Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Exp Rheumatol ; 41(7): 1434-1442, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36622109

RESUMEN

OBJECTIVES: Oestrogen deficiency is a rare disease and leads inter alia to arthralgia and osteoporosis in men. The clinical relevance of aromatase to a functioning male metabolism has become evident since 1991, when cases of patients with oestrogen deficiency caused by aromatase mutation were first described. Only few cases are known so far, which will now be presented in a case report and review of the literature. METHODS: All available publications since the first description in 1991 dealing with loss-of-function aromatase mutation in men were summarised and our case report was added. RESULTS: The mutations that cause the aromatase protein to lose function leads to a rather heterogeneous clinical picture. It is, however, clear that oestrogens play a central role in male patients, especially in bone metabolism. Most frequently, tall stature, unclosed epiphyseal joints, and osteoporosis are detected in affected individuals as a consequence of the change in hormonal status. CONCLUSIONS: As low oestrogen is associated with arthralgia, patients with aromatase mutation may be referred to a rheumatologist. Despite aromatase deficiency being a rare disease, the study of the effects of oestrogen on male bone development provides important insights for endocrine bone regulation. It has been demonstrated that androgens alone are not sufficient for adequate skeletal development in males. The described effects of loss of oestrogens are known from the aromatase inhibitor therapy in breast cancer treatment. This work highlights the important role of oestrogens in individual health and disease in men. Molecular effects of oestrogens on bone metabolism are summarised.


Asunto(s)
Aromatasa , Osteoporosis , Humanos , Masculino , Aromatasa/genética , Aromatasa/metabolismo , Enfermedades Raras , Estrógenos , Osteoporosis/tratamiento farmacológico , Osteoporosis/genética , Mutación
2.
Z Rheumatol ; 82(6): 479-490, 2023 Aug.
Artículo en Alemán | MEDLINE | ID: mdl-37488246

RESUMEN

BACKGROUND: Energy is the currency of life. The systemic and intracellular energy metabolism plays an essential role for the energy supply of the resting and activated immune system and this also applies to chronic inflammatory diseases. OBJECTIVE: This presentation examines both components of the systemic and cellular energy metabolism in health and chronic inflammation. MATERIAL AND METHODS: A literature search was conducted using PubMed, Embase and the Cochrane Library. The information is presented in the form of a narrative review. RESULTS: A chronically activated immune system acquires large amounts of energy-rich substrates that are lost for other functions of the body. In particular, the immune system and the brain are in competition. The consequences of this competition are many known diseases, such as fatigue, anxiety, depression, anorexia, sleep problems, sarcopenia, osteoporosis, insulin resistance, hypertension and others. The permanent change in the brain causes long-term alterations that stimulate disease sequelae even after disease remission. In the intracellular energy supply, chronic inflammation typically involves a conversion to glycolysis (to lactate, which has its own regulatory functions) and the pentose phosphate pathway in disorders of mitochondrial function. The chronic changes in immune cells of patients with rheumatoid arthritis (RA) lead to a disruption of the citric acid cycle (Krebs cycle). The hypoxic situation in the inflamed tissue stimulates many alterations. A differentiation is made between effector functions and regulatory functions of immune cells. CONCLUSION: Based on the energy changes mentioned, novel treatment suggestions can be made in addition to those already known in energy metabolism.


Asunto(s)
Artritis Reumatoide , Inflamación , Humanos , Sistema Inmunológico , Enfermedad Crónica , Metabolismo Energético/fisiología
3.
Metab Eng ; 72: 116-132, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35296430

RESUMEN

Osteoarthritis (OA) is the most common degenerative joint disease and a major cause of age-related disability worldwide, mainly due to pain, the disease's main symptom. Although OA was initially classified as a non-inflammatory joint disease, recent attention has been drawn to the importance of synovitis and fibroblast-like synoviocytes (FLS) in the pathogenesis of OA. FLS can be divided into two major populations: thymus cell antigen 1 (THY1)- FLS are currently classified as quiescent cells and assumed to destroy bone and cartilage, whereas THY1+ FLS are invasively proliferative cells that drive synovitis. Both THY1- and THY1+ FLS share many characteristics with fibroblast-like progenitors - mesenchymal stromal cells (MSC). However, it remains unclear whether synovitis-induced metabolic changes exist in FLS from OA patients and whether metabolic differences may provide a mechanistic basis for the identification of approaches to precisely convert the pathologically proliferative synovitis-driven FLS phenotype into a healthy one. To identify novel pathological mechanisms of the perpetuation and manifestation of OA, we analyzed metabolic, proteomic, and functional characteristics of THY1+ FLS from patients with OA. Proteome data and pathway analysis revealed that an elevated expression of pyruvate dehydrogenase kinase (PDK) 3 was characteristic of proliferative THY1+ FLS from patients with OA. These FLS also had the highest podoplanin (PDPN) expression and localized to the sublining but also the lining layer in OA synovium in contrast to the synovium of ligament trauma patients. Inhibition of PDKs reprogrammed metabolism from glycolysis towards oxidative phosphorylation and reduced FLS proliferation and inflammatory cytokine secretion. This study provides new mechanistic insights into the importance of FLS metabolism in the pathogenesis of OA. Given the selective overexpression of PDK3 in OA synovium and its restricted distribution in synovial tissue from ligament trauma patients and MSC, PDKs may represent attractive selective metabolic targets for OA treatment. Moreover, targeting PDKs does not affect cells in a homeostatic, oxidative state. Our data provide an evidence-based rationale for the idea that inhibition of PDKs could restore the healthy THY1+ FLS phenotype. This approach may mitigate the progression of OA and thereby fundamentally change the clinical management of OA from the treatment of symptoms to addressing causes.


Asunto(s)
Osteoartritis , Sinovitis , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Osteoartritis/genética , Osteoartritis/metabolismo , Osteoartritis/patología , Oxidorreductasas/metabolismo , Proteómica , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Piruvatos/metabolismo , Sinovitis/metabolismo , Sinovitis/patología
4.
FASEB J ; 35(7): e21684, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34159634

RESUMEN

Adenine nucleotides represent crucial immunomodulators in the extracellular environment. The ectonucleotidases CD39 and CD73 are responsible for the sequential catabolism of ATP to adenosine via AMP, thus promoting an anti-inflammatory milieu induced by the "adenosine halo". AMPD2 intracellularly mediates AMP deamination to IMP, thereby both enhancing the degradation of inflammatory ATP and reducing the formation of anti-inflammatory adenosine. Here, we show that this enzyme is expressed on the surface of human immune cells and its predominance may modify inflammatory states by altering the extracellular milieu. Surface AMPD2 (eAMPD2) expression on monocytes was verified by immunoblot, surface biotinylation, mass spectrometry, and immunofluorescence microscopy. Flow cytometry revealed enhanced monocytic eAMPD2 expression after TLR stimulation. PBMCs from patients with rheumatoid arthritis displayed significantly higher levels of eAMPD2 expression compared with healthy controls. Furthermore, the product of AMPD2-IMP-exerted anti-inflammatory effects, while the levels of extracellular adenosine were not impaired by an increased eAMPD2 expression. In summary, our study identifies eAMPD2 as a novel regulator of the extracellular ATP-adenosine balance adding to the immunomodulatory CD39-CD73 system.


Asunto(s)
5'-Nucleotidasa/metabolismo , AMP Desaminasa/metabolismo , Adenina/metabolismo , Adenosina Trifosfato/metabolismo , Leucocitos/metabolismo , Apirasa , Células Cultivadas , Proteínas Ligadas a GPI/metabolismo , Humanos
5.
Int J Mol Sci ; 23(9)2022 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-35563440

RESUMEN

Unspecific antibody binding takes a significant toll on researchers in the form of both the economic burden and the disappointed hopes of promising new therapeutic targets. Despite recent initiatives promoting antibody validation, a uniform approach addressing this issue has not yet been developed. Here, we demonstrate that the anti-glucocorticoid receptor (GR) antibody clone 5E4 predominantly targets two different proteins of approximately the same size, namely AMP deaminase 2 (AMPD2) and transcription intermediary factor 1-beta (TRIM28). This paper is intended to generate awareness of unspecific binding of well-established reagents and advocate the use of more rigorous verification methods to improve antibody quality in the future.


Asunto(s)
Receptores de Glucocorticoides , Factores de Transcripción , Células Cultivadas , Células Clonales/metabolismo , Receptores de Glucocorticoides/metabolismo , Factores de Transcripción/metabolismo
6.
Int J Mol Sci ; 23(19)2022 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-36232895

RESUMEN

The mechanisms of mast cell (MC) degranulation and MC-driven skin symptoms are well-described. In contrast, data about the role of mitochondrial respiration for immune functions of human skin MCs are lacking. Oxygen consumption rate (OCR) in primary human skin MCs during IgE-mediated activation in the absence of glucose was examined using a metabolic flux analyzer. Effects of the inhibition of mitochondrial complex I (by rotenone A) and III (by myxothiazol) on degranulation and cytokine secretion (IL-4, IL-5, IL-6, IL-13, TNF-α, and GM-CSF) were explored by the ß-hexosaminidase release assay and multiplex ELISA. IgE-mediated activation rapidly increased the mitochondrial OCR and extracellular acidification; the contribution of non-mitochondrial oxygen consumption remained unchanged at lower levels. Both myxothiazol and rotenone A reduced OCR, the mitochondrial parameters, and extracellular acidification; however, myxothiazol did not affect degranulation and cytokine secretion. In contrast, degranulation and the secretion of IL-6, IL-13, TNF-α, and GM-CSF were reduced by rotenone A, whereas the secretion of IL-4 and IL-5 was not significantly affected. The inhibitors did not affect cell viability. Our results highlight the important role played by mitochondrial respiration in primary human skin MCs and allow for a conclusion on a hierarchy of their effector functions. Drugs targeting specific pathways in mitochondria may provide future options to control MC-driven skin symptoms.


Asunto(s)
Degranulación de la Célula , Mastocitos , Transporte de Electrón , Complejo I de Transporte de Electrón/metabolismo , Glucosa/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inmunoglobulina E , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Mastocitos/metabolismo , Metacrilatos , Rotenona/metabolismo , Rotenona/farmacología , Tiazoles , Factor de Necrosis Tumoral alfa/metabolismo , beta-N-Acetilhexosaminidasas/metabolismo
7.
Z Rheumatol ; 81(8): 652-659, 2022 Oct.
Artículo en Alemán | MEDLINE | ID: mdl-35412048

RESUMEN

Rheumatoid arthritis and osteoarthritis are two related chronic diseases of the musculoskeletal system which are particularly pronounced in the region of joints and bones. Their pathogeneses are associated with chronic inflammation, which can disrupt homeostasis in bones and articular cartilage. Degradation products deriving from articular cartilage can contribute to the exacerbation of inflammation in the joint region. Mechanical stimuli and blood vessels also play a central role in both the regulation of bone growth as well as in the regeneration of bone tissue. Not only chronic inflammatory processes but also hormonal changes after menopause or undesired effects of glucocorticoid therapy have an influence on the balance between bone resorption and deposition, by promoting the former and reducing the latter. This results in decreased bone quality and, in some cases, considerable loss of bone or osteoporosis. An in-depth understanding of these processes at the molecular, cellular, and tissue level, as well as of the changes present in chronic inflammatory diseases, has been the focus of research at the German Rheumatism Research Center (Deutsches Rheuma-Forschungszentrum, DRFZ) since its foundation. Based on an improved understanding of these mechanisms, the DRFZ aims to develop improved prevention and treatment strategies with effects even in early disease stages.


Asunto(s)
Cartílago Articular , Osteoartritis , Femenino , Glucocorticoides , Humanos , Inflamación , Células del Estroma
8.
Int J Mol Sci ; 21(21)2020 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-33113770

RESUMEN

Rheumatoid arthritis (RA) is a chronic, inflammatory, and systemic autoimmune disease that affects the connective tissue and primarily the joints. If not treated, RA ultimately leads to progressive cartilage and bone degeneration. The etiology of the pathogenesis of RA is unknown, demonstrating heterogeneity in its clinical presentation, and is associated with autoantibodies directed against modified self-epitopes. Although many models already exist for RA for preclinical research, many current model systems of arthritis have limited predictive value because they are either based on animals of phylogenetically distant origin or suffer from overly simplified in vitro culture conditions. These limitations pose considerable challenges for preclinical research and therefore clinical translation. Thus, a sophisticated experimental human-based in vitro approach mimicking RA is essential to (i) investigate key mechanisms in the pathogenesis of human RA, (ii) identify targets for new therapeutic approaches, (iii) test these approaches, (iv) facilitate the clinical transferability of results, and (v) reduce the use of laboratory animals. Here, we summarize the most commonly used in vitro models of RA and discuss their experimental feasibility and physiological proximity to the pathophysiology of human RA to highlight new human-based avenues in RA research to increase our knowledge on human pathophysiology and develop effective targeted therapies.


Asunto(s)
Artritis Reumatoide/inmunología , Autoanticuerpos/inmunología , Cartílago/inmunología , Inflamación/inmunología , Membrana Sinovial/inmunología , Animales , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Huesos/inmunología , Huesos/metabolismo , Huesos/patología , Cartílago/metabolismo , Cartílago/patología , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamación/metabolismo , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
9.
Int J Mol Sci ; 21(23)2020 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-33256266

RESUMEN

The Janus kinase (JAK) signal transducer and activator of transcription (STAT) signaling pathway serves as an important downstream mediator for a variety of cytokines, hormones, and growth factors. Emerging evidence suggests JAK/STAT signaling pathway plays an important role in bone development, metabolism, and healing. In this light, pro-inflammatory cytokines are now clearly implicated in these processes as they can perturb normal bone remodeling through their action on osteoclasts and osteoblasts at both intra- and extra-articular skeletal sites. Here, we summarize the role of JAK/STAT pathway on development, homeostasis, and regeneration based on skeletal phenotype of individual JAK and STAT gene knockout models and selective inhibition of components of the JAK/STAT signaling including influences of JAK inhibition in osteoclasts, osteoblasts, and osteocytes.


Asunto(s)
Desarrollo Óseo , Homeostasis , Quinasas Janus/metabolismo , Regeneración , Factores de Transcripción STAT/metabolismo , Animales , Remodelación Ósea , Humanos
10.
Int J Mol Sci ; 22(1)2020 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-33374446

RESUMEN

Adequate tissue engineered models are required to further understand the (patho)physiological mechanism involved in the destructive processes of cartilage and subchondral bone during rheumatoid arthritis (RA). Therefore, we developed a human in vitro 3D osteochondral tissue model (OTM), mimicking cytokine-induced cellular and matrix-related changes leading to cartilage degradation and bone destruction in order to ultimately provide a preclinical drug screening tool. To this end, the OTM was engineered by co-cultivation of mesenchymal stromal cell (MSC)-derived bone and cartilage components in a 3D environment. It was comprehensively characterized on cell, protein, and mRNA level. Stimulating the OTM with pro-inflammatory cytokines, relevant in RA (tumor necrosis factor α, interleukin-6, macrophage migration inhibitory factor), caused cell- and matrix-related changes, resulting in a significantly induced gene expression of lactate dehydrogenase A, interleukin-8 and tumor necrosis factor α in both, cartilage and bone, while the matrix metalloproteases 1 and 3 were only induced in cartilage. Finally, application of target-specific drugs prevented the induction of inflammation and matrix-degradation. Thus, we here provide evidence that our human in vitro 3D OTM mimics cytokine-induced cell- and matrix-related changes-key features of RA-and may serve as a preclinical tool for the evaluation of both new targets and potential drugs in a more translational setup.


Asunto(s)
Artritis Reumatoide/metabolismo , Cartílago Articular/patología , Citocinas/metabolismo , Anciano , Huesos/metabolismo , Fosfatos de Calcio/metabolismo , Condrocitos/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Técnicas In Vitro , Interleucina-6/biosíntesis , Interleucina-8/biosíntesis , Lactato Deshidrogenasa 5/biosíntesis , Factores Inhibidores de la Migración de Macrófagos/biosíntesis , Masculino , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Membrana Sinovial/patología , Ingeniería de Tejidos/métodos , Investigación Biomédica Traslacional , Factor de Necrosis Tumoral alfa/biosíntesis
11.
Int J Mol Sci ; 21(3)2020 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-32013232

RESUMEN

Both inflammatory diseases like rheumatoid arthritis (RA) and anti-inflammatory treatment of RA with glucocorticoids (GCs) or non-steroidal anti-inflammatory drugs (NSAIDs) negatively influence bone metabolism and fracture healing. Janus kinase (JAK) inhibition with tofacitinib has been demonstrated to act as a potent anti-inflammatory therapeutic agent in the treatment of RA, but its impact on the fundamental processes of bone regeneration is currently controversially discussed and at least in part elusive. Therefore, in this study, we aimed to examine the effects of tofacitinib on processes of bone healing focusing on recruitment of human mesenchymal stromal cells (hMSCs) into the inflammatory microenvironment of the fracture gap, chondrogenesis, osteogenesis and osteoclastogenesis. We performed our analyses under conditions of reduced oxygen availability in order to mimic the in vivo situation of the fracture gap most optimal. We demonstrate that tofacitinib dose-dependently promotes the recruitment of hMSCs under hypoxia but inhibits recruitment of hMSCs under normoxia. With regard to the chondrogenic differentiation of hMSCs, we demonstrate that tofacitinib does not inhibit survival at therapeutically relevant doses of 10-100 nM. Moreover, tofacitinib dose-dependently enhances osteogenic differentiation of hMSCs and reduces osteoclast differentiation and activity. We conclude from our data that tofacitinib may influence bone healing by promotion of hMSC recruitment into the hypoxic microenvironment of the fracture gap but does not interfere with the cartilaginous phase of the soft callus phase of fracture healing process. We assume that tofacitinib may promote bone formation and reduce bone resorption, which could in part explain the positive impact of tofacitinib on bone erosions in RA. Thus, we hypothesize that it will be unnecessary to stop this medication in case of fracture and suggest that positive effects on osteoporosis are likely.


Asunto(s)
Inhibidores de las Cinasas Janus/farmacología , Quinasas Janus/metabolismo , Osteogénesis/efectos de los fármacos , Piperidinas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Condrogénesis/efectos de los fármacos , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Quinasas Janus/antagonistas & inhibidores , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo
12.
Microvasc Res ; 116: 34-44, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28993199

RESUMEN

Hypoxia driven angiogenesis is a prominent feature of tissue regeneration, inflammation and tumor growth and is regulated by hypoxia-inducible factor (HIF)-1 and -2. The distinct functions of HIFs in the hypoxia-induced angiogenesis and metabolic switch of endothelial cells are still unknown and therefore aim of this study. We investigated the role of HIF-1 and -2 in the adaptation of immortalized human microvascular endothelial cells (HMEC-1) to hypoxic conditions (1% O2) in terms of angiogenesis, cytokine secretion, gene expression and ATP/ADP-ratio using shRNA-mediated reduction of the oxygen sensitive α-subunits of either HIF-1 or HIF-2 or the combination of both. Reduction of HIF-1α diminished cellular energy, hypoxia-induced glycolytic gene expression, and angiogenesis not altering pro-angiogenic factors. Reduction of HIF-2α diminished hypoxia-induced pro-angiogenic factors, enhanced anti-angiogenic factors and attenuated angiogenesis not altering glycolytic gene expression. Reduction of both HIFs reduced cell survival, gene expression of glycolytic enzymes and pro-angiogenic factors as compared to the corresponding control. Finally, we identified the macrophage migration inhibitory factor (MIF) to be redundantly regulated by HIF-1 and HIF-2 and to be essential in the process of hypoxia-driven angiogenesis. Our results demonstrate a major impact of HIF-1 and HIF-2 on hypoxia-induced angiogenesis indicating distinct but also overlapping functions of HIF-1 and HIF-2. These findings open new possibilities for therapeutic approaches by specifically targeting the HIF-1 and HIF-2 or their target MIF.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Endoteliales/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Microvasos/metabolismo , Neovascularización Fisiológica , Adaptación Fisiológica , Antígenos de Diferenciación de Linfocitos B/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hipoxia de la Célula , Línea Celular , Microambiente Celular , Regulación Enzimológica de la Expresión Génica , Glucólisis , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Microvasos/citología , Neovascularización Fisiológica/genética , Transducción de Señal
13.
Int J Mol Sci ; 19(8)2018 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-30087255

RESUMEN

Mesenchymal stem/stromal cells (MSCs) are stem cells of the connective tissue, possess a plastic phenotype, and are able to differentiate into various tissues. Besides their role in tissue regeneration, MSCs perform additional functions as a modulator or inhibitor of immune responses. Due to their pleiotropic function, MSCs have also gained therapeutic importance for the treatment of autoimmune diseases and for improving fracture healing and cartilage regeneration. However, the therapeutic/immunomodulatory mode of action of MSCs is largely unknown. Here, we describe that MSCs express the inhibitory receptor CTLA-4 (cytotoxic T lymphocyte antigen 4). We show that depending on the environmental conditions, MSCs express different isoforms of CTLA-4 with the secreted isoform (sCTLA-4) being the most abundant under hypoxic conditions. Furthermore, we demonstrate that the immunosuppressive function of MSCs is mediated mainly by the secretion of CTLA-4. These findings open new ways for treatment when tissue regeneration/fracture healing is difficult.


Asunto(s)
Antígeno CTLA-4/inmunología , Células Madre Mesenquimatosas/inmunología , Adipogénesis , Antígeno CTLA-4/análisis , Células Cultivadas , Humanos , Tolerancia Inmunológica , Células Madre Mesenquimatosas/citología , Osteogénesis
14.
Int J Mol Sci ; 18(3)2017 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-28282868

RESUMEN

Immunologically restricted patients such as those with autoimmune diseases or malignancies often suffer from delayed or insufficient fracture healing. In human fracture hematomas and the surrounding bone marrow obtained from immunologically restricted patients, we analyzed the initial inflammatory phase on cellular and humoral level via flow cytometry and multiplex suspension array. Compared with controls, we demonstrated higher numbers of immune cells like monocytes/macrophages, natural killer T (NKT) cells, and activated T helper cells within the fracture hematomas and/or the surrounding bone marrow. Also, several pro-inflammatory cytokines such as Interleukin (IL)-6 and Tumor necrosis factor α (TNFα), chemokines (e.g., Eotaxin and RANTES), pro-angiogenic factors (e.g., IL-8 and Macrophage migration inhibitory factor: MIF), and regulatory cytokines (e.g., IL-10) were found at higher levels within the fracture hematomas and/or the surrounding bone marrow of immunologically restricted patients when compared to controls. We conclude here that the inflammatory activity on cellular and humoral levels at fracture sites of immunologically restricted patients considerably exceeds that of control patients. The initial inflammatory phase profoundly differs between these patient groups and is probably one of the reasons for prolonged or insufficient fracture healing often occurring within immunologically restricted patients.


Asunto(s)
Curación de Fractura/inmunología , Huésped Inmunocomprometido , Inflamación/inmunología , Inductores de la Angiogénesis/metabolismo , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/patología , Estudios de Casos y Controles , Citocinas/metabolismo , Femenino , Fracturas Óseas/inmunología , Fracturas Óseas/patología , Hematoma/inmunología , Hematoma/patología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Leucocitos/patología , Masculino , Neovascularización Fisiológica , Fenotipo
15.
Clin Exp Rheumatol ; 34(5): 848-856, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27385076

RESUMEN

OBJECTIVES: To compare sleep quality, disease activity and patient-reported outcomes such as fatigue and immune parameters in patients with rheumatoid arthritis treated with etanercept (ETA) or methotrexate (MTX). METHODS: Of 36 patients (28-joint Disease Activity Score, DAS28CRP≥3.2) in this 16-week (w), open, prospective study, 19 (11 women) received MTX 12.5-17 mg/w, and 17 (14 women) received ETA 25 mg x 2/w, alone or in combination with MTX. Clinical (DAS28CRP, visual analogue scale), laboratory (C-reactive protein [CRP]), sleep (polysomnography), functional (Multidimensional Fatigue Inventory; Health Assessment Questionnaire-Disability Index (HAQ-DI); 36-item Short-Form Health Survey (SF-36), immunological (humoral/cellular) and neuroendocrine (hormonal) parameters were recorded at baseline (BL), w8 and w16. RESULTS: BL characteristics did not differ significantly between the ETA and MTX groups except disease duration: mean age (years): 48.6±8.8 vs. 49.4±16.6; mean disease duration (months): 19.6±46.3 vs. 81.2±79.2; and DAS28CRP: 4.4±0.9 vs. 4.4±1.7, respectively. DAS28CRP, SF-36, and HAQ-DI improved significantly in both groups from BL to w16 (p≤0.05). The DAS28CRP improvements at w16 (mean changes -1.8 in the ETA group, and -1.4 in MTX group), were not statistically significant from each other. The absolute values of sleep efficiency, total sleep time, and stage 2 sleep duration increased significantly in the ETA group, but no significant changes were reported in the MTX group. CONCLUSIONS: Both therapies improved disease activity, CRP, SF-36 and HAQ-DI, with faster, more pronounced changes in DAS28CRP in the ETA group, which alone had significantly improved sleep parameters.


Asunto(s)
Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Etanercept/uso terapéutico , Fatiga/prevención & control , Metotrexato/uso terapéutico , Trastornos del Sueño-Vigilia/prevención & control , Sueño/efectos de los fármacos , Adolescente , Adulto , Anciano , Antirreumáticos/efectos adversos , Artritis Reumatoide/complicaciones , Artritis Reumatoide/inmunología , Artritis Reumatoide/fisiopatología , Evaluación de la Discapacidad , Quimioterapia Combinada , Etanercept/efectos adversos , Fatiga/inmunología , Fatiga/fisiopatología , Femenino , Humanos , Masculino , Metotrexato/efectos adversos , Persona de Mediana Edad , Medición de Resultados Informados por el Paciente , Proyectos Piloto , Polisomnografía , Valor Predictivo de las Pruebas , Estudios Prospectivos , Inducción de Remisión , Trastornos del Sueño-Vigilia/inmunología , Trastornos del Sueño-Vigilia/fisiopatología , Encuestas y Cuestionarios , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
16.
Clin Exp Rheumatol ; 33(1): 34-43, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25535886

RESUMEN

OBJECTIVES: The circadian rhythm of clinical symptoms in rheumatoid arthritis (RA) has been primarily attributed to circadian variations in humoral factors and hormones. In this study, we investigated circadian rhythms of cellular immunity in RA (CiRA study). METHODS: Peripheral blood of female postmenopausal patients with active RA (DAS 28 ≥ 4.2) (n=5) and female postmenopausal non-RA controls (n=5) was collected every 2 hours for 24 hours and analysed by flow cytometry, cytokine multiplex suspension array and quantitative RT-PCR of clock gene expression in isolated CD14+ monocytes. Endogenous circadian rhythms of macrophages were investigated by BMAL1-luciferase bioluminescence. Significance of circadian rhythms was tested by Cosinor analysis. RESULTS: We found (i) circadian rhythms in the relative frequency of peripheral blood cell populations that were present in postmenopausal non-RA controls but absent in patients with active RA, (ii) circadian rhythms that were absent in non-RA controls but present in patients with RA and (iii) circadian rhythms that were present in both groups but with differences in peak phase or amplitude or amplitude/magnitude. The circadian rhythm in expression of the clock genes PER2 and PER3 in CD14+ monocytes was lost in patients with RA. The amplitude of BMAL1-luciferase bioluminescence tended to be lower in patients with RA than in non-RA controls. CONCLUSIONS: We conclude that (i) in RA some immune cell populations lose their normal circadian rhythms whereas others establish new 'inflammatory' circadian rhythms and (ii) these findings provide a good basis for further identifying pathophysiological aspects of RA chronobiology with potential therapeutic implications.


Asunto(s)
Artritis Reumatoide/inmunología , Ritmo Circadiano , Inmunidad Celular , Macrófagos/inmunología , Monocitos/inmunología , Adaptación Fisiológica , Anciano , Artritis Reumatoide/sangre , Artritis Reumatoide/genética , Artritis Reumatoide/fisiopatología , Biomarcadores/sangre , Estudios de Casos y Controles , Ritmo Circadiano/genética , Citocinas/sangre , Femenino , Humanos , Inmunidad Humoral , Mediadores de Inflamación/sangre , Receptores de Lipopolisacáridos/sangre , Macrófagos/metabolismo , Persona de Mediana Edad , Monocitos/metabolismo , Proteínas Circadianas Period/genética , Proyectos Piloto , Posmenopausia/inmunología , ARN Mensajero/sangre , Factores de Tiempo
17.
Eur J Immunol ; 43(6): 1588-97, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23519896

RESUMEN

Inflamed areas are characterized by infiltration of immune cells, local hypoxia and alterations of cellular redox states. We investigated the impact of hypoxia on survival, proliferation, cytokine secretion, intracellular energy and redox state of human CD4(+) T cells. We found that pathophysiological hypoxia (<2% O2 ) significantly decreased CD4(+) T-cell survival after mitogenic stimulation. This effect was not due to an increased caspase-3/7-mediated apoptosis or adenosine-5'-triphosphate (ATP) consumption/depletion. However, the ability of stimulated T cells to proliferate was reduced under hypoxic conditions, despite increased expression of CD25. Pathophysiological hypoxia was also found to modify intracellular ROS (iROS) levels in stimulated T cells over time as compared with levels found in normoxia. Physiological hypoxia (5% O2 ) did not decrease CD4(+) T-cell survival and proliferation or modify iROS levels as compared with normoxia. We conclude that pathophysiological hypoxia affects T-cell proliferation and viability via disturbed IL-2R signalling downstream of STAT5a phosphorylation, but not as a result of impaired cellular energy homeostasis. We suggest iROS links early events in T-cell stimulation to the inhibition of the lymphoproliferative response under pathophysiological hypoxic conditions. The level of iROS may therefore act as a mediator of immune functions leading to down-regulation of long-term T-cell activity in inflamed tissues.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Hipoxia de la Célula/inmunología , Supervivencia Celular , Células Cultivadas , Humanos , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal
18.
J Gene Med ; 16(11-12): 352-63, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25382123

RESUMEN

BACKGROUND: Gene therapy appears to have the potential for achieving a long-term remedy for osteoarthritis (OA). However, there is a risk of adverse reactions, especially when using cytomegalovirus-controlled expression. To provide a safe application, we focused on the expression of therapeutic cytokines [e.g. interleukin (IL)-4] in a disease-responsive manner by use of the previously cloned Cox-2 promoter as 'genetic switch'. In the present study, we report the functionality of a controlled gene therapeutic system in an equine osteoarthritic cell model. METHODS: Different nonviral transfection reagents were tested for their efficiency on equine chondrocytes stimulated with equine IL-1ß or lipopolysaccharide to create an inflammatory environment. To optimize the transfection, we successfully redesigned the vector by excluding the internal ribosomal entry site (IRES). The functionality of our Cox-2 promoter construct with respect to expressing IL-4 was proven at the mRNA and protein levels and the anti-inflammatory potential of IL-4 was confirmed by analyzing the expression of IL-1ß, IL-6, IL-8, matrix metalloproteinase (MMP)-1, MMP-3 and tumor necrosis factor (TNF)-α using a quantitative polymerase chain reaction. RESULTS: Nonviral transfection reagents yielded transfection rates from 21% to 44% with control vectors with and without IRES, respectively. Stimulation of equine chondrocytes resulted in a 20-fold increase of mRNA expression of IL-1ß. Such exogenous stimulation of chondrocytes transfected with pNCox2-IL4 led to an increase of IL-4 mRNA expression, whereas expression of inflammatory mediators decreased. The timely link between these events confirms the anti-inflammatory potential of synthesized IL-4. CONCLUSIONS: We consider that this approach has significant potential for translation into a useful anti-inflammation therapy. Molecular tools such as the described therapeutic plasmid pave the way for a local-controlled, self-limiting gene therapy.


Asunto(s)
Ciclooxigenasa 2/genética , Terapia Genética , Interleucina-4/biosíntesis , Osteoartritis/terapia , Transfección , Animales , Células Cultivadas , Condrocitos/inmunología , Condrocitos/metabolismo , Regulación hacia Abajo , Expresión Génica , Vectores Genéticos , Caballos , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-4/genética , Lipopolisacáridos/farmacología , Osteoartritis/genética , Regiones Promotoras Genéticas
19.
BMC Musculoskelet Disord ; 15: 31, 2014 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-24491163

RESUMEN

BACKGROUND: The role of endogenous glucocorticoids (GC) in the initiation and maintenance of rheumatoid arthritis (RA) remains unclear. We demonstrated previously that disruption of GC signalling in osteoblasts results in a profound attenuation of K/BxN serum-induced arthritis, a mouse model of RA. To determine whether or not the modulation of the inflammatory response by osteoblasts involves T cells, we studied the effects of disrupted osteoblastic GC-signalling in the T cell-dependent model of antigen-induced arthritis (AIA). METHODS: Acute arthritis was induced in pre-immunised 11-week-old male 11ß-hydroxysteroid dehydrogenase type 2 transgenic (tg) mice and their wild-type (WT) littermates by intra-articular injection of methylated bovine serum albumine (mBSA) into one knee joint. Knee diameter was measured every 1-2 days until euthanasia on day 14 post injection. In a separate experiment, arthritis was maintained for 28 days by weekly reinjections of mBSA. Tissues were analysed by histology, histomorphometry and microfocal-computed tomography. Serum cytokines levels were determined by multiplex suspension array. RESULTS: In both short and long term experiments, arthritis developed in tg and WT mice with no significant difference between both groups. Histological indices of inflammation, cartilage damage and bone erosion were similar in tg and WT mice. Bone volume and turnover at the contralateral tibia and systemic cytokine levels were not different. CONCLUSIONS: Acute murine AIA is not affected by a disruption in osteoblastic GC signalling. These data indicate that osteoblasts do not modulate the T cell-mediated inflammatory response via a GC-dependent pathway.


Asunto(s)
Artritis Experimental/metabolismo , Glucocorticoides/metabolismo , Articulación de la Rodilla/metabolismo , Osteoblastos/metabolismo , Transducción de Señal , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/diagnóstico , Artritis Experimental/enzimología , Artritis Experimental/genética , Artritis Experimental/inmunología , Citocinas/sangre , Mediadores de Inflamación/sangre , Articulación de la Rodilla/diagnóstico por imagen , Articulación de la Rodilla/enzimología , Articulación de la Rodilla/inmunología , Masculino , Ratones , Ratones Transgénicos , Osteoblastos/enzimología , Osteoblastos/inmunología , Albúmina Sérica Bovina , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo , Microtomografía por Rayos X
20.
Front Immunol ; 15: 1385006, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38895122

RESUMEN

Osteoarthritis (OA) is the most common form of arthritis, characterized by osteophyte formation, cartilage degradation, and structural and cellular alterations of the synovial membrane. Activated fibroblast-like synoviocytes (FLS) of the synovial membrane have been identified as key drivers, secreting humoral mediators that maintain inflammatory processes, proteases that cause cartilage and bone destruction, and factors that drive fibrotic processes. In normal tissue repair, fibrotic processes are terminated after the damage has been repaired. In fibrosis, tissue remodeling and wound healing are exaggerated and prolonged. Various stressors, including aging, joint instability, and inflammation, lead to structural damage of the joint and micro lesions within the synovial tissue. One result is the reduced production of synovial fluid (lubricants), which reduces the lubricity of the cartilage areas, leading to cartilage damage. In the synovial tissue, a wound-healing cascade is initiated by activating macrophages, Th2 cells, and FLS. The latter can be divided into two major populations. The destructive thymocyte differentiation antigen (THY)1─ phenotype is restricted to the synovial lining layer. In contrast, the THY1+ phenotype of the sublining layer is classified as an invasive one with immune effector function driving synovitis. The exact mechanisms involved in the transition of fibroblasts into a myofibroblast-like phenotype that drives fibrosis remain unclear. The review provides an overview of the phenotypes and spatial distribution of FLS in the synovial membrane of OA, describes the mechanisms of fibroblast into myofibroblast activation, and the metabolic alterations of myofibroblast-like cells.


Asunto(s)
Fibroblastos , Fibrosis , Osteoartritis , Fenotipo , Sinoviocitos , Humanos , Osteoartritis/patología , Osteoartritis/inmunología , Osteoartritis/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/inmunología , Animales , Sinoviocitos/metabolismo , Sinoviocitos/patología , Sinoviocitos/inmunología , Membrana Sinovial/patología , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA