Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Intervalo de año de publicación
1.
Biomolecules ; 11(10)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34680053

RESUMEN

Analysis of liver biopsy specimens showed that SARS-CoV-2 might have led to liver damage. This study aimed to evaluate the role of selected hepatokines and myokines in the development and progression of COVID-19. Seventy patients with laboratory-confirmed COVID-19 and 20 healthy volunteers were enrolled in the study. Irisin, pentraxin 3, fetuin-A, and FGF-21 serum concentrations and biochemical parameters were assessed using an immunoenzymatic method with commercially available enzyme immunoassay (EIA) or enzyme-linked immunosorbent assay (ELISA) kits. Serum fetuin-A concentrations were significantly decreased in COVID-19 patients compared to healthy volunteers. The serum concentration of FGF-21 was significantly increased in obese COVID-19 patients compared to overweight ones. Moreover, the FGF-21 level was higher in COVID-19 patients diagnosed with metabolic syndrome than in patients without metabolic syndrome. PTX3 concentration was higher in COVID-19 patients with higher HOMA-IR values than those with lower HOMA-IR values. COVID-19 patients with HOMA-IR ≤ 3 and >3 had significantly lower fetuin-A levels than the control group. Irisin concentration was significantly decreased in the HOMA-IR ≤ 3 COVID-19 subgroup when comparing with the control group. Lower levels of fetuin-A observed in COVID-19 patients despite higher HOMA-IR, CRP, and ferritin levels, pneumonia, patients requiring ICU care suggests that fetuin-A deficiency predisposes to more severe COVID-19 course. Upregulated pentraxin 3 may be used as a potential predictor of COVID-19 severity.


Asunto(s)
COVID-19/metabolismo , alfa-2-Glicoproteína-HS/metabolismo , Animales , COVID-19/patología , Masculino , Ratas , Ratas Wistar , alfa-2-Glicoproteína-HS/deficiencia
2.
PLoS One ; 15(2): e0228503, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32074120

RESUMEN

The plasma protein fetuin-A mediates the formation of protein-mineral colloids known as calciprotein particles (CPP)-rapid clearance of these CPP by the reticuloendothelial system prevents errant mineral precipitation and therefore pathological mineralization (calcification). The mutant mouse strain D2,Ahsg-/- combines fetuin-A deficiency with the calcification-prone DBA/2 genetic background, having a particularly severe compound phenotype of microvascular and soft tissue calcification. Here we studied mechanisms leading to soft tissue calcification, organ damage and death in these mice. We analyzed mice longitudinally by echocardiography, X-ray-computed tomography, analytical electron microscopy, histology, mass spectrometry proteomics, and genome-wide microarray-based expression analyses of D2 wildtype and Ahsg-/- mice. Fetuin-A-deficient mice had calcified lesions in myocardium, lung, brown adipose tissue, reproductive organs, spleen, pancreas, kidney and the skin, associated with reduced growth, cardiac output and premature death. Importantly, early-stage calcified lesions presented in the lumen of the microvasculature suggesting precipitation of mineral containing complexes from the fluid phase of blood. Genome-wide expression analysis of calcified lesions and surrounding (not calcified) tissue, together with morphological observations, indicated that the calcification was not associated with osteochondrogenic cell differentiation, but rather with thrombosis and fibrosis. Collectively, these results demonstrate that soft tissue calcification can start by intravascular mineral deposition causing microvasculopathy, which impacts on growth, organ function and survival. Our study underscores the importance of fetuin-A and related systemic regulators of calcified matrix metabolism to prevent cardiovascular disease, especially in dysregulated mineral homeostasis.


Asunto(s)
Calcinosis/complicaciones , Calcinosis/genética , Microvasos/patología , Insuficiencia Multiorgánica/genética , Calcificación Vascular/genética , alfa-2-Glicoproteína-HS/genética , Animales , Calcinosis/metabolismo , Calcinosis/patología , Calcio/metabolismo , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Microcirculación/fisiología , Microvasos/metabolismo , Minerales/metabolismo , Sistema Mononuclear Fagocítico/metabolismo , Sistema Mononuclear Fagocítico/patología , Insuficiencia Multiorgánica/patología , Calcificación Vascular/metabolismo , Calcificación Vascular/patología , alfa-2-Glicoproteína-HS/deficiencia
3.
Biochem Pharmacol ; 166: 46-55, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31077645

RESUMEN

Acetaminophen (APAP)-induced liver injury (AILI) is initiated by the generation of a reactive metabolite and ultimately leads to hepatocyte necrosis. Necrotic cells secrete damage-associated molecular patterns that activate hepatic nonparenchymal cells and induce an inflammatory response. Fetuin-A is a hepatokine with reported involvement in low-grade inflammation in many diseases, due to acting as an endogenous ligand for TLR4. However, little is known about the role of fetuin-A in AILI. In this study, we showed that fetuin-A is involved in the aggravation of hepatotoxicity during the initial phase of AILI progression. Treatment with APAP increased the expression and serum levels of fetuin-A in mice. Fetuin-A upregulated transcription of pro-inflammatory cytokines and chemokines through activation of TLR4 and also increased monocyte infiltration into the liver, leading to necroinflammatory reactions in AILI. However, these reactions were attenuated with the silencing of fetuin-A using adenoviral shRNA. As a result, mice with silenced fetuin-A exhibited less centrilobular necrosis and liver injury compared to controls in response to APAP. In conclusion, our results suggest that fetuin-A is an important hepatokine that mediates the hepatotoxicity of APAP through production of chemokines and thus regulates the infiltration of monocytes into the liver, a critical event in the inflammatory response during the initial phase of AILI. Our results indicate that a strategy based on the antagonism of fetuin-A may be a novel therapeutic approach to the treatment of acetaminophen-induced acute liver failure.


Asunto(s)
Acetaminofén/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Receptor Toll-Like 4/metabolismo , alfa-2-Glicoproteína-HS/deficiencia , Animales , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , alfa-2-Glicoproteína-HS/genética
4.
PLoS One ; 12(10): e0187030, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29088242

RESUMEN

Fetuin-A / α2-Heremans-Schmid-glycoprotein (gene name Ahsg) is a systemic inhibitor of ectopic calcification. Due to its high affinity for calcium phosphate, fetuin-A is highly abundant in mineralized bone matrix. Foreshortened femora in fetuin-A-deficient Ahsg-/- mice indicated a role for fetuin-A in bone formation. We studied early postnatal bone development in fetuin-A-deficient mice and discovered that femora from Ahsg-/- mice exhibited severely displaced distal epiphyses and deformed growth plates, similar to the human disease slipped capital femoral epiphysis (SCFE). The growth plate slippage occurred in 70% of Ahsg-/- mice of both sexes around three weeks postnatal. At this time point, mice weaned and rapidly gained weight and mobility. Epiphysis slippage never occurred in wildtype and heterozygous Ahsg+/- mice. Homozygous fetuin-A-deficient Ahsg-/- mice and, to a lesser degree, heterozygous Ahsg+/- mice showed lesions separating the proliferative zone from the hypertrophic zone of the growth plate. The hypertrophic growth plate cartilage in long bones from Ahsg-/- mice was significantly elongated and V-shaped until three weeks of age and thus prior to the slippage. Genome-wide transcriptome analysis of laser-dissected distal femoral growth plates from 13-day-old Ahsg-/- mice revealed a JAK-STAT-mediated inflammatory response including a 550-fold induction of the chemokine Cxcl9. At this stage, vascularization of the elongated growth plates was impaired, which was visualized by immunofluorescence staining. Thus, fetuin-A-deficient mice may serve as a rodent model of growth plate pathologies including SCFE and inflammatory cartilage degradation.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Epífisis Desprendida/genética , Fémur/anomalías , Miembro Posterior/anomalías , alfa-2-Glicoproteína-HS/genética , Animales , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica/métodos , Placa de Crecimiento/anomalías , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Destete , alfa-2-Glicoproteína-HS/deficiencia
5.
Am J Physiol Renal Physiol ; 309(8): F744-54, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26180236

RESUMEN

Although dietary phosphate restriction is important for treating hyperphosphatemia in patients with chronic kidney disease, it remains unclear whether a low-protein diet (LPD), which contains low phosphate, has beneficial effects on malnutrition, inflammation, and vascular calcification. The effects of LPD on inflammation, malnutrition, and vascular calcification were therefore assessed in rats. Rats were fed a normal diet or diets containing 0.3% adenine and low/normal protein and low/high phosphate. After 6 wk, serum and urinary biochemical parameters, systemic inflammation, and vascular calcification were examined. The protective effect of fetuin-A and albumin were assessed in cultured vascular smooth muscle cells. Rats fed the diet containing 0.3% adenine developed severe azotemia. LPD in rats fed high phosphate induced malnutrition (decreases in body weight, food intake, serum albumin and fetuin-A levels, and urinary creatinine excretion) and systemic inflammation (increases in serum tumor necrosis factor-α and urinary oxidative stress marker). LPD decreased the serum fetuin-A level and fetuin-A synthesis in the liver and increased serum calcium-phosphate precipitates. A high-phosphate diet increased aortic calcium content, which was enhanced by LPD. Reduced fetal calf serum in the medium of cultured vascular smooth muscle cells enhanced phosphate-induced formation of calcium-phosphate precipitates in the media and calcification of vascular smooth muscle cells, both of which were prevented by fetuin-A administration. Our results suggest that phosphate restriction by restricting dietary protein promotes vascular calcification by lowering the systemic fetuin-A level and increasing serum calcium-phosphate precipitates and induces inflammation and malnutrition in uremic rats fed a high-phosphate diet.


Asunto(s)
Dieta con Restricción de Proteínas , Hiperfosfatemia/complicaciones , Uremia/complicaciones , Calcificación Vascular/etiología , alfa-2-Glicoproteína-HS/metabolismo , Albúminas/farmacología , Animales , Fosfatos de Calcio/metabolismo , Células Cultivadas , Hiperfosfatemia/metabolismo , Masculino , Músculo Liso Vascular/efectos de los fármacos , Fósforo Dietético/farmacología , Desnutrición Proteico-Calórica/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo , Uremia/metabolismo , Calcificación Vascular/metabolismo , alfa-2-Glicoproteína-HS/deficiencia , alfa-2-Glicoproteína-HS/farmacología
6.
J Am Soc Nephrol ; 23(10): 1744-52, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22956818

RESUMEN

Vascular and soft tissue calcification contributes to cardiovascular morbidity and mortality in both the general population and CKD. Because calcium and phosphate serum concentrations are near supersaturation, the balance of inhibitors and promoters critically influences the development of calcification. An assay that measures the overall propensity for calcification to occur in serum may have clinical use. Here, we describe a nanoparticle-based assay that detects, in the presence of artificially elevated calcium and phosphate concentrations, the spontaneous transformation of spherical colloidal primary calciprotein particles (CPPs) to elongate crystalline secondary CPPs. We used characteristics of this transition to describe the intrinsic capacity of serum to inhibit the precipitation of calcium and phosphate. Using this assay, we found that both the sera of mice deficient in fetuin-A, a serum protein that inhibits calcification, and the sera of patients on hemodialysis have reduced intrinsic properties to inhibit calcification. In summary, we developed a nanoparticle-based test that measures the overall propensity for calcification in serum. The clinical use of the test requires evaluation in a prospective study.


Asunto(s)
Calcinosis/sangre , Calcio/sangre , Nanopartículas , Nanotecnología/métodos , Animales , Estudios de Casos y Controles , Precipitación Química , Coloides , Cristalización , Humanos , Luz , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Nefelometría y Turbidimetría , Fosfatos/sangre , Diálisis Renal , Dispersión de Radiación , Solubilidad , alfa-2-Glicoproteína-HS/deficiencia , alfa-2-Glicoproteína-HS/genética
7.
Joint Bone Spine ; 78(6): 561-7, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21550285

RESUMEN

Extracellular matrix mineralization is important for mechanical stability of the skeleton and for calcium and phosphate storage. Professional mineral-disposing cell types are hypertrophic chondrocytes, odontoblasts, ameloblasts and osteoblasts. Since ectopic mineralization causes tissue dysfunction mineralization inhibitors and promoting factors have to be kept in close balance. The most prominent inhibitors are fetuin-A, matrix-Gla-protein (MGP), SIGBLING proteins and pyrophosphate. In spite of their ubiquitous presence, their loss entails a specific rather than a stereotypic pattern of ectopic mineralization. Typical sites of pathological mineral accumulation are connective tissues, articular cartilage, and vessels. Associated common human pathologies are degenerative joint disorders and arteriosclerosis. This article gives a summary on what we have learned from different mouse models with pathologic mineralization phenotypes about the role of these inhibitors and the regulation of mineralization promoting factors.


Asunto(s)
Enfermedades Óseas Metabólicas/fisiopatología , Calcificación Fisiológica/fisiología , Modelos Animales de Enfermedad , Fenotipo , Animales , Enfermedades Óseas Metabólicas/metabolismo , Fosfatos de Calcio/metabolismo , Glucuronidasa/deficiencia , Glucuronidasa/metabolismo , Proteínas Klotho , Ratones , Ratones Noqueados , Osteocalcina/deficiencia , Osteocalcina/metabolismo , Osteoprotegerina/deficiencia , Osteoprotegerina/metabolismo , alfa-2-Glicoproteína-HS/deficiencia , alfa-2-Glicoproteína-HS/metabolismo
8.
PLoS One ; 6(2): e16945, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21347455

RESUMEN

BACKGROUND: A liver-derived protein, fetuin-A, was first purified from calf fetal serum in 1944, but its potential role in lethal systemic inflammation was previously unknown. This study aims to delineate the molecular mechanisms underlying the regulation of hepatic fetuin-A expression during lethal systemic inflammation (LSI), and investigated whether alterations of fetuin-A levels affect animal survival, and influence systemic accumulation of a late mediator, HMGB1. METHODS AND FINDINGS: LSI was induced by endotoxemia or cecal ligation and puncture (CLP) in fetuin-A knock-out or wild-type mice, and animal survival rates were compared. Murine peritoneal macrophages were challenged with exogenous (endotoxin) or endogenous (IFN-γ) stimuli in the absence or presence of fetuin-A, and HMGB1 expression and release was assessed. Circulating fetuin-A levels were decreased in a time-dependent manner, starting between 26 h, reaching a nadir around 24-48 h, and returning towards base-line approximately 72 h post onset of endotoxemia or sepsis. These dynamic changes were mirrored by an early cytokine IFN-γ-mediated inhibition (up to 50-70%) of hepatic fetuin-A expression. Disruption of fetuin-A expression rendered animals more susceptible to LSI, whereas supplementation of fetuin-A (20-100 mg/kg) dose-dependently increased animal survival rates. The protection was associated with a significant reduction in systemic HMGB1 accumulation in vivo, and parallel inhibition of IFN-γ- or LPS-induced HMGB1 release in vitro. CONCLUSIONS: These experimental data suggest that fetuin-A is protective against lethal systemic inflammation partly by inhibiting active HMGB1 release.


Asunto(s)
Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , alfa-2-Glicoproteína-HS/metabolismo , Proteínas de Fase Aguda/metabolismo , Animales , Línea Celular , Citocinas/metabolismo , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Susceptibilidad a Enfermedades , Endotoxemia/metabolismo , Endotoxemia/patología , Endotoxemia/prevención & control , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Proteína HMGB1/metabolismo , Hígado , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Síndrome de Respuesta Inflamatoria Sistémica/patología , Síndrome de Respuesta Inflamatoria Sistémica/prevención & control , Factores de Tiempo , alfa-2-Glicoproteína-HS/deficiencia , alfa-2-Glicoproteína-HS/genética , alfa-2-Glicoproteína-HS/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA