunología , Membrana Sinovial/patología , Sinoviocitos/inmunología , Sinoviocitos/metabolismo , Sinovitis/inmunología , Sinovitis/patología , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA