Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Int J Mol Sci ; 25(5)2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38474000

RESUMEN

Gouty arthritis results from monosodium urate (MSU) crystal deposition in joints, initiating (pro)-interleukin (IL)-1ß maturation, inflammatory mediator release, and neutrophil infiltration, leading to joint swelling and pain. Parathyroid hormone-related protein (107-111) C-terminal peptide (osteostatin) has shown anti-inflammatory properties in osteoblasts and collagen-induced arthritis in mice, but its impact in gouty arthritis models remains unexplored. We investigated the effect of osteostatin on pyroptosis, inflammation, and oxidation in macrophages, as well as its role in the formation of calcium pyrophosphate dihydrate crystals and MSU-induced gouty arthritis in mice models. Osteostatin ameliorated pyroptosis induced by lipopolysaccharide and adenosine 5'-triphosphate (LPS + ATP) in mice peritoneal macrophages by reducing the expression of caspase-1, lactate dehydrogenase release, and IL-1ß and IL-18 secretion. Additionally, IL-6 and tumor necrosis factor-α (TNF-α) were also decreased due to the reduced activation of the NF-κB pathway. Furthermore, osteostatin displayed antioxidant properties in LPS + ATP-stimulated macrophages, resulting in reduced production of mitochondrial and extracellular reactive oxygen species and enhanced Nrf2 translocation to the nuclei. In both models of gouty arthritis, osteostatin administration resulted in reduced pro-inflammatory cytokine production, decreased leukocyte migration, and reduced caspase-1 and NF-κB activation. These results highlight the potential of osteostatin as a therapeutic option for gouty arthritis.


Asunto(s)
Artritis Gotosa , Proteína Relacionada con la Hormona Paratiroidea , Fragmentos de Péptidos , Ratones , Animales , Artritis Gotosa/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Regulación hacia Arriba , Lipopolisacáridos/efectos adversos , Ácido Úrico , Inflamación/metabolismo , Adenosina Trifosfato , Caspasas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
2.
Stem Cells Transl Med ; 11(12): 1177-1185, 2022 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-36318277

RESUMEN

Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes progressive joint destruction. Despite the advances in the treatment of this condition there remains a clinical need for safe therapies leading to clinical remission. Mesenchymal stem/stromal cells (MSCs) play immunomodulatory and regenerative roles which can be partly mediated by their secretome. In recent years, the important contribution of extracellular vesicles (EVs) to MSC actions has received an increasing interest as a new therapeutic approach. We provide an extensive overview of the immunomodulatory properties of MSC EVs and their effects on articular cells such as fibroblast-like synoviocytes that play a central role in joint destruction. This review discusses the anti-arthritic effects of MSC EVs in vitro and in animal models of RA as well as their potential mechanisms. Recent preclinical data suggest that transfer of non-coding RNAs by MSC EVs regulates key signaling pathways involved in the pathogenesis of RA. We also examine a number of EV modifications for improving their anti-arthritic efficacy and carrier ability for drug delivery.


Asunto(s)
Artritis Reumatoide , Vesículas Extracelulares , Células Madre Mesenquimatosas , Animales , Artritis Reumatoide/terapia , Vesículas Extracelulares/metabolismo , Células Cultivadas , Transducción de Señal , Células Madre Mesenquimatosas/metabolismo
3.
Int J Mol Sci ; 23(15)2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35955685

RESUMEN

Parathyroid hormone-related protein (PTHrP) C-terminal peptides regulate the metabolism of bone cells. PHTrP [107-111] (osteostatin) promotes bone repair in animal models of bone defects and prevents bone erosion in inflammatory arthritis. In addition to its positive effects on osteoblasts, osteostatin may inhibit bone resorption. The aim of this study was to determine the effects of osteostatin on human osteoclast differentiation and function. We used macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor κB ligand (RANKL) to induce the osteoclast differentiation of adherent human peripheral blood mononuclear cells. Tartrate-resistant acid phosphatase (TRAP) staining was performed for the detection of the osteoclasts. The function of mature osteoclasts was assessed with a pit resorption assay. Gene expression was evaluated with qRT-PCR, and nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) nuclear translocation was studied by immunofluorescence. We observed that osteostatin (100, 250 and 500 nM) decreased the differentiation of osteoclasts in a concentration-dependent manner, but it did not modify the resorptive ability of mature osteoclasts. In addition, osteostatin decreased the mRNA levels of cathepsin K, osteoclast associated Ig-like receptor (OSCAR) and NFATc1. The nuclear translocation of the master transcription factor in osteoclast differentiation NFATc1 was reduced by osteostatin. Our results suggest that the anti-resorptive effects of osteostatin may be dependent on the inhibition of osteoclastogenesis. This study has shown that osteostatin controls human osteoclast differentiation in vitro through the downregulation of NFATc1.


Asunto(s)
Resorción Ósea , Ligando RANK , Animales , Resorción Ósea/metabolismo , Diferenciación Celular , Humanos , Leucocitos Mononucleares/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Factores de Transcripción NFATC/metabolismo , Osteoclastos/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Fragmentos de Péptidos , Ligando RANK/metabolismo , Ligando RANK/farmacología
4.
Clin Chem Lab Med ; 59(9): 1592-1599, 2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-33908223

RESUMEN

OBJECTIVES: The aim of the present study was to validate a commercially available automated assay for the measurement of total adenosine deaminase (tADA) and its isoenzymes (ADA1 and ADA2) in saliva in a fast and accurate way, and evaluate the possible changes of these analytes in individuals with SARS-CoV-2 infection. METHODS: The validation, in addition to the evaluation of precision and accuracy, included the analysis of the effects of the main procedures that are currently being used for SARS-CoV-2 inactivation in saliva and a pilot study to evaluate the possible changes in salivary tADA and isoenzymes in individuals infected with SARS-CoV-2. RESULTS: The automated assay proved to be accurate and precise, with intra- and inter-assay coefficients of variation below 8.2%, linearity under dilution linear regression with R2 close to 1, and recovery percentage between 80 and 120% in all cases. This assay was affected when the sample is treated with heat or SDS for virus inactivation but tolerated Triton X-100 and NP-40. Individuals with SARS-CoV-2 infection (n=71) and who recovered from infection (n=11) had higher mean values of activity of tADA and its isoenzymes than healthy individuals (n=35). CONCLUSIONS: tADA and its isoenzymes ADA1 and ADA2 can be measured accurately and precisely in saliva samples in a rapid, economical, and reproducible way and can be analyzed after chemical inactivation with Triton X-100 and NP-40. Besides, the changes observed in tADA and isoenzymes in individuals with COVID-19 open the possibility of their potential use as non-invasive biomarkers in this disease.


Asunto(s)
Adenosina Desaminasa/metabolismo , Bioensayo/métodos , Biomarcadores/metabolismo , COVID-19/diagnóstico , SARS-CoV-2/enzimología , Saliva/enzimología , Adulto , COVID-19/virología , Estudios de Casos y Controles , Femenino , Humanos , Isoenzimas , Masculino , Persona de Mediana Edad , Proyectos Piloto , Adulto Joven
5.
Salud ment ; 44(2): 83-90, Mar.-Apr. 2021. tab, graf
Artículo en Inglés | LILACS-Express | LILACS | ID: biblio-1252154

RESUMEN

Abstract Introduction Premenstrual syndrome/premenstrual dysphoric disorder (PMS/PMDD) may be neuropsychologically understood as impairments of executive functions (EF), since these are related to the regulation of complex behavior and cognition. Objective To test the utility of self-report of EF versus performance-based measures, for the understanding of PMS/PMDD, and to analyze interactive effects between symptoms of these pathologies and EF on daily-life functionality. Method Mexican women were recruited through non-probabilistic procedures. The Premenstrual Symptoms Screening Tool (PSST) was used to determine severity of symptoms and functional impairment in daily-life activities, and the Behavioral Rating Inventory of Executive Functions-Adults (BRIEF-A) (short Spanish-translated version) adapted to collect information on EF during luteal versus follicular phases. Performance was evaluated with Stroop, Trail Making Test and Letter-Number Sequencing. Results A total of 157 were analyzed. Three groups were formed: No diagnosis ( n = 78); PMS ( n = 67) and PMDD ( n = 12). Between-group differences were observed for both BRIEF-A-Luteal and BRIEF-A-Follicular. Bivariate correlations between these measures and the PSST were found, with double the magnitude relative to BRIEF-A-Luteal. Only two indicators of performance-based measures were weakly associated to the PSST. The regression model showed high multicollinearity between self-reported EF and PMS/PMDD symptoms, and no interaction was found. Discussion and conclusion Self-report probed a better association than based-performance tests for the assessment of EF in PMS/PMDD. EF deficits and PMS/PMDD symptoms, particularly during luteal phase, may be as closely link as to allow for the consideration of these diagnoses as partial forms of dysexecutive syndrome.


Resumen Introducción El síndrome premenstrual/trastorno disfórico premenstrual (SPM/TDPM) pueden entenderse neuropsicológicamente como alteraciones de las funciones ejecutivas (FE), ya que éstas permiten la regulación del comportamiento complejo y la cognición. Objetivo Evaluar la utilidad del autorreporte de las FE versus pruebas de desempeño para comprender el SPM/TDPM, y analizar los efectos interactivos entre los síntomas de estas patologías y las FE sobre el funcionamiento diario. Método Mujeres mexicanas fueron reclutadas por medio de procedimientos no probabilísticos. El Instrumento de Detección de Síntomas Premenstruales (PSST) se utilizó para determinar la gravedad de los síntomas y el deterioro funcional en las actividades de la vida diaria, y el Inventario de Evaluación Conductual de la Función Ejecutiva-Adultos (BRIEF-A) (versión breve traducida al español) para recopilar información sobre EF durante las fases lútea versus folicular. Se emplearon también las pruebas de desempeño: Stroop, Trail Making Test y Secuencia de Letras y Números. Resultados Se analizó un total de 157 participantes. Se formaron tres grupos: sin diagnóstico ( n = 78); SPM ( n = 67) y TDPM ( n = 12). Se observaron diferencias entre los grupos para BRIEF-A-Lútea y BRIEF-A-Folicular. Se encontraron correlaciones bivariadas entre estas medidas y el PSST, con el doble de magnitud en relación con BRIEF-A-Lútea. Solo dos indicadores de medidas basadas en el desempeño mostraron una asociación débil con el PSST. El modelo de regresión mostró alta multicolinealidad entre el autorreporte de FE y SPM/TDPM, y no se encontró la interacción esperada. Discusión y conclusión El autorreporte mostró una mejor asociación que las pruebas de rendimiento para la evaluación de FE en SPM/TDPM. Los déficits de EF y los síntomas de SPM/TDPM, particularmente durante la fase lútea, pueden estar tan estrechamente vinculados como para permitir la consideración de estos diagnósticos como formas parciales de síndrome disejecutivo.

6.
Methods Mol Biol ; 2269: 221-231, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33687682

RESUMEN

Adipose tissue-derived mesenchymal stem cells (AD-MSCs) offer great therapeutic potential for osteoarthritis (OA) treatment. Recent investigations have revealed the contribution of extracellular vesicles (EVs) to AD-MSC actions. Here, we describe a method to study the in vitro effects of EVs from AD-MSCs in OA chondrocytes. This chapter includes the isolation and analysis of human AD-MSCs and their EVs as well as the isolation and treatment of OA chondrocytes.


Asunto(s)
Tejido Adiposo/metabolismo , Condrocitos/metabolismo , Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoartritis/metabolismo , Tejido Adiposo/patología , Separación Celular , Condrocitos/patología , Técnicas de Cocultivo , Humanos , Células Madre Mesenquimatosas/patología , Osteoartritis/patología
7.
Int J Mol Sci ; 22(3)2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33573086

RESUMEN

Adipose tissue represents an abundant source of mesenchymal stem cells (MSC) for therapeutic purposes. Previous studies have demonstrated the anti-inflammatory potential of adipose tissue-derived MSC (ASC). Extracellular vesicles (EV) present in the conditioned medium (CM) have been shown to mediate the cytoprotective effects of human ASC secretome. Nevertheless, the role of EV in the anti-inflammatory effects of mouse-derived ASC is not known. The current study has investigated the influence of mouse-derived ASC CM and its fractions on the response of mouse-derived peritoneal macrophages against lipopolysaccharide (LPS). CM and its soluble fraction reduced the release of pro-inflammatory cytokines, adenosine triphosphate and nitric oxide in stimulated cells. They also enhanced the migration of neutrophils or monocytes, in the absence or presence of LPS, respectively, which is likely related to the presence of chemokines, and reduced the phagocytic response. The anti-inflammatory effect of CM may be dependent on the regulation of toll-like receptor 4 expression and nuclear factor-κB activation. Our results demonstrate the anti-inflammatory effects of mouse-derived ASC secretome in mouse-derived peritoneal macrophages stimulated with LPS and show that they are not mediated by EV.


Asunto(s)
Vesículas Extracelulares/metabolismo , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Células Cultivadas , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Ratones
8.
Int J Mol Sci ; 20(16)2019 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-31394717

RESUMEN

In chronic inflammatory joint diseases, such as rheumatoid arthritis, there is an important bone loss. Parathyroid hormone-related protein (PTHrP) and related peptides have shown osteoinductive properties in bone regeneration models, but there are no data on inflammatory joint destruction. We have investigated whether the PTHrP (107-111) C-terminal peptide (osteostatin) could control the development of collagen-induced arthritis in mice. Administration of osteostatin (80 or 120 µg/kg s.c.) after the onset of disease decreased the severity of arthritis as well as cartilage and bone degradation. This peptide reduced serum IgG2a levels as well as T cell activation, with the downregulation of RORγt+CD4+ T cells and upregulation of FoxP3+CD8+ T cells in lymph nodes. The levels of key cytokines, such as interleukin(IL)-1ß, IL-2, IL-6, IL-17, and tumor necrosis factor-α in mice paws were decreased by osteostatin treatment, whereas IL-10 was enhanced. Bone protection was related to reductions in receptor activator of nuclear factor-κB ligand, Dickkopf-related protein 1, and joint osteoclast area. Osteostatin improves arthritis and controls bone loss by inhibiting immune activation, pro-inflammatory cytokines, and osteoclastogenesis. Our results support the interest of osteostatin for the treatment of inflammatory joint conditions.


Asunto(s)
Artritis Experimental/etiología , Artritis Experimental/metabolismo , Citocinas/metabolismo , Susceptibilidad a Enfermedades/inmunología , Mediadores de Inflamación/metabolismo , Osteogénesis , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Fragmentos de Péptidos/farmacología , Animales , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/patología , Biomarcadores , Biopsia , Huesos/metabolismo , Huesos/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inmunoglobulina G/inmunología , Masculino , Ratones , Peroxidasa/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
9.
Biochem Pharmacol ; 165: 4-16, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30826327

RESUMEN

Osteoarthritis (OA) is the most common joint disorder and a leading cause of disability. Current treatments for OA can improve symptoms but do not delay the progression of disease. In the last years, much effort has been devoted to developing new treatments for OA focused on pain control, inflammatory mediators or degradation of articular tissues. Although promising results have been obtained in ex vivo studies and animal models of OA, few of these agents have completed clinical trials. Available clinical data support the interest of nerve growth factor as a target in pain control as well as the disease-modifying potential of inhibitors of Wnt signaling or catabolic enzymes such as aggrecanases and cathepsin K, and anabolic strategies like fibroblast growth factor-18 or cellular therapies. Carefully controlled studies in patients selected according to OA phenotypes and with a long follow-up will help to confirm the relevance of these new approaches as emerging therapeutic treatments in OA.


Asunto(s)
Osteoartritis/tratamiento farmacológico , Animales , Citocinas/fisiología , Humanos , Mediadores de Inflamación/fisiología , Canales Iónicos/fisiología , Trasplante de Células Madre Mesenquimatosas , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Factor de Crecimiento Nervioso/fisiología , Transducción de Señal/fisiología , Vía de Señalización Wnt/fisiología
10.
Cells ; 9(1)2019 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-31906087

RESUMEN

Mesenchymal stem/stromal cells (MSCs) represent a promising therapy for musculoskeletal diseases. There is compelling evidence indicating that MSC effects are mainly mediated by paracrine mechanisms and in particular by the secretion of extracellular vesicles (EVs). Many studies have thus suggested that EVs may be an alternative to cell therapy with MSCs in tissue repair. In this review, we summarize the current understanding of MSC EVs actions in preclinical studies of (1) immune regulation and rheumatoid arthritis, (2) bone repair and bone diseases, (3) cartilage repair and osteoarthritis, (4) intervertebral disk degeneration and (5) skeletal muscle and tendon repair. We also discuss the mechanisms underlying these actions and the perspectives of MSC EVs-based strategies for future treatments of musculoskeletal disorders.


Asunto(s)
Terapia Biológica , Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Enfermedades Musculoesqueléticas/metabolismo , Enfermedades Musculoesqueléticas/terapia , Animales , Terapia Biológica/métodos , Manejo de la Enfermedad , Humanos , Inmunomodulación , Trasplante de Células Madre Mesenquimatosas , Enfermedades Musculoesqueléticas/diagnóstico , Enfermedades Musculoesqueléticas/etiología , Regeneración
11.
Cell Physiol Biochem ; 47(1): 11-25, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29763932

RESUMEN

BACKGROUND/AIMS: Chronic inflammation contributes to cartilage degeneration during the progression of osteoarthritis (OA). Adipose tissue-derived mesenchymal stem cells (AD-MSC) show great potential to treat inflammatory and degradative processes in OA and have demonstrated paracrine effects in chondrocytes. In the present work, we have isolated and characterized the extracellular vesicles from human AD-MSC to investigate their role in the chondroprotective actions of these cells. METHODS: AD-MSC were isolated by collagenase treatment from adipose tissue from healthy individuals subjected to abdominal lipectomy surgery. Microvesicles and exosomes were obtained from conditioned medium by filtration and differential centrifugation. Chondrocytes from OA patients were used in primary culture and stimulated with 10 ng/ml interleukin(IL)-1ß in the presence or absence of AD-MSC microvesicles, exosomes or conditioned medium. Protein expression was investigated by ELISA and immunofluorescence, transcription factor-DNA binding by ELISA, gene expression by real-time PCR, prostaglandin E2 (PGE2) by radioimmunoassay, and matrix metalloproteinase (MMP) activity and nitric oxide (NO) production by fluorometry. RESULTS: In OA chondrocytes stimulated with IL-1ß, microvesicles and exosomes reduced the production of inflammatory mediators tumor necrosis factor-α, IL-6, PGE2 and NO. The downregulation of cyclooxygenase-2 and microsomal prostaglandin E synthase-1 would lead to the decreased PGE2 production while the effect on NO could depend on the reduction of inducible nitric oxide synthase expression. Treatment of OA chondrocytes with extracellular vesicles also decreased the release of MMP activity and MMP-13 expression whereas the production of the anti-inflammatory cytokine IL-10 and the expression of collagen II were significantly enhanced. The reduction of inflammatory and catabolic mediators could be the consequence of a lower activation of nuclear factor-κB and activator protein-1. The upregulation of annexin A1 specially in MV may contribute to the anti-inflammatory and chondroprotective effects of AD-MSC. CONCLUSIONS: Our data support the interest of AD-MSC extracellular vesicles to develop new therapeutic approaches in joint conditions.


Asunto(s)
Condrocitos/inmunología , Vesículas Extracelulares/inmunología , Células Madre Mesenquimatosas/inmunología , Osteoartritis/terapia , Tejido Adiposo/citología , Anciano , Supervivencia Celular , Células Cultivadas , Condrocitos/citología , Condrocitos/patología , Citocinas/inmunología , Dinoprostona/inmunología , Femenino , Humanos , Masculino , Metaloproteinasas de la Matriz/inmunología , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Óxido Nítrico/inmunología , Osteoartritis/inmunología , Osteoartritis/patología
12.
Pharmacol Rev ; 70(2): 348-383, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29507103

RESUMEN

Systems medicine has a mechanism-based rather than a symptom- or organ-based approach to disease and identifies therapeutic targets in a nonhypothesis-driven manner. In this work, we apply this to transcription factor nuclear factor (erythroid-derived 2)-like 2 (NRF2) by cross-validating its position in a protein-protein interaction network (the NRF2 interactome) functionally linked to cytoprotection in low-grade stress, chronic inflammation, metabolic alterations, and reactive oxygen species formation. Multiscale network analysis of these molecular profiles suggests alterations of NRF2 expression and activity as a common mechanism in a subnetwork of diseases (the NRF2 diseasome). This network joins apparently heterogeneous phenotypes such as autoimmune, respiratory, digestive, cardiovascular, metabolic, and neurodegenerative diseases, along with cancer. Importantly, this approach matches and confirms in silico several applications for NRF2-modulating drugs validated in vivo at different phases of clinical development. Pharmacologically, their profile is as diverse as electrophilic dimethyl fumarate, synthetic triterpenoids like bardoxolone methyl and sulforaphane, protein-protein or DNA-protein interaction inhibitors, and even registered drugs such as metformin and statins, which activate NRF2 and may be repurposed for indications within the NRF2 cluster of disease phenotypes. Thus, NRF2 represents one of the first targets fully embraced by classic and systems medicine approaches to facilitate both drug development and drug repurposing by focusing on a set of disease phenotypes that appear to be mechanistically linked. The resulting NRF2 drugome may therefore rapidly advance several surprising clinical options for this subset of chronic diseases.


Asunto(s)
Enfermedad Crónica/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Factor 2 Relacionado con NF-E2/metabolismo , Análisis de Sistemas , Animales , Antiinflamatorios/uso terapéutico , Descubrimiento de Drogas , Reposicionamiento de Medicamentos , Humanos , Factor 2 Relacionado con NF-E2/genética
13.
Oxid Med Cell Longev ; 2018: 5053091, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29599896

RESUMEN

Heme oxygenase-1 (HO-1) is induced by many stimuli to modulate the activation and function of different cell types during innate immune responses. Although HO-1 has shown anti-inflammatory effects in different systems, there are few data on the contribution of myeloid HO-1 and its role in inflammatory processes is not well understood. To address this point, we have used HO-1M-KO mice with myeloid-restricted deletion of HO-1 to specifically investigate its influence on the acute inflammatory response to zymosan in vivo. In the mouse air pouch model, we have shown an exacerbated inflammation in HO-1M-KO mice with increased neutrophil infiltration accompanied by high levels of inflammatory mediators such as interleukin-1ß, tumor necrosis factor-α, and prostaglandin E2. The expression of the degradative enzyme matrix metalloproteinase-3 (MMP-3) was also enhanced. In addition, we observed higher levels of serum MMP-3 in HO-1M-KO mice compared with control mice, suggesting the presence of systemic inflammation. Altogether, these findings demonstrate that myeloid HO-1 plays an anti-inflammatory role in the acute response to zymosan in vivo and suggest the interest of this target to regulate inflammatory processes.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Inflamación/enzimología , Proteínas de la Membrana/metabolismo , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/inmunología , Inflamación/inducido químicamente , Inflamación/inmunología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Masculino , Metaloproteinasa 3 de la Matriz/sangre , Metaloproteinasa 3 de la Matriz/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Neutrófilos/enzimología , Neutrófilos/inmunología , Zimosan/toxicidad
14.
Biochem Pharmacol ; 153: 134-146, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29427625

RESUMEN

Extracellular vesicles (EVs) are attracting increasing interest since they might represent a more convenient therapeutic tool with respect to their cells of origin. In the last years much time and effort have been expended to determine the biological properties of EVs from mesenchymal stem cells (MSCs) and other sources. The immunoregulatory, anti-inflammatory and regenerative properties of MSC EVs have been demonstrated in in vitro studies and animal models of rheumatoid arthritis or osteoarthritis. This cell-free approach has been proposed as a possible better alternative to MSC therapy in autoimmune conditions and tissue regeneration. In addition, EVs show great potential as biomarkers of disease or delivery systems for active molecules. The standardization of isolation and characterization methods is a key step for the development of EV research. A better understanding of EV mechanisms of action and efficacy is required to establish the potential therapeutic applications of this new approach in joint conditions.


Asunto(s)
Vesículas Extracelulares/trasplante , Artropatías/terapia , Células Madre Mesenquimatosas , Animales , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Artritis Reumatoide/terapia , Vesículas Extracelulares/metabolismo , Humanos , Inmunidad Celular/fisiología , Artropatías/inmunología , Artropatías/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Trasplante de Células Madre Mesenquimatosas/tendencias , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Osteoartritis/inmunología , Osteoartritis/metabolismo , Osteoartritis/terapia
15.
Oxid Med Cell Longev ; 2017: 7197598, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29230269

RESUMEN

Osteoarthritis (OA) affects all articular tissues leading to pain and disability. The dysregulation of bone metabolism may contribute to the progression of this condition. Adipose-derived mesenchymal stem cells (ASC) are attractive candidates in the search of novel strategies for OA treatment and exert anti-inflammatory and cytoprotective effects on cartilage. Chronic inflammation in OA is a relevant factor in the development of cellular senescence and joint degradation. In this study, we extend our previous observations of ASC paracrine effects to study the influence of conditioned medium and extracellular vesicles from ASC on senescence induced by inflammatory stress in OA osteoblasts. Our results in cells stimulated with interleukin- (IL-) 1ß indicate that conditioned medium, microvesicles, and exosomes from ASC downregulate senescence-associated ß-galactosidase activity and the accumulation of γH2AX foci. In addition, they reduced the production of inflammatory mediators, with the highest effect on IL-6 and prostaglandin E2. The control of mitochondrial membrane alterations and oxidative stress may provide a mechanism for the protective effects of ASC in OA osteoblasts. We have also shown that microvesicles and exosomes mediate the paracrine effects of ASC. Our study suggests that correction of abnormal osteoblast metabolism by ASC products may contribute to their protective effects.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoartritis/genética , Osteoblastos/metabolismo , Regulación hacia Abajo , Humanos , Osteoartritis/metabolismo
16.
J Control Release ; 248: 60-70, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28069553

RESUMEN

Acute inflammation is a protective response of the body to harmful stimuli, such as pathogens or damaged cells. However, dysregulated inflammation can cause secondary damage and could thus contribute to the pathophysiology of many diseases. Inflammasomes, the macromolecular complexes responsible for caspase-1 activation, have emerged as key regulators of immune and inflammatory responses. Therefore, modulation of inflammasome activity has become an important therapeutic approach. Here we describe the design of a smart nanodevice that takes advantage of the passive targeting of nanoparticles to macrophages and enhances the therapeutic effect of caspase-1 inhibitor VX-765 in vivo. The functional hybrid systems consisted of MCM-41-based nanoparticles loaded with anti-inflammatory drug VX-765 (S2-P) and capped with poly-L-lysine, which acts as a molecular gate. S2-P activity has been evaluated in cellular and in vivo models of inflammation. The results indicated the potential advantage of using nanodevices to treat inflammatory diseases.


Asunto(s)
Antiinflamatorios/administración & dosificación , Inhibidores de Caspasas/administración & dosificación , Dipéptidos/administración & dosificación , Portadores de Fármacos/química , Inflamasomas/antagonistas & inhibidores , Nanopartículas/química , Dióxido de Silicio/química , para-Aminobenzoatos/administración & dosificación , Animales , Antiinflamatorios/farmacología , Inhibidores de Caspasas/farmacología , Línea Celular , Preparaciones de Acción Retardada/química , Dipéptidos/farmacología , Humanos , Inflamasomas/inmunología , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Nanopartículas/ultraestructura , para-Aminobenzoatos/farmacología
17.
J Gerontol A Biol Sci Med Sci ; 72(5): 624-631, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-27271252

RESUMEN

Osteoarthritis (OA) is characterized by degenerative changes in the whole joint leading to physical disability in the elderly population. This condition is associated with altered bone metabolism in subchondral areas suggesting that therapeutic strategies aimed at modifying bone cell metabolism may be of interest. We have investigated the effects of several parathyroid hormone-related protein (PTHrP)-derived peptides (1-37): (N-terminal), (107-111) and (107-139) (C-terminal) on senescence features induced by inflammatory stress in human OA osteoblasts. Incubation of these primary cells with interleukin(IL)-1ß led to an increased expression of senescence markers senescence-associated-ß-galactosidase activity, γH2AX foci, p16, p21, p53, and caveolin-1. PTHrP (107-111) and PTHrP (107-139) significantly reduced all these parameters. Both peptides decreased the production of IL-6 and prostaglandin E2 which was the consequence of cyclo-oxygenase-2 downregulation. PTHrP (107-139) also reduced tumor necrosis factor-α release. These anti-inflammatory effects would be related to the reduction of nuclear factor-κB activation by both peptides and activator protein-1 by PTHrP (107-139). The three PTHrP peptides favored osteoblastic function although the C-terminal domain of PTHrP was more efficient than its N-terminal domain. Our data support an anti-senescence and anti-inflammatory role for the C-terminal moiety of PTHrP with potential applications in chronic inflammatory conditions such as OA.


Asunto(s)
Senescencia Celular/fisiología , Osteoartritis/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Anciano , Células Cultivadas , Dinoprostona/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Mediadores de Inflamación/farmacología , Interleucina-1beta/farmacología , Interleucina-6/metabolismo , Masculino , Osteoartritis/prevención & control , Osteoblastos/citología , Fragmentos de Péptidos/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/metabolismo
18.
Aging (Albany NY) ; 8(8): 1703-17, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27490266

RESUMEN

Aging and exposure to stress would determine the chondrocyte phenotype in osteoarthritis (OA). In particular, chronic inflammation may contribute to stress-induced senescence of chondrocytes and cartilage degeneration during OA progression. Recent studies have shown that adipose-derived mesenchymal stem cells exert paracrine effects protecting against degenerative changes in chondrocytes. We have investigated whether the conditioned medium (CM) from adipose-derived mesenchymal stem cells may regulate senescence features induced by inflammatory stress in OA chondrocytes. Our results indicate that CM down-regulated senescence markers induced by interleukin-1ß including senescence-associated ß-galactosidase activity, accumulation of γH2AX foci and morphological changes with enhanced formation of actin stress fibers. Treatment of chondrocytes with CM also decreased the production of oxidative stress, the activation of mitogen-activated protein kinases, and the expression of caveolin-1 and p21. The effects of CM were related to the reduction in p53 acetylation which would be dependent on the enhancement of Sirtuin 1 expression. Therefore, CM may exert protective effects in degenerative joint conditions by countering the premature senescence of OA chondrocytes induced by inflammatory stress.


Asunto(s)
Tejido Adiposo/metabolismo , Senescencia Celular/fisiología , Condrocitos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoartritis/metabolismo , Comunicación Paracrina/fisiología , Tejido Adiposo/patología , Caveolina 1/metabolismo , Condrocitos/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Células Madre Mesenquimatosas/patología , Osteoartritis/patología , Estrés Oxidativo/fisiología , beta-Galactosidasa/metabolismo
19.
Biomed Pharmacother ; 79: 120-8, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27044820

RESUMEN

CONTEXT: The efficacy of the combination chondroitin sulfate-glucosamine (CS-GlcN) in the treatment of knee osteoarthritis (OA) has been suggested in recent clinical studies. In vitro reports have also suggested anti-inflammatory and anti-resorptive effects of this combination. OBJECTIVE: The aim of this study was to characterize the effects of CS-GlcN on joint degradation in vivo including the assessment of inflammation and bone metabolism in a model of OA. MATERIALS AND METHODS: We have used the OA model induced by anterior cruciate ligament transection (ACLT) in ovariectomised rats. CS-GlcN was administered daily (oral gavage) from week 0 until week 12 after ovariectomy at the dose of 140 (CS)+175 (GlcN)(HCl) mg/kg. Histochemical analyses were performed, the levels of biomarkers and inflammatory mediators were measured by luminex or ELISA and bone microstructure was determined by µCT. RESULTS: CS-GlcN protected against cartilage degradation and reduced the levels of inflammatory mediators such as interleukin-1ß and tumor necrosis factor-α in the affected knee. In addition, serum biomarkers of inflammation and cartilage and bone degradation including matrix metalloproteinase-3, C-telopeptide of type II collagen and the ratio receptor activator of nuclear factor κB ligand/osteoprotegerin were significantly decreased by CS-GlcN. This treatment also tended to improve some bone microstructural parameters without reaching statistical significance. DISCUSSION AND CONCLUSIONS: These results demonstrate the chondroprotective effects of CS-GlcN in vivo, in the experimental model of ACLT in ovariectomised rats, and suggest that this combination may be useful to control the joint catabolic effects of inflammatory stress. These findings could have clinical relevance related to the prevention of joint degradation by CS-GlcN and support the potential development of OA treatments based on this combination.


Asunto(s)
Lesiones del Ligamento Cruzado Anterior/tratamiento farmacológico , Ligamento Cruzado Anterior/patología , Cartílago Articular/patología , Sulfatos de Condroitina/uso terapéutico , Glucosamina/uso terapéutico , Osteoartritis de la Rodilla/tratamiento farmacológico , Sustancias Protectoras/uso terapéutico , Animales , Ligamento Cruzado Anterior/efectos de los fármacos , Lesiones del Ligamento Cruzado Anterior/patología , Biomarcadores/sangre , Huesos/efectos de los fármacos , Huesos/patología , Cartílago Articular/efectos de los fármacos , Sulfatos de Condroitina/farmacología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Glucosamina/farmacología , Mediadores de Inflamación/metabolismo , Articulaciones/efectos de los fármacos , Articulaciones/patología , Osteoartritis de la Rodilla/sangre , Osteoartritis de la Rodilla/patología , Ovariectomía , Sustancias Protectoras/farmacología , Ratas Wistar , Microtomografía por Rayos X
20.
Cytotherapy ; 17(9): 1230-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26276006

RESUMEN

BACKGROUND AIMS: Excessive or unresolved inflammation leads to tissue lesions. Adipose tissue-derived mesenchymal stromal cells (AMSCs) have shown protective effects that may be dependent on the modulation of inflammation by secreted factors. METHODS: We used the zymosan-induced mouse air pouch model at two time points (4 h and 18 h) to evaluate the in vivo effects of AMSCs and their conditioned medium (CM) on key steps of the early inflammatory response. We assessed the effects of AMSCs and CM on leukocyte migration and myeloperoxidase activity. The levels of chemokines, cytokines and eicosanoids in exudates were measured by use of enzyme-linked immunoassay or radio-immunoassay. In addition, the expression of cyclooxygenase-2 and microsomal prostaglandin E synthase-1 (mPGES-1) was studied by use of Western blotting and the phosphorylation of p65 nuclear factor-κB (NF-κB) by immunofluorescence. RESULTS: All inflammatory parameters were significantly reduced by CM and AMSCs to a similar extent at 4 h after zymosan injection with lower effects at 18 h. The observed inhibition of leukocyte migration was associated with reduced levels of chemokines and leukotriene B4. Interleukin-1ß, interleukin-6, tumor necrosis factor-α and tumor necrosis factor-stimulated gene 6 levels were significantly decreased. The downregulation of mPGES-1 was associated with inhibition of prostaglandin E2 production. Our results suggest that these anti-inflammatory effects are related, in part, to the inhibition of NF-κB activation. CONCLUSIONS: AMSCs dampen the early process of inflammation in the zymosan-induced mouse air pouch model through paracrine mechanisms. These results support the potential utility of these cells as a source of novel treatment approaches for inflammatory pathologies.


Asunto(s)
Tejido Adiposo/citología , Dinoprostona/antagonistas & inhibidores , Inflamación/inmunología , Células Madre Mesenquimatosas/metabolismo , Comunicación Paracrina , Factor de Transcripción ReIA/antagonistas & inhibidores , Tejido Adiposo/metabolismo , Animales , Movimiento Celular , Medios de Cultivo Condicionados/farmacología , Ciclooxigenasa 2/biosíntesis , Citocinas/metabolismo , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Oxidorreductasas Intramoleculares/biosíntesis , Leucocitos/fisiología , Leucotrieno B4/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratones , Prostaglandina-E Sintasas , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Zimosan/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA