Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(1)2022 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-36614150

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune chronic inflammatory disease that is still not well understood in terms of its pathogenesis and presents diagnostic and therapeutic challenges. Monocytes are key players in initiating and maintaining inflammation through the production of pro-inflammatory cytokines and S100 proteins in RA. This study aimed to test a specific DNA methylation inhibitor (RG108) and activator (budesonide) in the regulation of pro-inflammatory mediators-especially the S100 proteins. We also searched for new biomarkers of high disease activity in RA patients. RNA sequencing analysis of healthy controls (HCs) and RA monocytes was performed. Genes such as the S100 family, TNF, and IL-8 were validated by qRT-PCR following DNA-methylation-targeted drug treatment in a monocytic THP-1 cell line. The concentrations of the S100A8, S100A11, and S100A12 proteins in the sera and synovial fluids of RA patients were tested and correlated with clinical parameters. We demonstrated that RA monocytes had significantly increased levels of S100A8, S100A9, S100A11, S100A12, MYD88, JAK3, and IQGAP1 and decreased levels of IL10RA and TGIF1 transcripts. In addition, stimulation of THP-1 cells with budesonide statistically reduced the expression of the S100 family, IL-8, and TNF genes. In contrast, THP-1 cells treated with RG108 had increased levels of the S100 family and TNF genes. We also revealed a significant upregulation of S100A8, S100A11, and S100A12 in RA patients, especially in early RA compared to HC sera. In addition, protein levels of S100A8, S100A11, and S100A12 in RA synovial fluids compared to HC sera were significantly increased. Overall, our data suggest that the S100A8 and S100A12 proteins are strongly elevated during ongoing inflammation, so they could be used as a better biomarker of disease activity than CRP. Interestingly, epigenetic drugs can regulate these S100 proteins, suggesting their potential use in targeting RA inflammation.


Asunto(s)
Artritis Reumatoide , Proteína S100A12 , Humanos , Proteína S100A12/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/genética , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Proteínas S100/metabolismo , Inflamación/metabolismo , Biomarcadores , Budesonida/uso terapéutico , Epigénesis Genética , Proteínas Represoras/metabolismo , Proteínas de Homeodominio/metabolismo
2.
Int J Mol Sci ; 23(10)2022 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-35628127

RESUMEN

Complex pathogenesis of systemic lupus erythematosus (SLE) and systemic sclerosis (SSc) is associated with an imbalance of various Th-cell subpopulations. Mesenchymal stem cells (MSCs) have the ability to restore this balance. However, bone marrow-derived MSCs of SLE and SSc patients exhibit many abnormalities, whereas the properties of adipose derived mesenchymal stem cells (ASCS) are much less known. Therefore, we examined the effect of ASCs obtained from SLE (SLE/ASCs) and SSc (SSc/ASCs) patients on Th subset differentiation, using cells from healthy donors (HD/ASCs) as controls. ASCs were co-cultured with activated CD4+ T cells or peripheral blood mononuclear cells. Expression of transcription factors defining Th1, Th2, Th17, and regulatory T cell (Tregs) subsets, i.e., T-bet, GATA3, RORc, and FoxP3, were analysed by quantitative RT-PCR, the concentrations of subset-specific cytokines were measured by ELISA, and Tregs formation by flow cytometry. Compared with HD/ASCs, SLE/ASCs and especially SSc/ASCs triggered Th differentiation which was disturbed at the transcription levels of genes encoding Th1- and Tregs-related transcription factors. However, we failed to find functional consequences of this abnormality, because all tested ASCs similarly switched differentiation from Th1 to Th2 direction with accompanying IFNγ/IL-4 ratio decrease, up-regulated Th17 formation and IL-17 secretion, and up-regulated classical Tregs generation.


Asunto(s)
Lupus Eritematoso Sistémico , Células Madre Mesenquimatosas , Enfermedades Reumáticas , Esclerodermia Sistémica , Tejido Adiposo/metabolismo , Diferenciación Celular , Humanos , Leucocitos Mononucleares , Lupus Eritematoso Sistémico/metabolismo , Células Madre Mesenquimatosas/metabolismo , Enfermedades Reumáticas/metabolismo , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo
3.
Eur J Immunol ; 50(7): 1057-1066, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32087087

RESUMEN

Dysregulation in type I IFN and IFN-stimulated genes (ISGs) induced by monocytes is one of the key features of systemic sclerosis (SSc) pathogenesis. Abnormalities in microRNA (miRNA) expression are related to excessive IFN production, however the role of miRNA remains largely elusive in SSc monocytes. This study explores global miRNA-mRNA profiling of SSc monocytes and functional attenuation of IFN and ISGs by specific miRNAs. Global sequencing of mRNA (mRNA-seq) and miRNA (miRNA-seq) samples were performed simultaneously on healthy controls and SSc monocytes. Following computational analysis, selected miRNAs-mRNA candidates were validated, correlated with clinical parameters, and tested by functional assays. Transcriptomics data and qPCR analysis confirmed IFN signature in SSc but not in rheumatoid arthritis monocytes. Based on miRNA-seq analysis, five miRNAs were selected for further validation. Only the expression patterns of miRNA-26a-2-3p and miRNA-485-3p were confirmed and negatively correlated with clinical parameters. Exogenous delivery of miRNA-26a-2-3p to TLR-stimulated monocytic THP-1 cells specifically inhibited ISGs but not inflammasome activity in functional assays. In conclusion, our miRNA-mRNA co-sequencing and functional analysis identify miRNA-26a-2-3p as a new candidate, which is predicated to negatively regulate ISGs. This implies that reduced expression of miRNA-26a-2-3 may be involved in pathogenic IFN signature in SSc monocytes.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Interferones/inmunología , MicroARNs/inmunología , Monocitos/inmunología , ARN Mensajero/inmunología , Esclerodermia Sistémica/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Esclerodermia Sistémica/patología , Células THP-1
4.
Rheumatology (Oxford) ; 60(11): 5424-5435, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34009317

RESUMEN

OBJECTIVE: To explore global miRNA and transcriptomic profiling of monocytes from RA patients compared with healthy controls in order to predict which aberrantly expressed miRNA can negatively modulate inflammatory molecules. METHODS: Using next-generation sequencing, we have performed simultaneous global analysis of miRNA (miRNA-seq) and transcriptome (RNA-seq) of monocytes from RA patients and healthy controls. Global analysis of miRNA of SSc monocytes was also performed. Following differential analysis and negative correlation, miRNA-RNA pairs were selected. RESULTS: We found that 20 specific miRNA candidates are predicted to silence inflammatory mediators, out of 191 significantly changed miRNAs in RA monocytes. Based on the highest scoring in terms of negative correlation (r = -0.97, P = 1.75e-07, false discovery rate = 0.04) and the number of seeds in miRNA responsible for negative regulation, we selected miRNA-146b and its target gene anti-inflammatory retinoic acid receptor alpha (RARA). Similarly to next-generation sequencing, qPCR analysis also confirmed negative correlation between miRNA-146b and RARA expression (r = -0.45, P = 0.04). Additionally, miRNA-146b expression in RA monocytes significantly correlated with clinical parameters including DAS28 for RA with CRP (DAS28-CRP) and ESR (DAS28-ESR), whereas overexpression of miRNA-146b was able to functionally reduce RARA expression in the human monocytic cell line THP-1. Finally, circulating miRNA-146b expression in sera and SFs was significantly elevated in RA patients. CONCLUSIONS: Overall, in this study we have identified a new miRNA-146b candidate that is predicted to negatively regulate the anti-inflammatory RARA transcript, whereas circulating miRNA-146b level can be used as a biomarker predicting pro-inflammatory RA progression and disease activity.


Asunto(s)
Artritis Reumatoide/metabolismo , MicroARNs/sangre , Monocitos/metabolismo , Estudios de Casos y Controles , Línea Celular , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Receptor alfa de Ácido Retinoico/genética , Líquido Sinovial/metabolismo , Transcriptoma
5.
Eur J Clin Invest ; 47(8): 555-564, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28639412

RESUMEN

BACKGROUND: Systemic sclerosis (SSc) is a chronic autoimmune disease characterised by tissue fibrosis and immune abnormalities. Recent evidence suggests that activated circulating monocytes from patients with SSc play an important role in early stages of SSc pathogenesis due to enhanced expression of tissue inhibitor of metalloproteinases 1 (TIMP-1), IL-8 and reactive oxygen species (ROS) induction. However, the exact factors that contribute to chronic inflammation and subsequently fibrosis progression are still unknown. MATERIALS AND METHODS: The expression pattern of IL-8, TIMP-1, AP-1 transcription factor-Fra2 and ROS induction in peripheral blood monocytes following DZNep (histone methyltransferase inhibitor) and TLR8 agonist stimulation was investigated. Exogenous microRNA-5196, which is predicted to bind 3'UTR of Fra2 gene, was delivered to reverse profibrotic phenotype in monocytes. Expression of circulating microRNA-5196 was correlated with SSc parameters. RESULTS: DZNep + TLR8 agonist stimulation enhanced profibrotic TIMP-1, IL-8 and ROS generation in HC and SSc monocytes. As opposed by the decrease of miRNA-5196 and antioxidant SOD1 expression in SSc monocytes. Exogenous delivery of microRNA-5196 reduced Fra2 and TIMP-1 expression suggesting that it may be used as a potential modulator of fibrogenesis in SSc. Circulating microRNA-5196 was significantly increased in SSc and positively correlated with CRP level but not with Rodnan skin score or ESR. CONCLUSIONS: These results suggest that microRNA-5196 can be used as a potential biomarker characterising SSc. Overall, this study may open new possibilities for the development of microRNA-5196-based diagnostics and therapy in early phases of SSc.


Asunto(s)
MicroARNs/metabolismo , Esclerodermia Sistémica/etiología , Adenosina/análogos & derivados , Adenosina/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Estudios de Casos y Controles , Femenino , Antígeno 2 Relacionado con Fos/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Interleucina-8/metabolismo , Leucocitos Mononucleares/metabolismo , Masculino , Metaloproteinasas de la Matriz/metabolismo , MicroARNs/fisiología , Persona de Mediana Edad , Estrés Oxidativo/fisiología , Péptidos Cíclicos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Receptor Toll-Like 8/antagonistas & inhibidores , Transfección
6.
Mediators Inflamm ; 2016: 9607946, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27594771

RESUMEN

Systemic inflammatory rheumatic diseases are considered as autoimmune diseases, meaning that the balance between recognition of pathogens and avoidance of self-attack is impaired and the immune system attacks and destroys its own healthy tissue. Treatment with conventional Disease Modifying Antirheumatic Drugs (DMARDs) and/or Nonsteroidal Anti-Inflammatory Drugs (NSAIDs) is often associated with various adverse reactions due to unspecific and toxic properties of those drugs. Although biologic drugs have largely improved the outcome in many patients, such drugs still pose significant problems and fail to provide a solution to all patients. Therefore, development of more effective treatments and improvements in early diagnosis of rheumatic diseases are badly needed in order to increase patient's functioning and quality of life. The reversible nature of epigenetic mechanisms offers a new class of drugs that modulate the immune system and inflammation. In fact, epigenetic drugs are already in use in some types of cancer or cardiovascular diseases. Therefore, epigenetic-based therapeutics that control autoimmunity and chronic inflammatory process have broad implications for the pathogenesis, diagnosis, and management of rheumatic diseases. This review summarises the latest information about potential therapeutic application of epigenetic modification in targeting immune abnormalities and inflammation of rheumatic diseases.


Asunto(s)
Epigénesis Genética/genética , Epigénesis Genética/fisiología , Enfermedades Reumáticas/tratamiento farmacológico , Enfermedades Reumáticas/terapia , Animales , Antiinflamatorios/efectos adversos , Antiinflamatorios/uso terapéutico , Antirreumáticos/efectos adversos , Antirreumáticos/uso terapéutico , Autoinmunidad/genética , Autoinmunidad/fisiología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/terapia , Enfermedades Reumáticas/genética
7.
J Biol Chem ; 289(14): 9952-60, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24550394

RESUMEN

Fibrosis is a common and intractable condition associated with various pathologies. It is characterized by accumulation of an excessive amount of extracellular matrix molecules that primarily include collagen type I. IL-6 is a profibrotic cytokine that is elevated in the prototypic fibrotic autoimmune condition systemic sclerosis and is known to induce collagen I expression, but the mechanism(s) behind this induction are currently unknown. Using healthy dermal fibroblasts in vitro, we analyzed the signaling pathways that underscore the IL-6-mediated induction of collagen. We show that IL-6 trans signaling is important and that the effect is dependent on STAT3; however, the effect is indirect and mediated through enhanced TGF-ß signaling and the classic downstream cellular mediator Smad3. This is due to induction of the bone morphogenetic protein (BMP) antagonist Gremlin-1, and we show that Gremlin-1 is profibrotic and is mediated through canonical TGF-ß signaling.


Asunto(s)
Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas Morfogenéticas Óseas/genética , Células Cultivadas , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/genética , Femenino , Fibroblastos/patología , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Interleucina-6/genética , Factor de Transcripción STAT3/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta/genética
8.
Exp Dermatol ; 24(6): 401-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25712746

RESUMEN

Systemic sclerosis is an autoimmune disease characterised by vascular dysfunction, impaired angiogenesis, inflammation and fibrosis. There is no currently accepted disease-modifying treatment with only autologous stem cell transplant showing clinically meaningful benefit. The lack of treatment options reflects our lack of understanding of the precise molecular mechanisms occurring in the disease. Recent investigations have begun to decipher the molecular pathways underpinning the different aspects of the disease and may provide a rational clinical target(s). Uncovering the molecular mechanisms of the disease is important in understanding systemic sclerosis treatment. The aim of this review was to examine the current thinking in SSc pathogenesis and will offer novel areas for research which may yield novel therapeutics.


Asunto(s)
Medicina Molecular/tendencias , Esclerodermia Sistémica/etiología , Manejo de la Enfermedad , Epigénesis Genética/fisiología , Perfilación de la Expresión Génica , Humanos , Esclerodermia Sistémica/fisiopatología , Esclerodermia Sistémica/terapia
9.
Immunology ; 143(3): 331-40, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24476318

RESUMEN

Systemic sclerosis is an autoimmune idiopathic connective tissue disease, characterized by vasculopathy, inflammation and fibrosis. There appears to be a link between inflammation and fibrosis, although the exact nature of the relationship is unknown. Serum amyloid A (SAA) is an acute-phase protein that is elevated up to 1000-fold in times of infection or inflammation. This acute-phase reactant, as well as being a marker of inflammation, may initiate signals in a cytokine-like manner, possibly through toll-like receptors (TLRs) promoting inflammation. This study addressed the role of SAA in initiating interleukin-6 (IL-6) production in dermal fibroblasts and the role of TLR2 in this system. We show that SAA induces IL-6 secretion in healthy dermal fibroblasts and that blockade of TLR2 with a neutralizing antibody to TLR2 or specific small interfering RNA attenuated the SAA-induced IL-6 secretion and that this was also mediated through the TLR adaptor protein IL-1 receptor-associated kinase 4. The effect is nuclear factor-κB-mediated because blockade of nuclear factor-κB reduced the induction. We also demonstrate that dermal fibroblasts express TLR2; this is functional and over-expressed in the fibroblasts of patients with systemic sclerosis. Taken together these data suggest that SAA is a danger signal that initiates IL-6 signalling in systemic sclerosis via enhanced TLR2 signalling.


Asunto(s)
Fibroblastos/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-6/metabolismo , FN-kappa B/metabolismo , Proteína Amiloide A Sérica/metabolismo , Piel/metabolismo , Receptor Toll-Like 2/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Expresión Génica , Humanos , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Proteína Amiloide A Sérica/farmacología , Piel/patología , Receptor Toll-Like 2/genética
10.
Immunology ; 141(1): 70-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24032649

RESUMEN

The majority of studies examining antigen-presenting cell (APC) function have focused on the capture and presentation of antigens released from pathogens or damaged cells. However, antigen-specific B cells are also capable of efficiently extracting antigens that are either tethered to, or integrally part of the plasma membrane of various target cells. In this study we show that B cells are also highly efficient at extracting integral components of the extracellular matrix (ECM) for subsequent presentation. In particular we demonstrate that B cells specific for aggrecan, an integral component of cartilage ECM, acquire this rheumatoid arthritis candidate autoantigen in both a B-cell-receptor-dependent and a contact-dependent manner. We also demonstrate that the subsequent presentation of aggregan from ECM leads to CD4(+) T-cell activation and effector cell formation. Recent studies have identified B-cell-mediated antigen presentation as essential for the development of autoimmunity, but a unique role for B cells compared with other APC has yet to be defined. Our findings lead us to propose that the acquisition of ECM-derived autoantigens represents a mechanism that defines the APC requirement for B cells in the development of autoimmunity.


Asunto(s)
Agrecanos/inmunología , Presentación de Antígeno , Autoantígenos/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Cartílago/inmunología , Matriz Extracelular/inmunología , Activación de Linfocitos , Animales , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Linfocitos B/patología , Linfocitos T CD4-Positivos/patología , Cartílago/patología , Bovinos , Línea Celular Tumoral , Matriz Extracelular/patología , Humanos , Ratones
11.
Sci Rep ; 14(1): 14416, 2024 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-38909091

RESUMEN

The COVID-19 pandemic has profoundly affected all aspects of our lives. Through real-time monitoring and rapid vaccine implementation, we succeeded in suppressing the spread of the disease and mitigating its consequences. Finally, conclusions can be summarized and drawn. Here, we use the example of Poland, which was seriously affected by the pandemic. Compared to other countries, Poland has not achieved impressive results in either testing or vaccination, which may explain its high mortality (case fatality rate, CFR 1.94%). Through retrospective analysis of data collected by the COVID-19 Data Portal Poland, we found significant regional differences in the number of tests performed, number of cases detected, number of COVID-19-related deaths, and vaccination rates. The Masovian, Greater Poland, and Pomeranian voivodeships, the country's leaders in vaccination, reported high case numbers but low death rates. In contrast, the voivodeships in the eastern and southern parts of Poland (Subcarpathian, Podlaskie, Lublin, Opole), which documented low vaccination levels and low case numbers, had higher COVID-19-related mortality rates. The strong negative correlation between the CFR and the percentage of the population that was vaccinated in Poland supports the validity of vaccination. To gain insight into virus evolution, we sequenced more than 500 genomes and analyzed nearly 80 thousand SARS-CoV-2 genome sequences deposited in GISAID by Polish diagnostic centers. We showed that the SARS-CoV-2 variant distribution over time in Poland reflected that in Europe. Haplotype network analysis allowed us to follow the virus transmission routes and identify potential superspreaders in each pandemic wave.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Pandemias , SARS-CoV-2 , Polonia/epidemiología , COVID-19/epidemiología , COVID-19/virología , COVID-19/prevención & control , Humanos , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Estudios Retrospectivos , Genoma Viral , Genómica/métodos , Vacunación
12.
Expert Rev Mol Med ; 15: e9, 2013 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-23985302

RESUMEN

Accumulative evidence demonstrates the crucial role of evolutionary conserved Toll-like receptors (TLRs) in identifying microbial or viral compounds. TLRs are also able to recognise endogenous molecules which are released upon cell damage or stress and have been shown to play a key role in numerous autoimmune diseases including systemic sclerosis (SSc). A classic feature of SSc, is vascular injury manifested as Raynaud's phenomenon and ischaemia of the skin, resulting in the release of endogenous TLR ligands during inflammation and local tissue damage. These locally released TLR ligands bind TLRs possibly complexed to autoantibodies, and initiate intracellular signalling pathways and may be one of the mechanisms that initiate and drive autoimmunity and subsequent fibrosis. Activation of the immune system results in interferon (IFN) sensitive gene transcription. There is also an IFN gene signature in SSc peripheral blood. TLRs may represent the link between immune activation, common in SSc, and tissue fibrosis. Therefore, a better understanding of the mechanisms of TLR-mediated pathogenesis and therapies targeting individual TLRs, may provide a more specific approach of treating multi-systemic autoimmune diseases. This review aims to integrate the current knowledge of TLR function in the autoimmune disorders with particular emphasis on SSc. We suggest the TLR system as a new therapeutic target.


Asunto(s)
Esclerodermia Sistémica/inmunología , Receptores Toll-Like/metabolismo , Animales , Autoanticuerpos/inmunología , Autoinmunidad , Humanos , Esclerodermia Sistémica/etiología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/terapia , Transducción de Señal , Receptores Toll-Like/inmunología , Transcripción Genética
13.
Ann Rheum Dis ; 72(8): 1382-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23223421

RESUMEN

OBJECTIVES: To investigate whether monocytes contribute to matrix deposition in systemic sclerosis (SSc) by production of tissue-inhibitor of metalloproteinase-1 (TIMP-1). METHODS: Matrix metalloproteinase-1 (MMP-1) and TIMP-1 expression and secretion were measured by qRT-PCR and ELISA in circulating monocytes from patients with SSc, patients with rheumatoid arthritis (RA) and healthy controls (HC) and in healthy monocytes cultured in the presence of SSc or HC serum samples. Production of TIMP-1 was determined in response to a panel of Toll-like receptor (TLR) agonists and MyD88 inhibitory peptide. The functional effect of conditioned media from SSc and HC serum samples or TLR8-stimulated monocytes was studied in an MMP-1 activity assay. RESULTS: TIMP-1 production by monocytes was upregulated in patients with SSc compared with patients with RA and HC. Incubation of HC monocytes with SSc serum samples resulted in functionally active TIMP-1 production. However, pretreatment with MyD88 inhibitor, but not control peptide, decreased TIMP-1 secretion. TIMP-1 production was significantly stronger when SSc and HC monocytes were stimulated with TLR8 (ssRNA) agonist, but the response was more pronounced in SSc monocytes. TIMP-1 production after TLR stimulation was also strongly reduced in the presence of MyD88 inhibitory peptide or in the monocytes isolated from a patient with a genetic TLR signalling defect. MMP-1 activity was significantly inhibited in media from serum samples or TLR8-stimulated monocytes indicative of functional TIMP activity. CONCLUSIONS: This study demonstrates profibrotic properties of circulating monocytes from patients with SSc and a key role for TLR signalling, particularly TLR8, in TIMP-1 secretion and matrix remodelling.


Asunto(s)
Metaloproteinasa 1 de la Matriz/metabolismo , Monocitos/metabolismo , Esclerodermia Sistémica/sangre , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Receptor Toll-Like 8/metabolismo , Artritis Reumatoide/sangre , Artritis Reumatoide/genética , Medios de Cultivo Condicionados/farmacología , Fibrosis/inmunología , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Receptores de Lipopolisacáridos/metabolismo , Metaloproteinasa 1 de la Matriz/genética , Monocitos/efectos de los fármacos , Monocitos/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Esclerodermia Sistémica/genética , Piel/inmunología , Piel/metabolismo , Piel/patología , Inhibidor Tisular de Metaloproteinasa-1/genética , Receptor Toll-Like 8/agonistas , Regulación hacia Arriba
14.
Biomed Pharmacother ; 165: 115254, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37542854

RESUMEN

Despite the development of new biological and synthetic targeted therapies, methotrexate remains one of the most commonly used immunomodulatory drugs in rheumatology. However, its effect on the immunogenicity of vaccines has been studied only to a limited extent until recently, resulting in the lack of clear guidelines on the use of methotrexate during vaccination. Significant progress was made during the COVID-19 pandemic due to the dynamic development of research on vaccines, including patients with autoimmune inflammatory rheumatic diseases. In the following literature review, we present a summary of what we know so far on the impact of methotrexate on post-vaccination response in adult rheumatology patients, taking into account the lessons learned from the COVID-19 pandemic. Studies on the effect of methotrexate on the immunogenicity of influenza, pneumococcal, herpes zoster, tetanus/diphtheria/pertussis, hepatitis A, yellow fever, and COVID-19 vaccines are described in detail, including the effect of methotrexate on the humoral and cellular response of individual vaccines. The available evidence for recommendations for withholding methotrexate in the post-vaccination period is presented. Lastly, an overview of potential immunological mechanisms through which MTX modulates the immunogenicity of vaccinations is also provided.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Vacunas contra la Influenza , Enfermedades Reumáticas , Reumatología , Adulto , Humanos , COVID-19/prevención & control , Vacunas contra la COVID-19/uso terapéutico , Metotrexato/efectos adversos , Pandemias , Enfermedades Reumáticas/tratamiento farmacológico , Vacunación
15.
J Inflamm Res ; 16: 3949-3965, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37706062

RESUMEN

It is well known that fatigue is a highly disabling symptom commonly observed in inflammatory rheumatic diseases (IRDs). Fatigue is strongly associated with a poor quality of life and seems to be an independent predictor of job loss and disability in patients with different rheumatic diseases. Although the pathogenesis of fatigue remains unclear, indirect data suggest the cooperation of the immune system, the central and autonomic nervous system, and the neuroendocrine system in the induction and sustainment of fatigue in chronic diseases. Fatigue does not correspond with disease activity and its mechanism in IRDs. It is suggested that it may change over time and vary between individuals. Abnormal production of pro-inflammatory cytokines such as interleukin-6 (IL-6), interferons (IFNs), granulocyte-macrophage colony-stimulating factor (GM-CSF), TNF, IL-15, IL-17 play a role in both IRDs and subsequent fatigue development. Some of these cytokines such as IL-6, IFNs, GM-CSF, and common gamma-chain cytokines (IL-15, IL-2, and IL-7) activate the Janus Kinases (JAKs) family of intracellular tyrosine kinases. Therapy blocking JAKs (JAK inhibitors - JAKi) has been recently proven to be an effective approach for IRDs treatment, more efficient in pain reduction than anti-TNF. Therefore, the administration of JAKi to IRDs patients experiencing fatigue may find rational implications as a therapeutic modulator not only of disease inflammatory symptoms but also fatigue with its components like pain and neuropsychiatric features as well. In this review, we demonstrate the latest information on the mechanisms of fatigue in rheumatic diseases and the potential effect of JAKi on fatigue reduction.

16.
Front Immunol ; 14: 980247, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37056771

RESUMEN

Introduction: A number of studies have demonstrated a key role of miRNA isolated from cells, tissue or body fluids as disease-specific biomarkers of autoimmune rheumatic diseases including rheumatoid arthritis (RA) and systemic sclerosis (SSc). Also, the expression level of miRNA is changing during disease development, therefore miRNA can be used as biomarkers monitoring RA progression and treatment response. In this study we have investigated the monocytes-specific miRNA that could serve as potential biomarkers of disease progression observed in sera and synovial fluids (SF) in early (eRA) and advanced (aRA) RA and in RA patients before and 3 months after selective JAK inhibitor (JAKi) -baricitinib treatment. Methods: Samples from healthy control (HC) (n=37), RA (n=44) and SSc (n=10) patients were used. MiRNA-seq of HC, RA, and SSc monocytes was performed to find versatile miRNA present in different rheumatic diseases. Selected miRNAs were validated in body fluids in eRA (<2 years disease onset) and aRA (>2 years disease onset) and RA patients receiving baricitinib. Results: Using miRNA-seq, we selected top 6 miRNA out of 95 that were significantly changed in both RA and SSc monocytes compared to HC. To identify circulating miRNA predicting RA progression, these 6 miRNA were measured in eRA and aRA sera and SF. Interestingly, miRNA (-19b-3p, -374a-5p, -3614-5p) were significantly increased in eRA sera vs HC and even further upregulated in SF vs aRA sera. In contrast, miRNA-29c-5p was significantly reduced in eRA sera vs HC and even further decreased in SF vs aRA sera. Kegg pathway analysis predicted that miRNA were involved in inflammatory-mediated pathways. ROC analysis demonstrated that miRNA-19b-3p (AUC=0.85, p=0.04) can be used as biomarker predicting JAKi response. Discussion: In conclusion, we identified and validated miRNA candidates which were present simultaneously in monocytes, sera, SF and that can be used as biomarkers predicting joint inflammation and monitoring therapy response to JAKi in RA patients.


Asunto(s)
Artritis Reumatoide , MicroARN Circulante , MicroARNs , Esclerodermia Sistémica , Humanos , Monocitos/metabolismo , MicroARNs/metabolismo , Artritis Reumatoide/diagnóstico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/genética , Biomarcadores , Progresión de la Enfermedad
17.
Viruses ; 15(3)2023 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-36992329

RESUMEN

Impaired immunogenicity of COVID-19 vaccinations in inflammatory arthritis (IA) patients results in diminished immunity. However, optimal booster vaccination regimens are still unknown. Therefore, this study aimed to assess the kinetics of humoral and cellular responses in IA patients after the COVID-19 booster. In 29 IA patients and 16 healthy controls (HC), humoral responses (level of IgG antibodies) and cellular responses (IFN-γ production) were assessed before (T0), after 4 weeks (T1), and after more than 6 months (T2) from the booster vaccination with BNT162b2. IA patients, but not HC, showed lower anti-S-IgG concentration and IGRA fold change at T2 compared to T1 (p = 0.026 and p = 0.031). Furthermore, in IA patients the level of cellular response at T2 returned to the pre-booster level (T0). All immunomodulatory drugs, except IL-6 and IL-17 inhibitors for the humoral and IL-17 inhibitors for the cellular response, impaired the immunogenicity of the booster dose at T2. Our study showed impaired kinetics of both humoral and cellular responses after the booster dose of the COVID-19 vaccine in IA patients, which, in the case of cellular response, did not allow the vaccination effect to be maintained for more than 6 months. Repetitive vaccination with subsequent booster doses seems to be necessary for IA patients.


Asunto(s)
Artritis , COVID-19 , Humanos , Vacunas contra la COVID-19 , Vacuna BNT162 , Interleucina-17 , COVID-19/prevención & control , Inmunoglobulina G , Vacunación , Anticuerpos Antivirales
18.
Eur J Immunol ; 41(7): 1850-61, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21590765

RESUMEN

The induction of an effective immune response requires the activation of CD4+ T lymphocytes by APCs. While DCs have been shown to be pivotal in this process, it is now apparent that optimal CD4+ T-cell activation also requires B-lymphocyte APC function. Along with the acquisition of soluble antigens, it is known that B cells also acquire membrane-tethered antigens. Recent reports have described the relocation of intracellular antigens to the cell surface following immunogenic apoptosis. This study was designed to determine whether B cells can acquire and present such antigens to CD4+ T cells. By targeting the model antigen tetanus toxin C fragment to various cellular locations, we show that antigen-specific B cells acquire intracellular antigens that have relocated to the surface of cells undergoing immunogenic apoptosis. Crucially, we also demonstrate that antigen-specific B cells acquiring relocated antigen from apoptotic targets are capable of efficiently inducing CD4+ T-cell activation. We propose that the acquisition and presentation of intracellular antigens that have relocated to the cell surface during immunogenic apoptosis represents a novel means by which antigen-specific B cells contribute to the generation of immunity.


Asunto(s)
Presentación de Antígeno , Antígenos de Superficie/inmunología , Apoptosis , Linfocitos B/inmunología , Linfocitos B/metabolismo , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Linfocitos T CD4-Positivos/inmunología , Membrana Celular/inmunología , Citometría de Flujo , Células HeLa , Humanos , Activación de Linfocitos , Ratones , Fragmentos de Péptidos/inmunología , Toxina Tetánica/inmunología
19.
RSC Adv ; 12(7): 4016-4028, 2022 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-35425452

RESUMEN

The biomaterial-cells interface is one of the most fundamental issues in tissue regeneration. Despite many years of scientific work, there is no clear answer to what determines the desired adhesion of cells and the synthesis of ECM proteins. Crystallinity is a characteristic of the structure that influences the surface and bulk properties of semicrystalline polymers used in medicine. The crystallinity of polycaprolactone (PCL) was varied by changing the molecular weight of the polymer and the annealing procedure. Measurements of surface free energy showed differences related to substrate crystallinity. Additionally, the water contact angle was determined to characterise surface wettability which was crucial in the analysis of protein absorption. X-ray photoelectron spectroscopy was used to indicate oxygen bonds amount on the surface. Finally, the impact of the crystallinity, and related properties were demonstrated on dermal fibroblasts' response. Cellular proliferation and expression of selected genes: α-SMA, collagen I, TIMP, integrin were analysed.

20.
Biomedicines ; 10(4)2022 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-35453643

RESUMEN

This study aimed to investigate the associations of microRNA (miRs) signatures with cytokines, serum lipids, and disease activity in patients with psoriatic arthritis (PsA), ankylosing spondylitis (AS), and rheumatoid arthritis (RA). In total, 65 patients (PsA n = 25, AS n = 25, RA n = 15) and 25 healthy controls (HC) were enrolled into the study. The expression of miR-223-5p, miR-92b-3p, miR-485-3p, miR-10b-5p, let-7d-5p, miR-26a-2-3p, miR-146b-3p, and cytokines levels were measured in sera. DIANA-mirPath analysis was used to predict pathways targeted by the dysregulated miRs. Disease activity scores were calculated. Lipid profile, uric acid, glucose level, and C-reactive protein (CRP) concentrations were determined in the blood. Based on lipid profiles, the PsA group had hypertriglyceridaemia, and RA patients revealed mixed dyslipidaemia, while in AS, no specific changes were found. miR expression analysis revealed upregulation of miR-26a-2-3p and miR-10b-5p in PsA, miR-485-3p in AS, and let-7d-5p in RA. Several correlations between disease activity indexes, metabolites levels, and expression of miRs were observed in PsA, RA, and AS patients. Finally, in ROC analysis, miR-26a-2-3p/miR-485-3p, and let-7d-5p/miR-146b-3p tandems revealed high sensitivity and specificity in distinguishing between PsA, AS, and RA. Our study illustrates the superiority of miR expressions in distinguishing between RA, PsA, and AS. In PsA, a unique regulatory pathway exists through miR-26a-2-3p, miR-223-5p, miR-10b-5p, and miR-92b-3p that converges proatherogenic metabolism and disease activity.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA