Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pflugers Arch ; 476(5): 735-753, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38424322

RESUMEN

Genetic variants of gene SCN5A encoding the alpha-subunit of cardiac voltage-gated sodium channel Nav1.5 are associated with various diseases, including long QT syndrome (LQT3), Brugada syndrome (BrS1), and progressive cardiac conduction disease (PCCD). In the last decades, the great progress in understanding molecular and biophysical mechanisms of these diseases has been achieved. The LQT3 syndrome is associated with gain-of-function of sodium channels Nav1.5 due to impaired inactivation, enhanced activation, accelerated recovery from inactivation or the late current appearance. In contrast, BrS1 and PCCD are associated with the Nav1.5 loss-of-function, which in electrophysiological experiments can be manifested as reduced current density, enhanced fast or slow inactivation, impaired activation, or decelerated recovery from inactivation. Genetic variants associated with congenital arrhythmias can also disturb interactions of the Nav1.5 channel with different proteins or drugs and cause unexpected reactions to drug administration. Furthermore, mutations can affect post-translational modifications of the channels and their sensitivity to pH and temperature. Here we briefly review the current knowledge on biophysical mechanisms of LQT3, BrS1 and PCCD. We focus on limitations of studies that use heterologous expression systems and induced pluripotent stem cells (iPSC) derived cardiac myocytes and summarize our understanding of genotype-phenotype relations of SCN5A mutations.


Asunto(s)
Canalopatías , Canal de Sodio Activado por Voltaje NAV1.5 , Humanos , Animales , Canalopatías/genética , Canalopatías/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Miocardio/metabolismo , Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Síndrome de QT Prolongado/fisiopatología
2.
Cardiology ; 147(1): 35-46, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34628415

RESUMEN

BACKGROUND: Brugada syndrome (BrS) is a rare inherited cardiac arrhythmia with increased risk of sudden cardiac death. Mutations in gene SCN5A, which encodes the α-subunit of cardiac voltage-gated sodium channel NaV1.5, have been identified in over 20% of patients with BrS. However, only a small fraction of NaV1.5 variants, which are associated with BrS, are characterized in electrophysiological experiments. RESULTS: Here we explored variants V281A and L1582P, which were found in our patients with BrS, and variants F543L and K1419E, which are reportedly associated with BrS. Heterologous expression of the variants in CHO-K1 cells and the Western blot analysis demonstrated that each variant appeared at the cell surface. We further measured sodium current in the whole-cell voltage clamp configuration. Variant F543L produced robust sodium current with a hyperpolarizing shift in the voltage dependence of steady-state fast inactivation. Other variants did not produce detectable sodium currents, indicating a complete loss of function. In a recent cryoEM structure of the hNaV1.5 channel, residues V281, K1419, and L1582 are in close contacts with residues whose mutations are reportedly associated with BrS, indicating functional importance of respective contacts. CONCLUSIONS: Our results support the notion that loss of function of NaV1.5 or decrease of the channel activity is involved in the pathogenesis of BrS.


Asunto(s)
Síndrome de Brugada , Canal de Sodio Activado por Voltaje NAV1.5 , Síndrome de Brugada/genética , Humanos , Mutación , Canal de Sodio Activado por Voltaje NAV1.5/genética
3.
Am J Hum Genet ; 102(5): 760-775, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29706349

RESUMEN

Finland provides unique opportunities to investigate population and medical genomics because of its adoption of unified national electronic health records, detailed historical and birth records, and serial population bottlenecks. We assembled a comprehensive view of recent population history (≤100 generations), the timespan during which most rare-disease-causing alleles arose, by comparing pairwise haplotype sharing from 43,254 Finns to that of 16,060 Swedes, Estonians, Russians, and Hungarians from geographically and linguistically adjacent countries with different population histories. We find much more extensive sharing in Finns, with at least one ≥ 5 cM tract on average between pairs of unrelated individuals. By coupling haplotype sharing with fine-scale birth records from more than 25,000 individuals, we find that although haplotype sharing broadly decays with geographical distance, there are pockets of excess haplotype sharing; individuals from northeast Finland typically share several-fold more of their genome in identity-by-descent segments than individuals from southwest regions. We estimate recent effective population-size changes through time across regions of Finland, and we find that there was more continuous gene flow as Finns migrated from southwest to northeast between the early- and late-settlement regions than was dichotomously described previously. Lastly, we show that haplotype sharing is locally enriched by an order of magnitude among pairs of individuals sharing rare alleles and especially among pairs sharing rare disease-causing variants. Our work provides a general framework for using haplotype sharing to reconstruct an integrative view of recent population history and gain insight into the evolutionary origins of rare variants contributing to disease.


Asunto(s)
Enfermedad/genética , Genética de Población , Haplotipos/genética , Finlandia , Flujo Génico , Variación Genética , Geografía , Migración Humana , Humanos , Parto , Densidad de Población , Factores de Tiempo
4.
Int J Mol Sci ; 22(14)2021 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-34298968

RESUMEN

Mitochondrial dysfunction is considered the major contributor to skeletal muscle wasting in different conditions. Genetically determined neuromuscular disorders occur as a result of mutations in the structural proteins of striated muscle cells and therefore are often combined with cardiac phenotype, which most often manifests as a cardiomyopathy. The specific roles played by mitochondria and mitochondrial energetic metabolism in skeletal muscle under muscle-wasting conditions in cardiomyopathies have not yet been investigated in detail, and this aspect of genetic muscle diseases remains poorly characterized. This review will highlight dysregulation of mitochondrial representation and bioenergetics in specific skeletal muscle disorders caused by mutations that disrupt the structural and functional integrity of muscle cells.


Asunto(s)
Cardiomiopatías/genética , Corazón/fisiopatología , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Enfermedades Neuromusculares/genética , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Modelos Animales de Enfermedad , Metabolismo Energético , Humanos , Ratones , Mitocondrias Cardíacas/metabolismo , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Proteínas Musculares/fisiología , Músculo Esquelético/ultraestructura , Atrofia Muscular/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Enfermedades Neuromusculares/metabolismo , Enfermedades Neuromusculares/patología , Fenotipo
5.
Cell Physiol Biochem ; 54(4): 696-706, 2020 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-32706220

RESUMEN

BACKGROUND/AIMS: Mutations of desmosomal genes are known to cause arrhythmogenic cardiomyopathy characterized by arrhythmias and sudden cardiac death. Previously, we described a novel genetic variant H1684R in desmoplakin gene (DSP), associated with a progressive cardiac conduction disease (PCCD). In the present study, we aimed to investigate an effect of the DSP-H1684R genetic variant on the activity of ion channels. METHODS: We used cardiomyocytes derived from induced pluripotent stem cells (iPSC cardiomyocytes) from a patient with DSP-H1684R genetic variant and from two healthy donors. Immunofluorescent staining and western blot analyses were used to characterize patient-specific cardiomyocytes. By the whole-cell voltage-clamp technique we estimated the activity of voltage-gated sodium, calcium, and potassium channels that are responsible for action potential generation and its shape. Action potentials' parameters were measured using whole-cell current-clamp technique. RESULTS: In patient-specific cardiomyocytes we observed both lower amplitudes of currents through sodium Nav1.5 channels and L-type calcium channels, but higher amplitude of current through transient-outward potassium channels in comparison to donor cardiomyocytes. Current-clamp measurements revealed shortening of action-potential in DSP-H1684R-carrying iPSC cardiomyocytes. Therefore, observed alterations in the channels activity might have a great impact on the properties of action potential and development of PCCD. CONCLUSION: Our results show that desmoplakin genetic variants, besides conduction slowing caused by structural heart remodeling, could affect multiple ion channel activity aggravating arrhythmia manifestation in PCCD.


Asunto(s)
Trastorno del Sistema de Conducción Cardíaco/genética , Desmoplaquinas/genética , Bloqueo Cardíaco/genética , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Canales Iónicos/fisiología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Potenciales de Acción/fisiología , Canales de Calcio/fisiología , Trastorno del Sistema de Conducción Cardíaco/metabolismo , Desmoplaquinas/metabolismo , Técnica del Anticuerpo Fluorescente , Bloqueo Cardíaco/metabolismo , Humanos , Canales Iónicos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje/fisiología , Canales de Sodio Activados por Voltaje/fisiología
6.
Cell Tissue Res ; 379(1): 169-179, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31781870

RESUMEN

Osteogenic differentiation is a tightly regulated process realized by progenitor cell osteoblasts. Notch signaling pathway plays a critical role in skeletal development and bone remodeling. Controversial data exist regarding the role of Notch activation in promoting or preventing osteogenic differentiation. This study aims to investigate the effect of several Notch components and their dosage on osteogenic differentiation of mesenchymal stem cells of adipose tissue. Osteogenic differentiation was induced in the presence of either of Notch components (NICD, Jag1, Dll1, Dll4) dosed by lentiviral transduction. We show that osteogenic differentiation was increased by NICD and Jag1 transduction in a dose-dependent manner; however, a high dosage of both NICD and Jag1 decreased the efficiency of osteogenic differentiation. NICD dose-dependently increased activity of the CSL luciferase reporter but a high dosage of NICD caused a decrease in the activity of the reporter. A high dosage of both Notch components NICD and Jag1 induced apoptosis. In co-culture experiments where only half of the cells were transduced with either NICD or Jag1, only NICD increased osteogenic differentiation according to the dosage, while Jag1-transduced cells differentiated almost equally independently on dosage. In conclusion, activation of Notch promotes osteogenic differentiation in a tissue-specific dose-dependent manner; both NICD and Jag1 are able to increase osteogenic potential but at moderate doses only and a high dosage of Notch activation is detrimental to osteogenic differentiation. This result might be especially important when considering possibilities of using Notch activation to promote osteogenesis in clinical applications to bone repair.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Osteogénesis , Receptores Notch/fisiología , Comunicación Celular , Células Cultivadas , Técnicas de Cocultivo , Humanos , Proteína Jagged-1/genética , Proteína Jagged-1/fisiología , Lentivirus/genética , Osteogénesis/genética , Receptores Notch/genética , Transducción Genética
7.
Pediatr Res ; 88(1): 38-47, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31952074

RESUMEN

BACKGROUND: Tetralogy of Fallot (TF) is a severe congenital defect of heart development. Fine-tuned sequential activation of Notch signaling genes is responsible for proper heart chamber development. Mutations in Notch genes have been associated with TF. The aim of this study was to analyze the activity of the Notch pathway in cardiac mesenchymal cells derived from ventricular tissue of TF patients. METHODS: Cardiac mesenchymal cells were isolated from 42 TF patients and from 14 patients with ventricular septal defects (VSDs), used as a comparison group. The Notch pathway was analyzed by estimating the expression of Notch-related genes by qPCR. Differentiation and proliferation capacity of the cells was estimated. RESULTS: The TF-derived cells demonstrated a dysregulated pattern of Notch-related gene expression comparing to VSD-derived cells. Correlation of Notch signaling activation level by HEY1/HES1 expression level with proliferation and cardiogenic-like differentiation of cardiac mesenchymal cells was observed but not with clinical parameters nor with the age of the patients. CONCLUSIONS: The data suggest a contribution of dysregulated Notch signaling to the pathogenesis of tetralogy of Fallot and importance of Notch signaling level for the functional state of cardiac mesenchymal cells, which could be critical considering these cells for potential cell therapy approaches.


Asunto(s)
Defectos del Tabique Interventricular/metabolismo , Células Madre Mesenquimatosas/citología , Miocardio/metabolismo , Receptores Notch/metabolismo , Tetralogía de Fallot/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Proliferación Celular , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Corazón/fisiopatología , Ventrículos Cardíacos/metabolismo , Humanos , Hipoxia , Inmunofenotipificación , Lactante , Recién Nacido , Masculino , Mutación , Transducción de Señal , Factor de Transcripción HES-1/metabolismo
8.
Cardiology ; 145(11): 746-756, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33049752

RESUMEN

INTRODUCTION: Left ventricular non-compaction (LVNC) represents a genetically heterogeneous cardiomyopathy which occurs in both children and adults. Its genetic spectrum overlaps with other types of cardiomyopathy. However, LVNC phenotypes in different age groups can have distinct genetic aetiologies. The aim of the study was to decipher the genetic spectrum of LVNC presented in childhood. Patient Group and Methods: Twenty patients under the age of 18 years diagnosed with LVNC were enrolled in the study. Target sequencing and whole-exome sequencing were performed using a panel of 108 cardiomyopathy-associated genes. Pathogenic, likely pathogenic, and variants of unknown significance found in genes highly expressed in cardiomyocytes were considered as variants of interest for further analysis. RESULTS: The median age at presentation was 8.0 (0.1-17) years, with 6 patients presenting before 1 year of age. Twelve (60%) patients demonstrated reduced ejection fraction. Right ventricular (RV) dilation was registered in 6 (30%), often in combination with reduced RV contractility (25%). Almost half (45%) of the patients demonstrated biventricular involvement already at disease presentation. For pathogenic and likely pathogenic variants, the positive genotyping rate was 45%, and these variants were found mainly in non-contractile structural sarcomeric genes (ACTN2, MYPN, and TTN) or in metabolic and signal transduction genes (BRAF and TAZ). Likely pathogenic TAZ variants were detected in all 5 patients suspected of having Barth syndrome. No pathogenic or likely pathogenic variants were found in genes encoding for sarcomeric contractile proteins, but variants of unknown significance were detected in 3 out of 20 patients (MYH6, MYH7, and MYLK2). In 4 patients, variants of unknown significance in ion-channel genes were detected. CONCLUSION: We detected a low burden of contractile sarcomeric variants in LVNC patients presenting below the age of 18 years, with the major number of variants residing in non-contractile structural sarcomeric genes. The identification of the variants in ion-channel and related genes not previously associated with LVNC in paediatric patients requires further examination of their functional role.


Asunto(s)
Cardiomiopatías , Cardiopatías Congénitas , Adolescente , Cardiomiopatías/genética , Niño , Ventrículos Cardíacos , Humanos , Mutación , Fenotipo
9.
Platelets ; 31(2): 226-235, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30977703

RESUMEN

Platelet concentrates are used in clinic for therapy and prophylaxis of conditions associated with platelet deficiency or malfunction. The characteristics of platelet concentrates gradually change during pretransfusion storage, affecting their clinical effectiveness and the risk of adverse transfusion reactions. The presence of platelet-derived membrane vesicles is an important characteristic of platelet concentrates. Due to their functionality, changes in the number and molecular compositions of platelet-derived vesicles have major effects on the clinical properties of platelet preparations. The existence of different subpopulations of membrane vesicles requires analytical methods capable of providing information at the individual vesicle level. Such methods include flow cytometry and electron microscopy. However, conventional flow cytometry has certain limitations, since the diameters of many platelet-derived membrane vesicles are smaller than its detection limit. The use of classical scanning electron microscopy is also limited due to the requirement for coating with a layer of conductive material, which impedes the detection of small extracellular vesicles. Here, a combination of high-sensitivity flow cytometry and low-voltage scanning electron microscopy was used to increase sensitivity and resolution in the detection of nanosized objects present in platelet concentrates during storage. Apheresis platelet concentrates from eight healthy adult donors were investigated on days 2 and 7 of storage. Fractions of nanosized objects were obtained by differential centrifugation. Fluorophore-conjugated antibodies were used to detect marker-positive vesicles derived from platelets (CD41), red blood cells (CD235a), leukocytes (CD45), and endothelial cells (VEGFR2). Near-spherical objects with diameters ranging from 25 to 700 nm were observed by low-voltage scanning electron microscopy in platelet concentrates and its fractions. On day 7 of storage, objects with diameters of less than 100 nm were attached to and clustered near the terminal ends of pseudopod-like projections. High-sensitivity flow cytometry showed that during storage numbers of CD41(pos) vesicles elevated more than fivefold and numbers of marker-negative nanosized objects, which did not carry any of the investigated cell type-specific markers elevated more than twofold. Major changes in both CD41(pos) vesicles and marker-negative nanosized objects abundances were observed for objects with diameters around 100 nm bead equivalents. Overall, these results emphasized the importance of application of high-sensitivity methods for monitoring the characteristics of cell-derived nanosized objects during platelet concentrate storage.


Asunto(s)
Plaquetas/ultraestructura , Conservación de la Sangre , Citometría de Flujo , Vesículas Extracelulares/ultraestructura , Humanos , Masculino , Microscopía Electrónica de Rastreo , Nanoestructuras/ultraestructura , Plasma/citología , Plasma/metabolismo , Transfusión de Plaquetas , Plasma Rico en Plaquetas/citología , Plasma Rico en Plaquetas/metabolismo , Plaquetoferesis , Factores de Tiempo
10.
Biochem Biophys Res Commun ; 514(2): 462-468, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31056255

RESUMEN

Cardiovascular calcification is one of the leading reasons of morbidity and mortality in Western countries and has many similarities to osteogenesis. The role of smooth muscle calcific transformation is well established for atherogenic lesions, but mechanisms driving initial stages of proosteogenic cell fate commitment in big vessels remain poorly understood. The role of endothelial and underlying interstitial cell interaction in driving cellular decisions is emerging from recent studies. The aim of this study was to analyze co-culture of endothelial and smooth muscle cells in vitro in acquiring proosteogenic phenotype. We co-cultured human aortic endothelial cells (EC) and human aortic smooth muscle cells (SMC) and analyzed osteogenic phenotype by ALP staining and proosteogenic gene expression by qPCR in co-cultures and in separate cellular types after magnetic CD31-sorting. In EC and SMC co-cultures osteogenic phenotype was induced as well as activated expression of RUNX2, POSTIN, BMP2/4, SOX5, COL1A SMC; co-culture of EC with SMC induced NOTCH1, NOTCH3, NOTCH4 and HEY1 expression; Notch activation by lentiviral activated Notch intracellular domain induced expression of RUNX2, OPN, POSTIN in SMC; NOTCH1 and NOTCH3 and HEY1 were selectively induced in EC during co-culture. We conclude that endothelial cells are capable of driving smooth muscle calcification via cell-cell contact and activation of Notch signaling.


Asunto(s)
Aorta/citología , Calcinosis , Células Endoteliales/citología , Células Endoteliales/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Osteogénesis , Receptores Notch/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Perfilación de la Expresión Génica , Humanos , Transducción de Señal
11.
Biochem Biophys Res Commun ; 516(3): 777-783, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31253402

RESUMEN

Mutations in gene SCN5A, which encodes cardiac voltage-gated sodium channel Nav1.5, are associated with multiple clinical phenotypes. Here we describe a novel A1294G genetic variant detected in a male patient with combined clinical phenotype including atrioventricular II block, Brugada-like ECG, septal fibrosis, right ventricular dilatation and decreased left ventricular contractility. Residue A1294 is located in the IIIS3-S4 extracellular loop, in proximity to several residues whose mutations are associated with sodium channelopathies. The wild-type channel Nav1.5 and mutant Nav1.5-A1294G were expressed in the CHO-K1 and HEK293T cells and whole-cell sodium currents were recorded using the patch-clamp method. The A1294G channels demonstrated a negative shift of steady-state inactivation, accelerated fast and slow inactivation and decelerated recovery from intermediate inactivation. Our study reveals biophysical mechanism of the Nav1.5-A1294G dysfunction, which may underlie the combined phenotypic manifestation observed in the patient.


Asunto(s)
Bloqueo Atrioventricular/genética , Síndrome de Brugada/genética , Predisposición Genética a la Enfermedad/genética , Canal de Sodio Activado por Voltaje NAV1.5/genética , Mutación Puntual , Adulto , Animales , Bloqueo Atrioventricular/fisiopatología , Síndrome de Brugada/fisiopatología , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Masculino , Canal de Sodio Activado por Voltaje NAV1.5/fisiología , Técnicas de Placa-Clamp , Fenotipo
12.
Int J Mol Sci ; 20(21)2019 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-31694310

RESUMEN

Abstract: Heart failure (HF) is associated with skeletal muscle wasting and exercise intolerance. This study aimed to evaluate the exercise-induced clinical response and histological alterations. One hundred and forty-four HF patients were enrolled. The individual training program was determined as a workload at or close to the lactate threshold (LT1); clinical data were collected before and after 12 weeks/6 months of training. The muscle biopsies from eight patients were taken before and after 12 weeks of training: histology analysis was used to evaluate muscle morphology. Most of the patients demonstrated a positive response after 12 weeks of the physical rehabilitation program in one or several parameters tested, and 30% of those showed improvement in all four of the following parameters: oxygen uptake (VO2) peak, left ventricular ejection fraction (LVEF), exercise tolerance (ET), and quality of life (QOL); the walking speed at LT1 after six months of training showed a significant rise. Along with clinical response, the histological analysis detected a small but significant decrease in both fiber and endomysium thickness after the exercise training course indicating the stabilization of muscle mechanotransduction system. Together, our data show that the beneficial effect of personalized exercise therapy in HF patients depends, at least in part, on the improvement in skeletal muscle physiological and biochemical performance.


Asunto(s)
Terapia por Ejercicio , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/rehabilitación , Músculo Esquelético/patología , Femenino , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/fisiopatología , Consumo de Oxígeno , Medicina de Precisión , Calidad de Vida , Volumen Sistólico
13.
Hum Mutat ; 39(9): 1161-1172, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29858533

RESUMEN

Mutations in FLNC for a long time are known in connection to neuromuscular disorders and only recently were described in association with various cardiomyopathies. Here, we report a new clinical phenotype of filaminopathy in four unrelated patients with early-onset restrictive cardiomyopathy (RCM) in combination with congenital myopathy due to FLNC mutations (NM_001458.4:c.3557C>T, p.A1186V, rs1114167361 in three probands and c.[3547G>C; 3548C>T], p.A1183L, rs1131692185 in one proband). In all cases, concurrent myopathy was confirmed by neurological examination, electromyography, and morphological studies. Three of the patients also presented with arthrogryposis. The pathogenicity of the described missense variants was verified by cellular and morphological studies and by in vivo modeling in zebrafish. Combination of in silico and experimental approaches revealed that FLNC missense variants localized in Ig-loop segments often lead to development of RCM. The described FLNC mutations associated with early-onset RCMP extend cardiac spectrum of filaminopathies and facilitate the differential diagnosis of restrictive cardiac phenotype associated with neuromuscular involvement in children.


Asunto(s)
Cardiomiopatía Restrictiva/genética , Anomalías Congénitas/genética , Filaminas/genética , Enfermedades Musculares/genética , Adolescente , Cardiomiopatía Restrictiva/fisiopatología , Preescolar , Anomalías Congénitas/fisiopatología , Análisis Mutacional de ADN , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Enfermedades Musculares/fisiopatología , Mutación , Linaje , Fenotipo
14.
Genet Res (Camb) ; 100: e3, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29502537

RESUMEN

Maternal gestational diabetes mellitus (GDM) is considered to be an important factor that epigenetically predisposes offspring to metabolic and cardiovascular diseases. However, the mechanisms of how intrauterine hyperglycaemia affects offspring have not been thoroughly studied. The mammalian tribbles homologue 1 (TRIB1) gene is associated with plasma lipid concentrations and coronary artery disease (CAD). Our aim was to study the effect of GDM and its treatment terms on the level of TRIB1 gene expression in human umbilical vein endothelial cells (HUVECs) of newborns from women with and without GDM. The study included 50 women with GDM and 25 women without GDM (control group). Women with GDM were divided into three groups according to their gestational age when the treatment of GDM started: 24-28 weeks (GDM1, N = 16), 29-32 weeks (GDM2, N = 25) and >34 weeks (GDM3, N = 9). The levels of TRIB1 gene expression in GDM3, GDM2, GDM1 and control groups were 2.8 ± 1.1, 4.2 ± 2.4, 6.0 ± 3.4 and 8.1 ± 6.1, respectively (p = 0.001). After comparison in pairs the difference was significant for the following pairs: GDM2-control (p = 0.004), GDM3-control (p = 0.002), GDM1-GDM3 (p = 0.012). Notably, if treatment had been started before the 28th week of gestation, the difference in TRIB1 gene expression in HUVECs was not significant (p = 0.320 for comparison between GDM1 and control groups). Our findings support the hypothesis that TRIB1 gene expression in HUVECs depends on the duration of intrauterine exposure to hyperglycaemia.


Asunto(s)
Diabetes Gestacional/genética , Estudios de Asociación Genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Adulto , Femenino , Expresión Génica , Edad Gestacional , Humanos , Hiperglucemia/genética , Recién Nacido , Embarazo , Proteínas Serina-Treonina Quinasas/genética , Factores de Tiempo
15.
Amino Acids ; 49(11): 1815-1829, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28831623

RESUMEN

Idiopathic restrictive cardiomyopathy (RCM, MIM# 115210) is the least common type of cardiomyopathies, often of genetic origin. Recently we described a spectrum of variants-classified as pathogenic, likely pathogenic and variants of unknown significance-in 24 patients suffering from idiopathic RCM. Pathogenic variants, detected in half of the RCM cases, were found in sarcomeric and cytoskeletal genes that have a predominant role in the development of RCM. Here we have analyzed the structural consequences of these missense variants and predicted their effect on the function of three large groups of domains: intrinsically disordered regions (IDRs), fibronectin-type III (FnIII) domains, and immunoglobulin-like (Ig) domains. Our findings indicate that pathogenic mutations are likely to disrupt interdomain interfaces, interfere with protein interactions, and affect protein stability, potentially destabilizing the multi-domain architecture of myofibrils and leading to myocardial stiffness in patients with idiopathic RCM.


Asunto(s)
Cardiomiopatía Restrictiva/genética , Predisposición Genética a la Enfermedad , Modelos Moleculares , Mutación , Proteínas/química , Proteínas/metabolismo , Secuencia de Aminoácidos/genética , Sustitución de Aminoácidos/genética , Bases de Datos de Proteínas , Fibronectinas/química , Fibronectinas/genética , Humanos , Conformación Proteica , Procesamiento Proteico-Postraduccional
16.
Mol Genet Metab ; 115(2-3): 118-27, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25982065

RESUMEN

Various mutations in LMNA gene, encoding for nuclear lamin A/C protein, lead to laminopathies and contribute to over ten human disorders, mostly affecting tissues of mesenchymal origin such as fat tissue, muscle tissue, and bones. Recently it was demonstrated that lamins not only play a structural role providing communication between extra-nuclear structures and components of cell nucleus but also control cell fate and differentiation. In our study we assessed the effect of various LMNA mutations on the expression profile of mesenchymal multipotent stem cells (MMSC) during adipogenic and osteogenic differentiation. We used lentiviral approach to modify human MMSC with LMNA-constructs bearing mutations associated with different laminopathies--G465D, R482L, G232E, R527C, and R471C. The impact of various mutations on MMSC differentiation properties and expression profile was assessed by colony-forming unit analysis, histological staining, expression of the key differentiation markers promoting adipogenesis and osteogenesis followed by the analysis of the whole set of genes involved in lineage-specific differentiation using PCR expression arrays. We demonstrate that various LMNA mutations influence the differentiation efficacy of MMSC in mutation-specific manner. Each LMNA mutation promotes a unique expression pattern of genes involved in a lineage-specific differentiation and this pattern is shared by the phenotype-specific mutations.


Asunto(s)
Laminas/genética , Células Madre Mesenquimatosas/metabolismo , Mutación , Transcriptoma , Adipogénesis , Diferenciación Celular , Células Cultivadas , Histona Desacetilasas/metabolismo , Humanos , Osteogénesis
18.
J Vasc Interv Radiol ; 25(5): 789-796.e7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24508346

RESUMEN

PURPOSE: Restenosis caused by intimal hyperplasia (IH) remains a significant drawback for vascular interventions. It is crucial to understand the molecular mechanisms that control activation of smooth muscle cells (SMCs) after the injury in order to develop strategies to prevent IH. The purpose of the present study was to investigate the early alterations in arterial-wall gene expression after balloon injury in the rat carotid artery with focus on the induction of an inflammatory response. MATERIALS AND METHODS: Twenty-four male Sprague-Dawley rats were subjected to injury of the left common carotid artery by using a 2-F Fogarty catheter. The arteries were harvested 5, 10, and 20 hours after injury. Uninjured arteries from an additional eight rats were used as controls. RNA was isolated and used for genome-wide microarray expression analysis, followed by validation of selected genes with quantitative real-time polymerase chain reaction (qRT-PCR). Immunohistochemistry was performed on the cross-sectioned vessels. RESULTS: Analysis of gene expression by microarrays showed that the most differentially expressed genes were primarily associated with inflammation, cell proliferation, migration, and adhesion. As confirmed by qRT-PCR, microarray data showed a significant (P < .005) upregulation of cytokines and chemokines (IL-6, CCL2, CXCL1, AIMP1, and CD44) just 5 hours after injury. Immunohistochemistry demonstrated that CCL2 and the adhesion receptor CD44 were expressed by SMCs in the early response to injury and in the absence of leukocyte infiltration. CONCLUSIONS: Arterial injury is followed by an early induction of inflammatory genes in the vessel wall that appears to be confined to SMCs.


Asunto(s)
Arterias Carótidas/inmunología , Traumatismos de las Arterias Carótidas/inmunología , Citocinas/inmunología , Regulación de la Expresión Génica/inmunología , Mediadores de Inflamación/inmunología , Músculo Liso Vascular/inmunología , Miocitos del Músculo Liso/inmunología , Animales , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/patología , Masculino , Músculo Liso Vascular/patología , Ratas , Ratas Sprague-Dawley
19.
Gynecol Endocrinol ; 30 Suppl 1: 43-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25200829

RESUMEN

OBJECTIVE: To review the literature and to present the latest advances in the autotransplantation of cryopreserved ovarian tissue. MATERIALS AND METHODS: A literature review was conducted for all relevant articles assessing the fertility preservation, ovarian tissue transplantation, standard freezing and vitrification of ovarian tissue. RESULTS: One of the promising and effective methods for fertility preservation may be the autotransplantation of cryopreserved ovarian tissue. At present, 30 babies have been born after orthotopic autotransplantation of frozen-thawed human ovarian tissue. Restoration of ovarian activity occurs between 3.5 months and 6.5 months. The longevity of autotransplanted ovarian tissue is about 5-7 years. The follicles are similarly preserved after all freezing methods; however, the ovarian stroma is significantly better preserved after vitrification than after slow freezing. An important topic for further research is preparation of the "vascular bed", optimization of vitrification technique and the development of alternative procedures to avoid the transmission of cancer cells via ovarian tissue autotransplantation - "artificial ovary". CONCLUSIONS: Cryopreservation of ovarian tissue has unique advantages over other strategies. This method: (1) does not delay cancer treatment; (2) is safer for hormone dependent malignancy; (3) can be done independent of menstrual cycles; (4) is the only option for prepubertal girls; (5) can restore not only fertility but endocrine function.


Asunto(s)
Criopreservación/métodos , Preservación de la Fertilidad/métodos , Neoplasias/terapia , Ovario/fisiología , Trasplante Autólogo/normas , Criopreservación/normas , Femenino , Preservación de la Fertilidad/normas , Humanos , Ovario/trasplante
20.
Noncoding RNA ; 10(1)2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38392970

RESUMEN

There are many articles on the quantitative analysis of miRNAs contained in a population of EVs of different sizes under various physiological and pathological conditions. For such analysis, it is important to correctly quantify the miRNA contents of EVs. It should be considered that quantification is skewed depending on the isolation protocol, and different miRNAs are degraded by nucleases with different efficiencies. In addition, it is important to consider the contribution of miRNAs coprecipitating with the EVs population, because the amount of miRNAs in the EVs population under study is skewed without appropriate enzymatic treatment. By studying a population of EVs from the blood plasma of healthy donors, we found that the absolute amount of miRNA inside the vesicles is commensurate with the amount of the same type of miRNA adhered to the outside of the EVs. The inside/outside ratio ranged from 1.02 to 2.64 for different investigated miRNAs. According to our results, we propose the hypothesis that high occupancy of miRNAs on the outer surface of EVs influence on the transporting RNA repertoire no less than the inner cargo received from the host cell.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA