Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 35(5): e21506, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33811695

RESUMEN

Purinergic signaling regulates several renal physiological and pathophysiological processes. Extracellular vesicles (EVs) are nanoparticles released by most cell types, which, in non-renal tissues, modulate purinergic signaling. The aim of this study was to investigate the effect of EVs from renal proximal tubule (HK2) and collecting duct cells (HCD) on intra- and intersegment modulation of extracellular ATP levels, the underlying molecular mechanisms, and the impact on the expression of the alpha subunit of the epithelial sodium channel (αENaC). HK2 cells were exposed to HK2 EVs, while HCD cells were exposed to HK2 and HCD EVs. Extracellular ATP levels and αENaC expression were measured by chemiluminescence and qRT-PCR, respectively. ATPases in EV populations were identified by mass spectrometry. The effect of aldosterone was assessed using EVs from aldosterone-treated cells and urinary EVs (uEVs) from primary aldosteronism (PA) patients. HK2 EVs downregulated ectonucleoside-triphosphate-diphosphohydrolase-1 (ENTPD1) expression, increased extracellular ATP and downregulated αENaC expression in HCD cells. ENTPD1 downregulation could be attributed to increased miR-205-3p and miR-505 levels. Conversely, HCD EVs decreased extracellular ATP levels and upregulated αENaC expression in HCD cells, probably due to enrichment of 14-3-3 isoforms with ATPase activity. Pretreatment of donor cells with aldosterone or exposure to uEVs from PA patients enhanced the effects on extracellular ATP and αENaC expression. We demonstrated inter- and intrasegment modulation of renal purinergic signaling by EVs. Our findings postulate EVs as carriers of information along the renal tubules, whereby processes affecting EV release and/or cargo may impact on purinergically regulated processes.


Asunto(s)
Adenosina Trifosfato/metabolismo , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Vesículas Extracelulares/fisiología , Regulación de la Expresión Génica , Hiperaldosteronismo/patología , Túbulos Renales/metabolismo , Células Epiteliales/citología , Canales Epiteliales de Sodio/genética , Humanos , Hiperaldosteronismo/metabolismo , Túbulos Renales/citología
2.
J Am Soc Nephrol ; 32(5): 1210-1226, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33782168

RESUMEN

BACKGROUND: Urinary extracellular vesicles (uEVs) are a promising source for biomarker discovery, but optimal approaches for normalization, quantification, and characterization in spot urines are unclear. METHODS: Urine samples were analyzed in a water-loading study, from healthy subjects and patients with kidney disease. Urine particles were quantified in whole urine using nanoparticle tracking analysis (NTA), time-resolved fluorescence immunoassay (TR-FIA), and EVQuant, a novel method quantifying particles via gel immobilization. RESULTS: Urine particle and creatinine concentrations were highly correlated in the water-loading study (R2 0.96) and in random spot urines from healthy subjects (R2 0.47-0.95) and patients (R2 0.41-0.81). Water loading reduced aquaporin-2 but increased Tamm-Horsfall protein (THP) and particle detection by NTA. This finding was attributed to hypotonicity increasing uEV size (more EVs reach the NTA size detection limit) and reducing THP polymerization. Adding THP to urine also significantly increased particle count by NTA. In both fluorescence NTA and EVQuant, adding 0.01% SDS maintained uEV integrity and increased aquaporin-2 detection. Comparison of intracellular- and extracellular-epitope antibodies suggested the presence of reverse topology uEVs. The exosome markers CD9 and CD63 colocalized and immunoprecipitated selectively with distal nephron markers. Conclusions uEV concentration is highly correlated with urine creatinine, potentially replacing the need for uEV quantification to normalize spot urines. Additional findings relevant for future uEV studies in whole urine include the interference of THP with NTA, excretion of larger uEVs in dilute urine, the ability to use detergent to increase intracellular-epitope recognition in uEVs, and CD9 or CD63 capture of nephron segment-specific EVs.


Asunto(s)
Vesículas Extracelulares/metabolismo , Enfermedades Renales/diagnóstico , Enfermedades Renales/orina , Adulto , Biomarcadores/orina , Estudios de Casos y Controles , Creatinina/orina , Femenino , Humanos , Masculino , Reproducibilidad de los Resultados , Urinálisis
3.
Int J Mol Sci ; 22(7)2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33805981

RESUMEN

Mesenchymal stem cells have an important potential in the treatment of age-related diseases. In the last years, small extracellular vesicles derived from these stem cells have been proposed as cell-free therapies. Cellular senescence and proinflammatory activation are involved in the loss of therapeutic capacity and in the phenomenon called inflamm-aging. The regulators of these two biological processes in mesenchymal stem cells are not well-known. In this study, we found that p65 is activated during cellular senescence and inflammatory activation in human umbilical cord-derived mesenchymal stem cell. To demonstrate the central role of p65 in these two processes, we used small-molecular inhibitors of p65, such as JSH-23, MG-132 and curcumin. We found that the inhibition of p65 prevents the cellular senescence phenotype in human umbilical cord-derived mesenchymal stem cells. Besides, p65 inhibition produced the inactivation of proinflammatory molecules as components of a senescence-associated secretory phenotype (SASP) (interleukin-6 and interleukin-8 (IL-6 and IL-8)). Additionally, we found that the inhibition of p65 prevents the transmission of paracrine senescence between mesenchymal stem cells and the proinflammatory message through small extracellular vesicles. Our work highlights the important role of p65 and its inhibition to restore the loss of functionality of small extracellular vesicles from senescent mesenchymal stem cells and their inflamm-aging signature.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Factor de Transcripción ReIA/metabolismo , Adolescente , Adulto , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Curcumina/farmacología , Daño del ADN , Femenino , Humanos , Inflamación , Leupeptinas/farmacología , Nanopartículas , Comunicación Paracrina/efectos de los fármacos , Fenotipo , Fenilendiaminas/farmacología , Cordón Umbilical/citología
4.
J Cell Physiol ; 232(1): 225-33, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27138291

RESUMEN

The general consensus is that milk promotes bone growth and density because is a source of calcium and contains components that enhance intestinal calcium uptake or directly affect bone metabolism. In this study, we investigated the effect of bovine-derived milk 100,000 g pellet (P100), which contains nanoparticles (<220 nm) including extracellular vesicles, on osteoclast differentiation and bone resorption. Bone marrow-derived osteoclast precursor cells were differentiated into osteoclasts by M-CSF and RANKL (control) and in the presence of milk P100. Milk P100 treatment until day 4 increased the number of TRAP-positive mononuclear cells and small (≤5 nuclei) osteoclasts. The number of large (≥6 nuclei) osteoclasts remained the same. These alterations were associated with increased expression of TRAP, NFATc1, and c-Fos. Cells seeded in a calcium-phosphate coated plate or bone slices showed reduced resorption area when exposed to milk P100 during the differentiation phase and even after osteoclast formation. Interestingly, milk P100 treatment enhanced Cathepsin K expression but reduced Carbonic Anhydrase 2 gene expression. Moreover, intracellular acid production was also decreased by milk P100 treatment. Oral delivery of milk P100 to female DBA1/J mice for 7 weeks did not alter bone area; however, increased osteoclast number and area in tibia without changes in serum RANKL and CTX-I levels. We showed for the first time the effect of milk P100 on osteoclast differentiation both in vitro and in vivo and found that milk P100 increased the formation of small osteoclasts but this does not lead to more bone resorption probably due to reduced acid secretion. J. Cell. Physiol. 232: 225-233, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Leche/metabolismo , Nanopartículas/administración & dosificación , Osteoclastos/metabolismo , Animales , Resorción Ósea/metabolismo , Fosfatos de Calcio/farmacología , Diferenciación Celular/fisiología , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Osteoclastos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Ann Rheum Dis ; 74(11): 2084-91, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25028707

RESUMEN

OBJECTIVES: Rheumatoid arthritis is a chronic destructive autoimmune disease, but the course is unpredictable in individual patients. An attractive treatment would provide a disease-regulated therapy that offers personalised drug delivery. Therefore, we expressed the anti-inflammatory interleukin-10 (IL-10) gene under the control of inflammation-dependent promoters in a mouse model of arthritis. METHODS: Proximal promoters of S100a8, Cxcl1, Mmp13, Saa3, IL-1b and Tsg6 were selected by whole-genome expression analysis of inflamed synovial tissues from arthritic mice. Mice were injected intraarticularly in knee joints with lentiviral vectors expressing a luciferase reporter or the therapeutic protein IL-10 under control of the Saa3 or Mmp13 promoter. After 4 days, arthritis was induced by intraarticular injection of streptococcal cell walls (SCW). At different time points after arthritis induction, in vivo bioluminescent imaging was performed and knee joints were dissected for histological and RNA analysis. RESULTS: The disease-regulated promoter-luciferase reporter constructs showed different activation profiles during the course of the disease. The Saa3 and Mmp13 promoters were significantly induced at day 1 or day 4 after arthritis induction respectively and selected for further research. Overexpression of IL-10 using these two disease-inducible promoters resulted in less synovitis and markedly diminished cartilage proteoglycan depletion and in upregulation of IL-1Ra and SOCS3 gene expression. CONCLUSIONS: Our study shows that promoters of genes that are expressed locally during arthritis can be candidates for disease-regulated overexpression of biologics into arthritic joints, as shown for IL-10 in SCW arthritis. The disease-inducible approach might be promising for future tailor-made local gene therapy in arthritis.


Asunto(s)
Artritis Experimental/terapia , Cartílago Articular/metabolismo , Terapia Genética , Interleucina-10 , Proteoglicanos/metabolismo , ARN Mensajero/metabolismo , Membrana Sinovial/inmunología , Sinovitis/terapia , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Reumatoide , Pared Celular/inmunología , Expresión Génica , Proteína Antagonista del Receptor de Interleucina 1/genética , Masculino , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Proteína Amiloide A Sérica/genética , Streptococcus/inmunología , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Membrana Sinovial/patología , Sinovitis/inmunología , Sinovitis/patología
6.
Mol Imaging ; 13: 1-10, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24881106

RESUMEN

Rheumatoid arthritis (RA) and osteoarthritis (OA) are serologically and clinically distinctive, but at the local level, both diseases have many molecular pathways in common. In vivo molecular imaging can unravel the local pathologic processes involved in both diseases. In this study, we investigated matrix metalloproteinase (MMP) and cathepsin activity during cartilage destruction, in an RA and an OA mouse model, using biophotonic imaging of substrate-based probes. Mice with collagen-induced arthritis (CIA) or destabilization of the medial meniscus (DMM) were imaged using near-infrared fluorescent probes, activated by several cathepsins or MMPs. Fluorescence signal intensity was compared to synovial gene expression, histology, and cartilage staining of a neoepitope of aggrecan cleaved by MMPs with the amino acids DIPEN. Increased cathepsin and MMP activity was seen during CIA, whereas the DMM model only showed increased MMP activity. DIPEN expression was seen only during CIA. A possible explanation can be differences in gene expressions; MMP3 and -13, known to produce DIPEN neoepitopes, were upregulated in the CIA model, whereas MMP12, known to be involved in elastin degradation and chemokine inhibition, was upregulated in the DMM model. Thus, molecular imaging showed no cathepsin activity at the time of cartilage damage in the DMM model, whereas both cathepsins and MMPs are active in the CIA model during disease progression.


Asunto(s)
Artritis Reumatoide/metabolismo , Catepsinas/análisis , Metaloproteinasas de la Matriz/análisis , Imagen Molecular/métodos , Osteoartritis/metabolismo , Animales , Artritis Experimental/metabolismo , Catepsinas/metabolismo , Muerte Celular , Condrocitos/citología , Condrocitos/metabolismo , Colágeno Tipo II/efectos adversos , Colágeno Tipo II/inmunología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Rodilla de Cuadrúpedos/química , Rodilla de Cuadrúpedos/metabolismo , Rodilla de Cuadrúpedos/patología
7.
Mol Nutr Food Res ; 68(3): e2300445, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38087782

RESUMEN

SCOPE: Bovine milk extracellular vesicles (MEVs) have demonstrated therapeutic potential in regulating bone cell activity. However, the outcome of their use on alveolar bone loss has not yet been demonstrated. METHODS AND RESULTS: This study evaluates the effect of oral administration of MEVs on ovariectomized (OVX) mice. There is a reduced height of the alveolar bone crest in OVX mice by MEVs treatment, but the alveolar bone parameters are not altered. OVX mice are then submitted to a force-induced bone remodeling model by orthodontic tooth movement (OTM). MEVs-treated mice have markedly less bone remodeling movement, unlike the untreated OVX mice. Also, OVX mice treated with MEVs show an increased number of osteoblasts and osteocytes associated with higher sclerostin expression and reduce osteoclasts in the alveolar bone. Although the treatment with MEVs in OVX mice does not show differences in root structure in OTM, few odontoclasts are observed in the dental roots of OVX-treated mice. Compared to untreated mice, maxillary and systemic RANKL/OPG ratios are reduced in OVX mice treated with MEVs. CONCLUSION: Treatment with MEVs results in positive bone cell balance in the alveolar bone and dental roots, indicating its beneficial potential in treating alveolar bone loss in the nutritional context.


Asunto(s)
Pérdida de Hueso Alveolar , Ratones , Animales , Femenino , Humanos , Pérdida de Hueso Alveolar/prevención & control , Pérdida de Hueso Alveolar/metabolismo , Leche , Osteoclastos/metabolismo , Osteoblastos/metabolismo , Remodelación Ósea/fisiología , Ovariectomía
8.
Ann Rheum Dis ; 72(8): 1407-15, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23291389

RESUMEN

OBJECTIVE: A prominent role of Toll-like receptor 4 (TLR4) in arthritis is emerging. TLR4 is functional in immune cells and stromal cells. The aim was to investigate the involvement of TLR4 in bone marrow (BM)-derived and resident cells in arthritis. METHODS: Reciprocal sex-mismatched BM transplantation was performed between IL-1Ra(-/-)TLR4(+/+) and IL-1Ra(-/-)TLR4(-/-) double knockout animals in Balb/c background. Arthritis was assessed macroscopically and by histopathology. Immunity was evaluated by splenic cytokine production and flow cytometry in draining lymph node (DLN) cells. RESULTS: Arthritis progression was reduced to a similar extent in animals lacking TLR4 on BM-derived, resident cells or both. Histology revealed that joint inflammation was partially TLR4-dependent in either BM-derived or resident cells. TLR4 plays an additive role in BM-derived and resident cells in promoting cartilage erosion. By contrast, TLR4 was equally important in BM-derived and resident cells in mediating bone erosion. Systemically, TLR4 in both BM-derived and resident cells contributed to IL-17 production by splenic T-cells, whereas in the DLNs of arthritic joints this was not the case. Interestingly, in DLN, the dominant cells producing IL-17 were CD4 negative, and cell numbers were determined by TLR4 in the BM-derived cells. CONCLUSIONS: TLR4 is necessary in both BM-derived and resident cells for full-blown joint swelling, inflammation and bone erosion. Furthermore, TLR4 on BM-derived and tissue-resident cells show an additive effect in cartilage destruction. Interestingly, TLR4 on BM-derived and tissue-resident cells are both required for IL-17 production in spleen, but only in BM-derived cells in DLN.


Asunto(s)
Artritis Experimental/metabolismo , Células de la Médula Ósea/metabolismo , Rodilla de Cuadrúpedos/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Cartílago Articular/patología , Femenino , Interleucina-17/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Bazo/metabolismo , Rodilla de Cuadrúpedos/inmunología , Rodilla de Cuadrúpedos/patología
9.
Arthritis Rheum ; 64(6): 1838-47, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22147588

RESUMEN

OBJECTIVE: Increasing evidence indicates the involvement of Toll-like receptors (TLRs) in the progression of arthritis; however, the contribution of the two signaling pathways used by TLRs, which are mediated by myeloid differentiation factor 88 (MyD88) and TRIF, remains unclear. The objective of this study was to investigate the specific roles of MyD88 and TRIF in chronic experimental arthritis and the accompanying adaptive immune responses. METHODS: Chronic arthritis was induced in wild-type, MyD88(-/-) , and Trif(lps2) (TRIF(-/-) ) mice by repetitive intraarticular injections of streptococcal cell wall (SCW) fragments. SCW-specific T cell and B cell responses, joint swelling, and histopathologic changes were analyzed during chronic arthritis. RESULTS: Both MyD88 and TRIF pathways contributed to antigen-specific T cell proliferation and antibody production, with the MyD88 pathway playing the dominant role. The severity of joint swelling and synovial inflammation, as well as the histopathologic damage to cartilage and bone, was strongly dependent on MyD88 signaling, whereas TRIF was redundant. MyD88 signaling was critical for the development of pathogenic T cell response (i.e., interleukin-17 [IL-17] production) in response to SCW antigen. Interestingly, when the T cell-dependent phase was prolonged, TRIF signaling appeared to down-regulate bone erosion, an effect accompanied by an inhibitory effect on IL-17 production. CONCLUSION: This study reveals a central role of MyD88 and a counterregulatory function of TRIF in T cell-driven arthritis. The findings provide a rationale for a pathway-specific interference in order to block the pathogenic features and to preserve or stimulate the beneficial aspects of TLR signaling.


Asunto(s)
Inmunidad Adaptativa , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Artritis Experimental/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal/inmunología , Receptores Toll-Like/metabolismo , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Cartílago Articular/inmunología , Cartílago Articular/metabolismo , Cartílago Articular/patología , Proliferación Celular , Interleucina-17/inmunología , Interleucina-17/metabolismo , Articulaciones/inmunología , Articulaciones/metabolismo , Articulaciones/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Índice de Severidad de la Enfermedad , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Linfocitos T/metabolismo , Linfocitos T/patología
10.
Arthritis Rheum ; 64(10): 3313-23, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22576756

RESUMEN

OBJECTIVE: To determine the expression of suppressor of cytokine signaling 3 (SOCS-3) in human articular chondrocytes and its functional consequences. METHODS: Chondrocytes were isolated from the cartilage of patients with osteoarthritis (OA), patients with rheumatoid arthritis (RA), and trauma patients and from the healthy cartilage of patients with a femoral neck fracture. The human chondrocyte cell line G6 and primary bovine chondrocytes were used in validation experiments. SOCS-3 messenger RNA (mRNA) expression was measured by quantitative polymerase chain reaction, and SOCS-3 protein levels were determined by Western blotting and immunohistochemical analysis. To ascertain the role of SOCS-3 in the chondrocyte response to interleukin-1ß (IL-1ß) or lipopolysaccharide (LPS), the expression of SOCS3 was either reduced by small interfering RNA or enhanced by viral transduction. RESULTS: The expression of SOCS-3 mRNA (but not that of SOCS-1 mRNA) was significantly enhanced in chondrocytes obtained from OA cartilage (mean ± SD ΔC(t) 3.4 ± 1.0) and RA cartilage (ΔC(t) 3.4 ± 1.4) compared with cartilage obtained from patients with femoral neck fracture (ΔC(t) 5.3 ± 1.2). The expression of SOCS3 correlated significantly with that of other genes known to be expressed in arthritic chondrocytes, such as MMP13 (r = 0.743), ADAMTS4 (r = 0.779), and ADAMTS5 (r = 0.647), and an inverse relationship was observed with COL2A1 (r = -0.561). Up-regulation of SOCS-3 by IL-1 in G6 chondrocytes and its spontaneous expression in OA chondrocytes were reduced by mithramycin, a specific inhibitor of transcription factor Sp-1. Overexpression of SOCS-3 in bovine chondrocytes reduced IL-1- and LPS-induced nitric oxide production and insulin-like growth factor 1-induced proteoglycan synthesis. Interestingly, a similar impairment of function was observed in OA chondrocytes, which was partially restored by SOCS-3 gene knockdown. CONCLUSION: This study demonstrated that both SOCS-3 mRNA and SOCS-3 protein are expressed in human arthritic chondrocytes and affect cellular responses involved in cartilage pathology.


Asunto(s)
Artritis Reumatoide/metabolismo , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Osteoartritis de la Cadera/metabolismo , Osteoartritis de la Rodilla/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS4 , Proteína ADAMTS5 , Adulto , Anciano , Anciano de 80 o más Años , Animales , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Bovinos , Línea Celular , Condrocitos/efectos de los fármacos , Condrocitos/patología , Femenino , Humanos , Interleucina-1/farmacología , Masculino , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Persona de Mediana Edad , Osteoartritis de la Cadera/genética , Osteoartritis de la Cadera/patología , Osteoartritis de la Rodilla/genética , Osteoartritis de la Rodilla/patología , Procolágeno N-Endopeptidasa/genética , Procolágeno N-Endopeptidasa/metabolismo , Proteoglicanos/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Regulación hacia Arriba/efectos de los fármacos
11.
PLoS One ; 18(12): e0295076, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38051739

RESUMEN

Mesenchymal stromal/stem cell derived-extracellular vesicles (MSC-EVs) have gained interest as drug delivery nanoparticles, having immunoregulatory and potentiating tissue repair property. To maintain growth of MSCs and obtain pure MSC-derived EVs, the culture media should contain fetal bovine serum (FBS) devoid of EVs, as the presence of FBS EVs confounds the properties of MSC-EVs. Therefore, we tested three methods: 18h ultracentrifugation (UC) and ultrafiltration (UF), which are common FBS EV depletion methods in current MSC-EV research, and polyethylene glycol (PEG) precipitation to obtain three EV depleted FBS (EVdFBS) batches, and compared them to FBS and commercial (Com) EVdFBS on human adipose stem cell (hADSC) growth, differentiation, enrichment of EVs in hADSC supernatant and their biological function on collagen metabolism. Our comparative study showed UC and UF vary in terms of depletion efficiency and do not completely deplete EVs and affects the growth-promoting quality of FBS. Specifically, FBS EV depletion was comparable between PEG (95.6%) and UF (96.6%) but less by UC (82%), as compared to FBS. FBS protein loss was markedly different among PEG (47%), UF (87%), and UC (51%), implying the ratio of EV depletion over protein loss was PEG (2.03), UF (1.11), and UC (1.61). A significant decrease of TGFß/Smad signaling, involving in MSC growth and physiology, was observed by UF. After 96 hours of exposure to 5% FBS or 5% four different EVdFBS cell growth media, the osteogenesis ability of hADSCs was not impaired but slightly lower mRNA expression level of Col2a observed in EVdFBS media during chondrogenesis. In consistent with low confluency of hADSCs observed by optical microscope, cell proliferation in response to 5% UF EVdFBS media was inhibited significantly. Importantly, more and purer ADSCs EVs were obtained from ADSCs cultured in 5% PEG EVdFBS media, and they retained bioactive as they upregulated the expression of Col1a1, TIMP1 of human knee synovial fibroblast. Taken together, this study showed that PEG precipitation is the most efficient method to obtain EV depleted FBS for growth of MSCs, and to obtain MSC EVs with minimal FBS EV contamination.


Asunto(s)
Vesículas Extracelulares , Albúmina Sérica Bovina , Humanos , Albúmina Sérica Bovina/metabolismo , Vesículas Extracelulares/metabolismo , Diferenciación Celular , Medios de Cultivo/farmacología , Medios de Cultivo/metabolismo , Polietilenglicoles/farmacología , Polietilenglicoles/metabolismo
12.
Front Med (Lausanne) ; 10: 1247778, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38274452

RESUMEN

Background: Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic synovial inflammation and cartilage/bone damage. Intercellular messengers such as IL-1 and TNF play a crucial role in the pathophysiology of RA but have limited diagnostic and prognostic values. Therefore, we assessed whether the protein content of the recently discovered extracellular vesicles (EVs), which have gained attention in the pathogenesis of RA, correlates with disease activity parameters in RA patients. Methods: We identified and quantified proteins in plasma-derived EVs (pEVs), isolated by size exclusion chromatography from 17 RA patients by mass spectrophotometry (MS). Quantified protein levels were correlated with laboratory and clinical parameters and the patient's own global assessment of their disease activity (PGA-VAS). In a second MS run, the pEV proteins of nine other RA patients were quantified and compared to those from nine healthy controls (HC). Results: No differences were observed in the concentration, size, and protein content of pEVs from RA patients. Proteomics revealed >95% overlapping proteins in RA-pEVs, compared to HC-pEVs (data are available via ProteomeXchange with identifier PXD046058). Remarkably, in both runs, the level of far more RA-pEV proteins correlated positively to PGA-VAS than to either clinical or laboratory parameters. Interestingly, all observed PGA-VAS positively correlated RA-pEV proteins were associated with the actin-cytoskeleton linker proteins, ezrin, and moesin. Conclusion: Our observation suggests that PGA-VAS (loss of vitality) may have a different underlying pathological mechanism in RA, possibly related to enhanced muscle actin-cytoskeleton activity. Furthermore, our study contributes to the growing awareness and evidence that pEVs contain valuable biomarkers for diseases, with added value for RA patients.

13.
Cells ; 12(14)2023 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-37508577

RESUMEN

Joint pain severity in arthritic diseases differs between sexes and is often more pronounced in women. This disparity is thought to stem from biological mechanisms, particularly innate immunity, yet the understanding of sex-specific differences in arthritic pain remains incomplete. This study aims to investigate these disparities using an innate immunity-driven inflammation model induced by intra-articular injections of Streptococcus Cell Wall fragments to mimic both acute and pre-sensitized joint conditions. Nociceptive behavior was evaluated via gait analysis and static weight-bearing, and inflammation was evaluated via joint histology and the synovial gene expression involved in immune response. Although acute inflammation and pain severity were comparable between sexes, distinct associations between synovial inflammatory gene expression and static nociceptive behavior emerged. These associations delineated sex-specific relationships with pain, highlighting differential gene interactions (Il6 versus Cybb on day 1 and Cyba/Gas6 versus Nos2 on day 8) between sexes. In conclusion, our study found that, despite similar pain severity between sexes, the association of inflammatory synovial genes revealed sex-specific differences in the molecular inflammatory mechanisms underlying pain. These findings suggest a path towards more personalized treatment strategies for pain management in arthritis and other inflammatory joint diseases.


Asunto(s)
Sinovitis , Masculino , Humanos , Ratones , Femenino , Animales , Sinovitis/metabolismo , Dolor , Inflamación/complicaciones , Artralgia , Inmunidad Innata
14.
Mol Nutr Food Res ; 66(6): e2100764, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34965027

RESUMEN

SCOPE: Data from the Osteoarthritis Initiative shows that females who drink milk regularly have less joint cartilage loss and OA progression, but the biologic mechanism is unclear. Bovine milk is a rich source of extracellular vesicles (EVs), which are small phospholipid bilayer bound structures that facilitate intercellular communication. In this study, the authors aim to evaluate whether these EVs may have the capacity to protect cartilage from osteoarthritis patients, ex vivo, by directly effecting chondrocytes. METHODS AND RESULTS: Human cartilage explants are exposed to cow's milk-derived EVs (CMEVs), which results in reduced sulfated glycosaminoglycan release and inhibition of metalloproteinase-1 expression. Incubation of articular chondrocytes with CMEVs also effectively reduces expression of cartilage destructive enzymes (ADAMTS5, MMPs), which play key roles in the disease progression. In part, these findings are attributed to the presence of TGFß on these vesicles, and in addition, a possible role is reserved for miR-148a, which is functionally transferred by CMEVs. CONCLUSION: These findings highlight the therapeutic potential of local CMEV delivery in osteoarthritic joints, where inflammatory and catabolic mediators are responsible for joint pathology. CMEVs are carriers of both TGFß and miR-148a, two essential regulators for maintaining chondrocyte homeostasis and protection against cartilage destruction.


Asunto(s)
Cartílago Articular , Vesículas Extracelulares , MicroARNs , Osteoartritis , Animales , Cartílago Articular/metabolismo , Cartílago Articular/patología , Bovinos , Femenino , Humanos , MicroARNs/metabolismo , Leche , Osteoartritis/metabolismo , Osteoartritis/patología , Osteoartritis/terapia , Factor de Crecimiento Transformador beta/metabolismo
15.
Ann Rheum Dis ; 70(7): 1311-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21474483

RESUMEN

OBJECTIVE: To evaluate the applicability of a lentiviral (LV) serum amyloid A3 (Saa3)-promoter luciferase (Luc) reporter for assessing inflammation in experimental arthritis, synovial fibroblasts (SF) from osteoarthritis (OA) and rheumatoid arthritis (RA) patients. METHODS: In mice, synovium was transduced in vivo by cholesterol optimised LV, and two flares of acute joint inflammation were induced by injection of streptococcal cell wall (SCW) material into the knee-joint cavity. The time course of synovial inflammation was assessed using ex vivo luciferase assays, and histology. Uptake of (99m)technetium (Tc) was used to assess oedema. SF (n=12) of RA and OA patients were stratified by hierarchical clustering of whole genome expression profiles. Relative Saa3-promoter responses were determined in cytokine- or toll-like receptor (TLR)-stimulated SF subgroups. RESULTS: In vivo, the Saa3-promoter reporter activity was strongly upregulated at 1 and 2 days after the first and second SCW challenge. The Saa3-promoter activities during acute inflammation correlated with Tc uptake measurements but were more sensitive and able to respond to the ongoing synovitis in the chronic phase of SCW arthritis. Molecular stratification defined two inflammatory SF subtypes, unrelated to disease classification. Relative Saa3-promoter responses to interleukin 1ß, tumour necrosis factor α and TLR4 agonist were significantly increased in OA/RA SF with a high compared to a low inflammatory profile subtype. Serum stimulation of the Saa3-promoter reporter cell-line could distinguish between healthy and RA patients. CONCLUSION: The Saa3-promoter reporter demonstrates a robust and feasible tool for assessing the course and severity of experimental arthritis and for distinguishing molecularly distinct inflammatory SF subtypes from a heterogeneous patient population.


Asunto(s)
Artritis Experimental/diagnóstico , Proteína Amiloide A Sérica/genética , Membrana Sinovial/patología , Adulto , Anciano , Animales , Artritis Experimental/patología , Artritis Reumatoide/diagnóstico , Colesterol/farmacología , Análisis por Conglomerados , Diagnóstico Diferencial , Relación Dosis-Respuesta a Droga , Estudios de Factibilidad , Fibroblastos/patología , Perfilación de la Expresión Génica/métodos , Vectores Genéticos , Humanos , Lentivirus/efectos de los fármacos , Lentivirus/genética , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Osteoartritis/diagnóstico , Regiones Promotoras Genéticas , Tecnecio , Transducción Genética
16.
Ann Rheum Dis ; 70(12): 2167-75, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21873688

RESUMEN

OBJECTIVE: Suppressor of cytokine signalling (SOCS) proteins constitute a class of intracellular proteins that are key physiological regulators of immune cell function. It has previously been shown that antigen-presenting cells (APCs) overexpressing SOCS3 steer T helper immune responses and protect against experimental arthritis. A study was undertaken to investigate the contribution of SOCS3 in regulating invariant natural killer T (iNKT) cell responses during collagen-induced arthritis (CIA). METHODS: DBA/1 mice were immunised with type II collagen and adenoviruses encoding SOCS3 were administered intravenously before the clinical onset of arthritis. Murine APCs overexpressing SOCS3 were co-cultured with an iNKT cell hybridoma and interleukin 2 (IL-2) release was measured by Luminex multi-analyte technology. The frequency and activation of primary iNKT cells was assessed by flow cytometry. Murine APCs were analysed for cytokine and CD1d expression following viral SOCS3 gene transfer. RESULTS: Viral overexpression of SOCS3 in APCs resulted in reduced activation of the iNKT cell hybridoma. Importantly, during initiation of CIA, adenovirus-mediated overexpression of SOCS3 in hepatic and splenic APCs inhibited iNKT cell expansion in both organs. The iNKT cell population from SOCS3-treated mice showed low expression of the early activation marker CD69 and primary liver iNKT cells produced less interferon γ and IL-4 upon α-galactosylceramide stimulation. No differences in CD1d surface expression were observed, but SOCS3-transduced APCs produced decreased levels of proinflammatory cytokines and increased levels of IL-10. CONCLUSION: These results demonstrate a critical role for SOCS3 in controlling the immunostimulatory capacities of APCs, which has direct implications for the effector function of iNKT cells during arthritis.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Artritis Experimental/inmunología , Células T Asesinas Naturales/inmunología , Proteínas Supresoras de la Señalización de Citocinas/sangre , Adenoviridae/genética , Animales , Antígenos CD1d/metabolismo , Artritis Experimental/prevención & control , Células Cultivadas , Citocinas/inmunología , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Hígado/inmunología , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos DBA , Bazo/inmunología , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética
17.
Rheumatology (Oxford) ; 50(7): 1216-25, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21335610

RESUMEN

OBJECTIVE: Activated synovial fibroblasts are key players in the pathogenesis of RA by driving inflammation and joint destruction. Numerous molecules including cytokines and Toll-like receptor (TLR) ligands induce pro-inflammatory signalling and gene expression through a hierarchical network of kinases. Upstream mitogen-activated protein kinase kinase kinases (MAP3Ks) represent an attractive target for RA treatment. In this study, we sought to determine the role of the MAP3K TGF-ß-activated kinase 1 (TAK1) in cytokine and TLR-mediated signalling. METHODS: TAK1 activity was inhibited using either a small molecule inhibitor or lentivirally overexpressed kinase-inactive TAK1-K63W mutant in murine embryonic and human dermal and synovial fibroblasts. Fibroblasts were stimulated with IL-1, TNF, TLR2 or TLR4 agonists and responses were evaluated using transcriptional reporters, western blotting and analysis of gene expression of collagenases (MMP3 and MMP13), cytokines (IL-1ß and IL-6) and chemokines (IL-8 and MCP-1). RESULTS: TAK1 inhibition abrogated cytokine- and TLR-induced nuclear factor-κB (NF-κB) and Saa3-promoter reporter activation in murine and human dermal fibroblasts. In synovial fibroblasts, TAK1 regulated IL-1 and TNF-mediated NF-κB, but not Saa3-promoter reporter activation. Inducible mRNA expression of cytokines, collagenases and chemokines, except MCP-1, was TAK1 dependent for IL-1, TNF and TLR2 signalling. Unexpectedly, TLR4-mediated NF-κB reporter activation and inducible mRNA expression was fully TAK1 independent. Accordingly, NF-κB p65 and p38 MAPK phosphorylation was unaffected by TAK1 inhibition. CONCLUSION: In general, TAK1 crucially regulates IL-1 and TNF signalling in fibroblasts. Interestingly, TLR4 signalling is specifically TAK1 independent in synovial fibroblasts. Consequently, therapeutic TAK1 inhibition in arthropathies may not dampen the damage-associated molecular pattern-mediated TLR4 activation of synovial fibroblasts.


Asunto(s)
Fibroblastos/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Membrana Sinovial/citología , Membrana Sinovial/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Análisis de Varianza , Animales , Artritis Reumatoide/metabolismo , Western Blotting , Células Cultivadas , Activación Enzimática , Femenino , Fibroblastos/citología , Masculino , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Transducción de Señal , Receptor Toll-Like 4/análisis
18.
Front Immunol ; 12: 703277, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34394100

RESUMEN

Many studies provided compelling evidence that extracellular vesicles (EVs) are involved in the regulation of the immune response, acting as both enhancers and dampeners of the immune system, depending on the source and type of vesicle. Research, including ours, has shown anti-inflammatory effects of milk-derived EVs, using human breast milk as well as bovine colostrum and store-bought pasteurized cow milk, in in vitro systems as well as therapeutically in animal models. Although it is not completely elucidated which proteins and miRNAs within the milk-derived EVs contribute to these immunosuppressive capacities, one proposed mechanism of action of the EVs is via the modulation of the crosstalk between the (intestinal) microbiome and their host health. There is increasing awareness that the gut plays an important role in many inflammatory diseases. Enhanced intestinal leakiness, dysbiosis of the gut microbiome, and bowel inflammation are not only associated with intestinal diseases like colitis and Crohn's disease, but also characteristic for systemic inflammatory diseases such as lupus, multiple sclerosis, and rheumatoid arthritis (RA). Strategies to target the gut, and especially its microbiome, are under investigation and hold a promise as a therapeutic intervention for these diseases. The use of milk-derived EVs, either as stand-alone drug or as a drug carrier, is often suggested in recent years. Several research groups have studied the tolerance and safety of using milk-derived EVs in animal models. Due to its composition, milk-derived EVs are highly biocompatible and have limited immunogenicity even cross species. Furthermore, it has been demonstrated that milk-derived EVs, when taken up in the gastro-intestinal tract, stay intact after absorption, indicating excellent stability. These characteristics make milk-derived EVs very suitable as drug carriers, but also by themselves, these EVs already have a substantial immunoregulatory function, and even without loading, these vesicles can act as therapeutics. In this review, we will address the immunomodulating capacity of milk-derived EVs and discuss their potential as therapy for RA patients. Review criteria: The search terms "extracellular vesicles", "exosomes", "microvesicles", "rheumatoid arthritis", "gut-joint axis", "milk", and "experimental arthritis" were used. English-language full text papers (published between 1980 and 2021) were identified from PubMed and Google Scholar databases. The reference list for each paper was further searched to identify additional relevant articles.


Asunto(s)
Artritis Reumatoide/inmunología , Vesículas Extracelulares/inmunología , Intestinos/inmunología , Leche Humana/inmunología , Animales , Artritis Reumatoide/terapia , Femenino , Humanos , Inmunomodulación , Articulaciones/inmunología
19.
Mol Ther ; 17(11): 1877-87, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19690516

RESUMEN

The promoter regions of genes that are differentially regulated in the synovial membrane during the course of rheumatoid arthritis (RA) represent attractive candidates for application in transcriptionally targeted gene therapy. In this study, we applied an unbiased computational approach to define proximal-promoters from a gene expression profiling study of murine experimental arthritis. Synovium expression profiles from progressing stages of collagen-induced arthritis (CIA) were classified into six distinct groups using k-means clustering. Using an algorithm based on local over-representation and comparative genomics, we identified putatively functional transcription factor-binding sites (TFBS) in TATA-dependent proximal-promoters. Applying a filter based on spacing between TATA box and transcription start site (TSS) combined with the presence of over-represented nuclear factor kappaB (NFkappaB), AP-1, or CCAAT/enhancer-binding protein beta (C/EBPbeta) sites, 382 candidate murine and human promoters were reduced to 66, corresponding to 45 genes. In vitro, 9 out of 10 computationally defined promoter regions conferred cytokine-inducible expression in murine cells and human synovial fibroblasts. Under these conditions, the serum amyloid A3 (Saa3) promoter showed the strongest transcriptional induction and strength. We applied this promoter for driving therapeutically efficacious levels of the interleukin-1 receptor antagonist (Il1rn) in a disease-regulated fashion. These results demonstrate the value of bioinformatics for guiding the selection of endogenous promoters for transcriptionally targeted gene therapy.


Asunto(s)
Artritis Reumatoide/terapia , Biología Computacional/métodos , Terapia Genética/métodos , Regiones Promotoras Genéticas/genética , Adenoviridae/genética , Algoritmos , Animales , Bovinos , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Células HeLa , Humanos , Proteína Antagonista del Receptor de Interleucina 1/genética , Proteína Antagonista del Receptor de Interleucina 1/fisiología , Lentivirus/genética , Masculino , Ratones , Modelos Genéticos , Células 3T3 NIH , Proteína Amiloide A Sérica/genética
20.
PLoS One ; 15(7): e0236508, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32726333

RESUMEN

Extracellular vesicles (EVs) are cell membrane-derived phospholipid bilayer nanostructures that contain bioactive proteins, enzymes, lipids and polymers of nucleotides. They play a role in intercellular communication and are present in body fluids. EVs can be isolated by methods like ultracentrifugation (UC), polyethylene-glycol-precipitation (PEG) or size exclusion chromatography (SEC). The co-presence of immunoglobulins (Ig) in EV samples isolated from plasma (pEVs) is often reported and this may influence the assessment of the biological function and phenotype of EVs in bio- and immunoassay. Here, we studied the presence of an Ig-based therapeutic (etanercept) in pEV samples isolated from rheumatoid arthritis (RA) patients and improved the isolation method to obtain purer pEVs. From plasma of etanercept (Tumor-necrosis-factor (TNF)-α antibodies)-treated RA patients pEVs were isolated by either UC, PEG or SEC. SEC isolated pEVs showed the highest particle-to-protein ratio. Strong TNF-α inhibition determined in a TNF-α sensitive reporter assay was observed by pEVs isolated by UC and PEG, and to a lesser extent by SEC, suggesting the presence of functional etanercept. SEC isolation of etanercept or labelled immunoglobulin G (IgG) showed co-isolation of these antibodies in the pEV fraction in the presence of plasma or a high protein (albumin) concentration. To minimize the presence of etanercept or immunoglobulins, we extended SEC (eSEC) column length from 56mm to 222mm (total stacking volume unchanged). No effect on the amount of isolated pEVs was observed while protein and IgG content were markedly reduced (90%). Next, from six etanercept- treated RA patients, pEVs were isolated on a eSEC or standard SEC column, in parallel. TNF-α inhibition was again observed in pEVs isolated by conventional SEC but not by eSEC. To confirm the purer pEVs isolated by eSEC the basal IL-8 promoter activation in human monocytes was determined and in 4 out of 5 SEC isolated pEVs activation was observed while eSEC isolated pEVs did not. This study shows that extended SEC columns yielded pEVs without detectable biologicals and with low protein and IgG levels. This isolation method will improve the characterization of pEVs as potential biomarkers and mediators of disease.


Asunto(s)
Productos Biológicos/sangre , Proteínas Sanguíneas/análisis , Vesículas Extracelulares/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Cromatografía en Gel , Etanercept/sangre , Etanercept/uso terapéutico , Vesículas Extracelulares/química , Humanos , Inmunoglobulina G/análisis , Interleucina-8/genética , Regiones Promotoras Genéticas , Activación Transcripcional , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA