Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
ACS Omega ; 9(17): 19566-19577, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38708281

RESUMEN

The present study introduces an advanced surface modification approach combining electrochemical anodization and non-thermal plasma treatment, tailored for biomedical applications on stainless steel grade 316L (SS316L) surfaces. Nanopores with various diameters (100-300 nm) were synthesized with electrochemical anodization, and samples were further modified with non-thermal oxygen plasma. The surface properties of SS316L surfaces were examined by scanning electron microscopy, atomic force microscopy, X-ray photoemission spectroscopy, and Water contact angle measurements. It has been shown that a combination of electrochemical anodization and plasma treatment significantly alters the surface properties of SS316L and affects its interactions with blood platelets and human coronary cells. Optimal performance is attained on the anodized specimen featuring pores within the 150-300 nm diameter range, subjected to subsequent oxygen plasma treatment; the absence of platelet adhesion was observed. At the same time, the sample demonstrated good endothelialization and a reduction in smooth muscle cell adhesion compared to the untreated SS316L and the sample with smaller pores (100-150 nm). This novel surface modification strategy has significant implications for improving biocompatibility and performance of SS316L in biomedical applications.

2.
Int J Mol Sci ; 24(13)2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37445972

RESUMEN

Studies on the serum biomarkers of granulomatous inflammation and pulmonary interstitial disease in intrathoracic sarcoidosis have shown conflicting results. We postulated that differences in the concentrations of serum biomarkers can be explained by the heterogenous patterns of sarcoidosis seen on thoracic HRCT. Serum biomarker levels in 79 consecutive patients, newly diagnosed with intrathoracic sarcoidosis, were compared to our control group of 56 healthy blood donors. An analysis was performed with respect to HRCT characteristics (the presence of lymph node enlargement, perilymphatic or peribronchovascular infiltrates, ground-glass lesions, or fibrosis), CXR, and disease extent. Serum levels of CXCL9, CXCL10, CTO, and CCL18 were statistically significantly increased in all patients compared to controls. Serum levels of CA15.3 were statistically significantly increased in all patients with parenchymal involvement. SAA was increased in patients with ground-glass lesions while SP-D levels were statistically significantly increased in patients with lung fibrosis. Only SP-D and CA15.3 showed a significant correlation to interstitial disease extent. In conclusion, we found that sarcoidosis patients with different HRCT patterns of intrathoracic sarcoidosis have underlying biochemical differences in their serum biomarkers transcending Scadding stages. The stratification of patients based on both radiologic and biochemical characteristics could enable more homogenous patient selection for further prognostic studies.


Asunto(s)
Enfermedades Pulmonares Intersticiales , Sarcoidosis , Humanos , Proteína D Asociada a Surfactante Pulmonar , Enfermedades Pulmonares Intersticiales/patología , Sarcoidosis/diagnóstico por imagen , Pulmón/patología , Tomografía Computarizada por Rayos X , Biomarcadores
3.
Int J Mol Sci ; 24(6)2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36982831

RESUMEN

Animal models are invaluable in the research of the pathophysiology of interstitial cystitis/bladder pain syndrome (IC/BPS), a chronic aseptic urinary bladder disease of unknown etiology that primarily affects women. Here, a mouse model of IC/BPS was induced with multiple low-dose cyclophosphamide (CYP) applications and thoroughly characterized by RNA sequencing, qPCR, Western blot, and immunolabeling to elucidate key inflammatory processes and sex-dependent differences in the bladder inflammatory response. CYP treatment resulted in the upregulation of inflammatory transcripts such as Ccl8, Eda2r, and Vegfd, which are predominantly involved in innate immunity pathways, recapitulating the crucial findings in the bladder transcriptome of IC/BPS patients. The JAK/STAT signaling pathway was analyzed in detail, and the JAK3/STAT3 interaction was found to be most activated in cells of the bladder urothelium and lamina propria. Sex-based data analysis revealed that cell proliferation was more pronounced in male bladders, while innate immunity and tissue remodeling processes were the most distinctive responses of female bladders to CYP treatment. These processes were also reflected in prominent histological changes in the bladder. The study provides an invaluable reference dataset for preclinical research on IC/BPS and an insight into the sex-specific mechanisms involved in the development of IC/BPS pathology, which may explain the more frequent occurrence of this disease in women.


Asunto(s)
Cistitis Intersticial , Ratones , Animales , Femenino , Masculino , Cistitis Intersticial/genética , Cistitis Intersticial/patología , Vejiga Urinaria/patología , Transcriptoma , Pelvis/patología , Proliferación Celular , Modelos Animales de Enfermedad , Receptor Xedar/metabolismo
4.
PLoS One ; 17(7): e0271737, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35877764

RESUMEN

More than 30 types of amyloids are linked to close to 50 diseases in humans, the most prominent being Alzheimer's disease (AD). AD is brain-related local amyloidosis, while another amyloidosis, such as AA amyloidosis, tends to be more systemic. Therefore, we need to know more about the biological entities' influencing these amyloidosis processes. However, there is currently no support system developed specifically to handle this extraordinarily complex and demanding task. To acquire a systematic view of amyloidosis and how this may be relevant to the brain and other organs, we needed a means to explore "amyloid network systems" that may underly processes that leads to an amyloid-related disease. In this regard, we developed the DES-Amyloidoses knowledgebase (KB) to obtain fast and relevant information regarding the biological network related to amyloid proteins/peptides and amyloid-related diseases. This KB contains information obtained through text and data mining of available scientific literature and other public repositories. The information compiled into the DES-Amyloidoses system based on 19 topic-specific dictionaries resulted in 796,409 associations between terms from these dictionaries. Users can explore this information through various options, including enriched concepts, enriched pairs, and semantic similarity. We show the usefulness of the KB using an example focused on inflammasome-amyloid associations. To our knowledge, this is the only KB dedicated to human amyloid-related diseases derived primarily through literature text mining and complemented by data mining that provides a novel way of exploring information relevant to amyloidoses.


Asunto(s)
Enfermedad de Alzheimer , Amiloidosis , Amiloide , Humanos , Bases del Conocimiento , Proteína Amiloide A Sérica
5.
Front Cell Dev Biol ; 10: 872688, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35573685

RESUMEN

We present an optimized dissociation protocol for preparing high-quality skin cell suspensions for in-depth single-cell RNA-sequencing (scRNA-seq) analysis of fresh and cultured human skin. Our protocol enabled the isolation of a consistently high number of highly viable skin cells from small freshly dissociated punch skin biopsies, which we use for scRNA-seq studies. We recapitulated not only the main cell populations of existing single-cell skin atlases, but also identified rare cell populations, such as mast cells. Furthermore, we effectively isolated highly viable single cells from ex vivo cultured skin biopsy fragments and generated a global single-cell map of the explanted human skin. The quality metrics of the generated scRNA-seq datasets were comparable between freshly dissociated and cultured skin. Overall, by enabling efficient cell isolation and comprehensive cell mapping, our skin dissociation-scRNA-seq workflow can greatly facilitate scRNA-seq discoveries across diverse human skin pathologies and ex vivo skin explant experimentations.

6.
Curr Opin Rheumatol ; 33(6): 505-513, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34534166

RESUMEN

PURPOSE OF REVIEW: Adipose tissue is closely associated with systemic sclerosis (SSc)-pathology, both anatomically and functionally. This review focuses on local effects of adipocytes in the context of adipose to mesenchymal transdifferentiation (AMT), effects of the adipose stromal vascular fraction on SSc pathogenesis and systemic effects of adipose tissue secretome. RECENT FINDINGS: Novel populations of fibroblasts evolving from adipose tissue were identified- for example COL11+ cancer-associated fibroblasts differentiated from adipose-derived stromal cells. Lipofibroblasts in human lungs were described using nonconventional markers that allow more effective population identification. These findings could make an important contribution to further clarification of adipocyte involvement in SSc.Recent studies confirmed that lipolysis contributes to fibrogenesis through AMT differentiation and release of fatty acids (FA). Unbalanced metabolism of FA has been reported in several studies in SSc. Other adipose tissue secretome molecules (e.g. lysophosphatidic acid), novel adipokines and extracellular vesicles from adipose mesenchymal stem cells make important contributions to the pro-/antifibrotic balance. SUMMARY: There is a growing evidence of important contribution of adipose tissue and its secretome to SSc pathogenesis. Novel techniques such as single-cell RNA sequencing (scRNAseq) and metabolomics, albeit challenging to use in adipose tissue, will provide further evidence.


Asunto(s)
Células Madre Mesenquimatosas , Esclerodermia Sistémica , Adipocitos , Tejido Adiposo , Diferenciación Celular , Humanos
7.
Int J Mol Sci ; 22(12)2021 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-34204585

RESUMEN

In this study, we explored expression of microRNA (miR), miR-target genes and matrix remodelling molecules in temporal artery biopsies (TABs) from treatment-naïve patients with giant cell arteritis (GCA, n = 41) and integrated these analyses with clinical, laboratory, ultrasound and histological manifestations of GCA. NonGCA patients (n = 4) served as controls. GCA TABs exhibited deregulated expression of several miRs (miR-21-5p, -145-5p, -146a-5p, -146b-5p, -155-5p, 424-3p, -424-5p, -503-5p), putative miR-target genes (YAP1, PELI1, FGF2, VEGFA, KLF4) and matrix remodelling factors (MMP2, MMP9, TIMP1, TIPM2) with key roles in Toll-like receptor signaling, mechanotransduction and extracellular matrix biology. MiR-424-3p, -503-5p, KLF4, PELI1 and YAP1 were identified as new deregulated molecular factors in GCA TABs. Quantities of miR-146a-5p, YAP1, PELI1, FGF2, TIMP2 and MMP9 were particularly high in histologically positive GCA TABs with occluded temporal artery lumen. MiR-424-5p expression in TABs and the presence of facial or carotid arteritis on ultrasound were associated with vision disturbances in GCA patients. Correlative analysis of miR-mRNA quantities demonstrated a highly interrelated expression network of deregulated miRs and mRNAs in temporal arteries and identified KLF4 as a candidate target gene of deregulated miR-21-5p, -146a-5p and -155-5p network in GCA TABs. Meanwhile, arterial miR and mRNA expression did not correlate with constitutive symptoms and signs of GCA, elevated markers of systemic inflammation nor sonographic characteristics of GCA. Our study provides new insights into GCA pathophysiology and uncovers new candidate biomarkers of vision impairment in GCA.


Asunto(s)
Regulación de la Expresión Génica , Redes Reguladoras de Genes , Arteritis de Células Gigantes/etiología , Arteritis de Células Gigantes/metabolismo , MicroARNs/genética , Interferencia de ARN , ARN Mensajero/genética , Arterias Temporales/metabolismo , Biomarcadores , Biopsia , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Arteritis de Células Gigantes/diagnóstico , Humanos , Inmunohistoquímica , Factor 4 Similar a Kruppel , Evaluación de Síntomas , Arterias Temporales/patología , Ultrasonografía
8.
Int J Mol Sci ; 22(8)2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33920997

RESUMEN

Deregulation of adiponectin is found in systemic autoimmune rheumatic diseases (SARDs). Its expression is downregulated by various inflammatory mediators, but paradoxically, elevated serum levels are present in SARDs with high inflammatory components, such as rheumatoid arthritis and systemic lupus erythematosus. Circulating adiponectin is positively associated with radiographic progression in rheumatoid arthritis as well as with cardiovascular risks and lupus nephritis in systemic lupus erythematosus. However, in SARDs with less prominent inflammation, such as systemic sclerosis, adiponectin levels are low and correlate negatively with disease activity. Regulators of adiponectin gene expression (PPAR-γ, Id3, ATF3, and SIRT1) and inflammatory cytokines (interleukin 6 and tumor necrosis factor α) are differentially expressed in SARDs and could therefore influence total adiponectin levels. In addition, anti-inflammatory therapy could also have an impact, as tocilizumab treatment is associated with increased serum adiponectin. However, anti-tumor necrosis factor α treatment does not seem to affect its levels. Our review provides an overview of studies on adiponectin levels in the bloodstream and other biological samples from SARD patients and presents some possible explanations why adiponectin is deregulated in the context of therapy and gene regulation.


Asunto(s)
Adiponectina/metabolismo , Enfermedades Autoinmunes/metabolismo , Enfermedades Reumáticas/metabolismo , Adiponectina/sangre , Adiponectina/química , Adiponectina/genética , Animales , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/terapia , Citocinas/metabolismo , Humanos , Modelos Biológicos , Enfermedades Reumáticas/sangre , Enfermedades Reumáticas/terapia , Factores de Transcripción/metabolismo
9.
Curr Res Transl Med ; 68(4): 217-224, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32843323

RESUMEN

BACKGROUND: Mesenchymal stromal cell (MSC) - based therapies are emerging as promising treatment of various autoimmune diseases, however the utility of different MSC tissue sources remains elusive. We aimed to characterize MSC from different origins, namely bone marrow (BM), adipose tissue (AT) and umbilical cord (UC) and determine their functional effects on normal human lung fibroblasts (NHLF). METHODS: BM-, AT- or UC-MSC were isolated each from 3 different healthy donors. The gene expression and protein secretion were analyzed at basal level, along with TNFα-, IL-1ß- and SAA- stimulated cells using real-time PCR and Luminex technology. Effect of conditioned medium (CM) from different MSC sources on migration was determined with wound scratch assay, while mitotic and apoptotic rates were studied using immunofluorescence microscopy. RESULTS: BM-MSC expressed highest basal mRNA levels of SDF1 and VCAM-1, while other genes were similarly expressed between MSC origins. TNFα priming of AT-MSC gained a prominent increase in IDO1 and CCL5 gene expression, with 928-fold and 4396-fold changes, respectively. Among all tissue sources, basal UC-MSC released highest protein levels of most measured analytes, including IL-6, IL-8, MCP-1, ICAM1, HGF, MMP1 and CH3L1. BM- and AT-MSC derived CM enhanced wound closure in NHLF, while an opposite effect was observed with UC-MSC derived CM. Our data also suggests that MSC-CM could contribute to decreased mitotic potential and increased apoptotic rate in lung fibroblasts. CONCLUSIONS: Our study highlights origin-specific MSC profile differences and emphasizes a heterogenic response of different MSC to inflammatory stimuli.


Asunto(s)
Tejido Adiposo , Células de la Médula Ósea , Células Madre Mesenquimatosas , Cordón Umbilical , Tejido Adiposo/citología , Células Cultivadas , Expresión Génica , Humanos , Células Madre Mesenquimatosas/fisiología , Cordón Umbilical/citología
10.
Atherosclerosis ; 291: 1-8, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31629987

RESUMEN

BACKGROUND AND AIMS: Patients with rheumatic diseases have an increased risk of atherosclerosis with up-regulated serum amyloid A (SAA), tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), which were reported to activate human coronary artery endothelial cells (HCAEC). We aimed to investigate the effects of TNF-α inhibitor infliximab and anti-infliximab antibodies on the TNF-α/IL-1ß/SAA activated HCAEC. METHODS: HCAEC were incubated with TNF-α, IL-1ß, SAA, infliximab, anti-infliximab antibodies and their combinations. The protein levels of pro- and anti-atherogenic analytes were measured in supernatants using ELISA and multiplex assays, while mRNA expression was determined by RT-PCR. Anti-infliximab antibodies were purified from sera samples by affinity chromatography. RESULTS: IL-6, IL-8, GM-CSF and GRO-α were synergistically up-regulated in triple stimulation with TNF-α, IL-1ß and SAA, while their levels in solely SAA- or TNF-α-stimulated HCAEC did not increase. IL-1Ra, IL-1α, VCAM-1, MCP-1, IL-10 and IL-17A were increased, but no synergistic responses were observed in triple stimulation. Infliximab was effective in lowering the synergistic effect of IL-6, IL-8, GM-CSF and GRO-α in triple stimulation, while anti-infliximab antibodies restored the levels. The changes were confirmed at the mRNA expression level for IL-6, IL-8 and GM-CSF. CONCLUSIONS: Triple stimulation with TNF-α, IL-1ß and SAA synergistically elevated IL-6, IL-8, GM-CSF and GRO-α release in supernatants of HCAEC, with infliximab substantially inhibiting their levels. An isolated, enriched fraction of polyclonal anti-infliximab antibodies was capable of neutralizing infliximab, in the presence of TNF-α/IL-1ß/SAA. The long-term presence of anti-infliximab antibodies in the circulation of patients with chronic rheumatic diseases is potentially important for promoting the atherosclerotic process.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Vasos Coronarios/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Infliximab/inmunología , Inhibidores del Factor de Necrosis Tumoral/inmunología , Anticuerpos Neutralizantes/sangre , Células Cultivadas , Vasos Coronarios/inmunología , Vasos Coronarios/metabolismo , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-1beta/farmacología , Proteína Amiloide A Sérica/farmacología , Factor de Necrosis Tumoral alfa/farmacología
11.
Clin Rheumatol ; 38(2): 361-370, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30014359

RESUMEN

Therapeutic drug monitoring of TNF-alpha inhibitors is crucial for evaluating patients with inflammatory diseases on a personalized level. It has been clinically observed that many patients receiving TNF-alpha inhibitors, with negative drug and anti-drug antibody results from bridging ELISA (bELISA), lose their drug response over time, despite dose optimization. Our aims were to develop innovative in-house competitive ELISAs (cELISAs) for the detection of neutralizing antibodies against infliximab and adalimumab and compare their results to reporter gene assay (RGA) and in-house bELISA. Furthermore, we aimed to evaluate patient anti-drug antibody results in regard to their clinical records and potential benefits of therapeutic drug monitoring with the novel cELISAs. Sera of patients treated with infliximab (n = 46) or adalimumab (n = 31), having undetectable drug levels, were tested with our in-house cELISA. Briefly, samples were incubated with a fixed amount of drug and the neutralizing capacity of the samples was determined. The cELISA results were compared to RGA and bELISA results using Spearman's correlation coefficient. Additionally, patient clinical data were evaluated in line with the results of cELISA, bELISA, and RGA using the Kaplan-Meier analysis and the Log Rank test. Both anti-infliximab and anti-adalimumab cELISAs showed very good correlation to RGA (r = 0.932, p < 0.0001 and r = 0.947, p < 0.0001, respectively). Furthermore, a positive result in anti-infliximab cELISA can predict treatment failure in 100% of patients with negative bELISA, while a positive result in anti-adalimumab cELISA can predict treatment failure in 80% of patients with negative bELISA. Taken together, we developed innovative cELISAs enabling quantification of functional and neutralizing anti-drug antibodies, comparable to RGA. The association between cELISA results and loss of drug response in patients identified clinically important anti-drug antibodies, as measured by cELISA.


Asunto(s)
Adalimumab/inmunología , Anticuerpos Neutralizantes/sangre , Ensayo de Inmunoadsorción Enzimática , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Infliximab/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Adalimumab/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Monitoreo de Drogas , Femenino , Genes Reporteros , Humanos , Enfermedades Inflamatorias del Intestino/sangre , Infliximab/uso terapéutico , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Insuficiencia del Tratamiento , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adulto Joven
12.
Clin Rheumatol ; 38(2): 307-316, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30069799

RESUMEN

Giant cell arteritis (GCA) is a systemic vasculitis in individuals older than 50 years, characterized by headaches, visual disturbances, painful scalp, jaw claudication, impairment of limb arteries, and systemic inflammation, among other symptoms. GCA diagnosis is confirmed by a positive temporal artery biopsy (TAB) or by imaging modalities. A prominent acute phase response with inflammation is the hallmark of the disease, predominantly targeting large- and medium-sized arteries leading to stenosis or occlusion of arterial lumen. To date, there are no reliable tissue markers specific for GCA. Scarce reports have indicated the importance of epigenetics in GCA. The current systematic review reports significantly changed candidate biomarkers in TABs of GCA patients compared to non-GCA patients using qPCR.


Asunto(s)
Expresión Génica , Arteritis de Células Gigantes/diagnóstico , Arteritis de Células Gigantes/genética , MicroARNs/genética , Arterias Temporales/patología , Biomarcadores , Citocinas/metabolismo , Metilación de ADN , Epigénesis Genética , Arteritis de Células Gigantes/fisiopatología , Humanos
13.
PLoS One ; 13(4): e0195346, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29617422

RESUMEN

Serum amyloid A (SAA) is a sensitive inflammatory marker rapidly increased in response to infection, injury or trauma during the acute phase. Resolution of the acute phase and SAA reduction are well documented, however the exact mechanism remains elusive. Two inducible SAA proteins, SAA1 and SAA2, with their variants could contribute to systemic inflammation. While unconjugated human variant SAA1α is already commercially available, the variants of SAA2 are not. Antibodies against SAA have been identified in apparently healthy blood donors (HBDs) in smaller, preliminary studies. So, our objective was to detect anti-SAA and anti-SAA1α autoantibodies in the sera of 300 HBDs using ELISA, characterize their specificity and avidity. Additionally, we aimed to determine the presence of anti-SAA and anti-SAA1α autoantibodies in intravenous immunoglobulin (IVIg) preparations and examine their effects on released IL-6 from SAA/SAA1α-treated peripheral blood mononuclear cells (PBMCs). Autoantibodies against SAA and SAA1α had a median (IQR) absorbance OD (A450) of 0.655 (0.262-1.293) and 0.493 (0.284-0.713), respectively. Both anti-SAA and anti-SAA1α exhibited heterogeneous to high avidity and reached peak levels between 41-50 years, then diminished with age in the oldest group (51-67 years). Women consistently exhibited significantly higher levels than men. Good positive correlation was observed between anti-SAA and anti-SAA1α. Both anti-SAA and anti-SAA1α were detected in IVIg, their fractions subsequently isolated, and shown to decrease IL-6 protein levels released from SAA/SAA1α-treated PBMCs. In conclusion, naturally occurring antibodies against SAA and anti-SAA1α could play a physiological role in down-regulating their antigen and proinflammatory cytokines leading to the resolution of the acute phase and could be an important therapeutic option in patients with chronic inflammatory diseases.


Asunto(s)
Autoanticuerpos/sangre , Interleucina-6/metabolismo , Leucocitos Mononucleares/inmunología , Proteína Amiloide A Sérica/inmunología , Adolescente , Adulto , Anciano , Envejecimiento/sangre , Envejecimiento/inmunología , Células Cultivadas , Humanos , Persona de Mediana Edad , Adulto Joven
14.
Sci Rep ; 7(1): 4397, 2017 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-28667272

RESUMEN

Skin fibrosis in systemic sclerosis (SSc) is accompanied by attrition of dermal white adipose tissue (dWAT) and reduced levels of circulating adiponectin. Since adiponectin has potent regulatory effects on fibroblasts, we sought to assess adiponectin signaling in SSc skin biopsies, and evaluate fibrosis in mice with adiponectin gain- and loss-of-function mutations. Furthermore, we investigated the effects and mechanism of action of agonist peptides targeting adiponectin receptors in vitro and in vivo. We found that adiponectin pathway activity was significantly reduced in a subset of SSc skin biopsies. Mice lacking adiponectin mounted an exaggerated dermal fibrotic response, while transgenic mice with constitutively elevated adiponectin showed selective dWAT expansion and protection from skin and peritoneal fibrosis. Adiponectin receptor agonists abrogated ex vivo fibrotic responses in explanted normal and SSc fibroblasts and in 3D human skin equivalents, in part by attenuating focal adhesion complex assembly, and prevented and reversed experimentally-induced organ fibrosis in mice. These results implicate aberrant adiponectin pathway activity in skin fibrosis, identifying a novel function for this pleiotropic adipokine in regulation of tissue remodeling. Restoring adiponectin signaling in SSc patients therefore might represent an innovative pharmacological strategy for intractable organ fibrosis.


Asunto(s)
Adiponectina/metabolismo , Fibrosis/metabolismo , Fibrosis/patología , Adiponectina/farmacología , Animales , Biopsia , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis/tratamiento farmacológico , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Oligopéptidos/farmacología , Receptores de Adiponectina/metabolismo , Esclerodermia Sistémica/etiología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Transducción de Señal/efectos de los fármacos , Piel/metabolismo , Piel/patología
16.
Nat Commun ; 7: 11703, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27256716

RESUMEN

The factors responsible for maintaining persistent organ fibrosis in systemic sclerosis (SSc) are not known but emerging evidence implicates toll-like receptors (TLRs) in the pathogenesis of SSc. Here we show the expression, mechanism of action and pathogenic role of endogenous TLR activators in skin from patients with SSc, skin fibroblasts, and in mouse models of organ fibrosis. Levels of tenascin-C are elevated in SSc skin biopsy samples, and serum and SSc fibroblasts, and in fibrotic skin tissues from mice. Exogenous tenascin-C stimulates collagen gene expression and myofibroblast transformation via TLR4 signalling. Mice lacking tenascin-C show attenuation of skin and lung fibrosis, and accelerated fibrosis resolution. These results identify tenascin-C as an endogenous danger signal that is upregulated in SSc and drives TLR4-dependent fibroblast activation, and by its persistence impedes fibrosis resolution. Disrupting this fibrosis amplification loop might be a viable strategy for the treatment of SSc.


Asunto(s)
Colágeno/genética , Fibroblastos/metabolismo , Pulmón/patología , Esclerodermia Sistémica/genética , Piel/metabolismo , Tenascina/genética , Adulto , Anciano , Animales , Estudios de Casos y Controles , Diferenciación Celular , Células Cultivadas , Colágeno/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Fibrosis/genética , Fibrosis/metabolismo , Regulación de la Expresión Génica , Humanos , Pulmón/efectos de los fármacos , Masculino , Ratones , Persona de Mediana Edad , Miofibroblastos/efectos de los fármacos , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Transducción de Señal , Piel/patología , Tenascina/metabolismo , Tenascina/farmacología , Receptor Toll-Like 4/metabolismo , Regulación hacia Arriba
17.
Metabolomics ; 12: 92, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27330522

RESUMEN

INTRODUCTION: Human primary cells originating from different locations within the body could differ greatly in their metabolic phenotypes, influencing both how they act during physiological/pathological processes and how susceptible/resistant they are to a variety of disease risk factors. A novel way to monitor cellular metabolism is through cell energetics assays, so we explored this approach with human primary cell types, as models of sclerotic disorders. OBJECTIVES: In order to better understand pathophysiological processes at the cellular level, our goals were to measure metabolic pathway activities of endothelial cells and fibroblasts, and determine their metabolic phenotype profiles. METHODS: Biolog Phenotype MicroArray™ technology was used for the first time to characterize metabolic phenotypes of diverse primary cells. These colorimetric assays enable detection of utilization of 367 specific biochemical substrates by human endothelial cells from the coronary artery (HCAEC), umbilical vein (HUVEC) and normal, healthy lung fibroblasts (NHLF). RESULTS: Adenosine, inosine, d-mannose and dextrin were strongly utilized by all three cell types, comparable to glucose. Substrates metabolized solely by HCAEC were mannan, pectin, gelatin and prevalently tricarballylic acid. HUVEC did not show any uniquely metabolized substrates whereas NHLF exhibited strong utilization of sugars and carboxylic acids along with amino acids and peptides. CONCLUSION: Taken together, we show for the first time that this simple energetics assay platform enables metabolic characterization of primary cells and that each of the three human cell types examined gives a unique and distinguishable profile.

18.
Stem Cells Int ; 2016: 3585362, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26880955

RESUMEN

Mesenchymal stem cells (MSCs) are recognised as a promising tool to improve renal recovery in experimental models of cisplatin-induced acute kidney injury. However, these preclinical studies were performed on severely immunodeficient animals. Here, we investigated whether human umbilical cord derived MSC treatment could equally ameliorate acute kidney injury induced by cisplatin and prolong survival in mice with a normal immune system and those with a suppressed immune system by polyclonal antithymocyte globulin (ATG). We demonstrated that ATG pretreatment, when followed by MSC transplantation, significantly improved injured renal function parameters, as evidenced by decreased blood urea nitrogen and serum creatinine concentration, as well as improved renal morphology. This tissue restoration was also supported by increased survival of mice. The beneficial effects of ATG were associated with reduced level of inflammatory protein serum amyloid A3 and induced antioxidative expression of superoxide dismutase-1 (SOD-1), glutathione peroxidase (GPx), and hem oxygenase-1 (HO-1). Infused MSCs became localised predominantly in peritubular areas and acted to reduce renal cell death. In conclusion, these results show that ATG diminished in situ inflammation and oxidative stress associated with cisplatin-induced acute kidney injury, the effects that may provide more favourable microenvironment for MSC action, with consequential synergistic improvements in renal injury and animal survival as compared to MSC treatment alone.

19.
PLoS One ; 10(1): e0110820, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25629975

RESUMEN

Inflammation in systemic sclerosis (SSc) is a prominent, but incompletely characterized feature in early stages of the disease. The goal of these studies was to determine the circulating levels, clinical correlates and biological effects of the acute phase protein serum amyloid A (SAA), a marker of inflammation, in patients with SSc. Circulating levels of SAA were determined by multiplex assays in serum from 129 SSc patients and 98 healthy controls. Correlations between SAA levels and clinical and laboratory features of disease were analyzed. The effects of SAA on human pulmonary fibroblasts were studied ex vivo. Elevated levels of SAA were found in 25% of SSc patients, with the highest levels in those with early-stage disease and diffuse cutaneous involvement. Significant negative correlations of SAA were found with forced vital capacity and diffusion capacity for carbon monoxide. Patients with elevated SAA had greater dyspnea and more frequent interstitial lung disease, and had worse scores on patient-reported outcome measures. Incubation with recombinant SAA induced dose-dependent stimulation of IL-6 and IL-8 in normal lung fibroblasts in culture. Serum levels of the inflammatory marker SAA are elevated in patients with early diffuse cutaneous SSc, and correlate with pulmonary involvement. In lung fibroblasts, SAA acts as a direct stimulus for increased cytokine production. These findings suggest that systemic inflammation in SSc may be linked to lung involvement and SAA could serve as a potential biomarker for this complication.


Asunto(s)
Pulmón/metabolismo , Pulmón/patología , Esclerodermia Sistémica/sangre , Esclerodermia Sistémica/patología , Proteína Amiloide A Sérica/metabolismo , Biomarcadores/sangre , Sedimentación Sanguínea , Proteína C-Reactiva , Estudios de Casos y Controles , Fibroblastos/metabolismo , Humanos , Interleucina-6/metabolismo , Pulmón/fisiopatología , Evaluación del Resultado de la Atención al Paciente , Pruebas de Función Respiratoria , Esclerodermia Sistémica/epidemiología , Autoinforme
20.
Mediators Inflamm ; 2014: 876395, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24782597

RESUMEN

Serum amyloid A (SAA) production is increased by inflamed arthritic synovial tissue, where it acts as a cytokine/chemoattractant for inflammatory and immune cells and as an inducer of matrix degrading enzymes. SAA has been shown to bind lipoxin A4 receptor, a member of the formyl-peptide related 2 G-protein coupled receptor family (ALX) and elicit proinflammatory activities in human primary fibroblast-like synoviocytes (FLS). We report on the identification of uteroglobin, a small globular protein with potent anti-inflammatory activities, as a possible ligand of ALX. Uteroglobin-specific association with ALX was demonstrated by an enzyme immunoassay experiment employing a cell line engineered to express the human ALX receptor. Uteroglobin's interaction with ALX resulted in the inhibition of SAA responses, such as attenuation of phospholipase A2 activation and cellular chemotaxis. In FLS, uteroglobin showed an antagonism against SAA-induced interleukin-8 release and decreased cell migration. These novel roles described for uteroglobin via ALX may help elucidate genetic and clinical observations indicating that a polymorphism in the uteroglobin promoter is linked to disease outcome, specifically prediction of bone erosion in patients with rheumatoid arthritis or severity of IgA glomerulonephritis and sarcoidosis.


Asunto(s)
Receptores de Lipoxina/metabolismo , Proteína Amiloide A Sérica/metabolismo , Uteroglobina/metabolismo , Animales , Células CHO , Movimiento Celular , Quimiotaxis , Cricetulus , Endometrio/citología , Femenino , Fibroblastos/citología , Células HL-60 , Humanos , Técnicas para Inmunoenzimas , Inflamación , Ligandos , Fosfolipasas A2/metabolismo , Regiones Promotoras Genéticas , Membrana Sinovial/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA