Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Ther ; 31(3): 657-675, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36457248

ABSTRACT

Lysosomal storage diseases (LSDs) are multisystem inherited metabolic disorders caused by dysfunctional lysosomal activity, resulting in the accumulation of undegraded macromolecules in a variety of organs/tissues, including the central nervous system (CNS). Treatments include enzyme replacement therapy, stem/progenitor cell transplantation, and in vivo gene therapy. However, these treatments are not fully effective in treating the CNS as neither enzymes, stem cells, nor viral vectors efficiently cross the blood-brain barrier. Here, we review the latest advancements in improving delivery of different therapeutic agents to the CNS and comment upon outstanding questions in the field of neurological LSDs.


Subject(s)
Blood-Brain Barrier , Lysosomal Storage Diseases , Humans , Blood-Brain Barrier/metabolism , Lysosomal Storage Diseases/therapy , Lysosomal Storage Diseases/drug therapy , Central Nervous System/metabolism , Enzyme Replacement Therapy , Genetic Therapy/methods
2.
Int J Mol Sci ; 24(11)2023 May 31.
Article in English | MEDLINE | ID: mdl-37298497

ABSTRACT

Brugada syndrome (BrS) is an inherited autosomal dominant cardiac channelopathy. Pathogenic rare mutations in the SCN5A gene, encoding the alpha-subunit of the voltage-dependent cardiac Na+ channel protein (Nav1.5), are identified in 20% of BrS patients, affecting the correct function of the channel. To date, even though hundreds of SCN5A variants have been associated with BrS, the underlying pathogenic mechanisms are still unclear in most cases. Therefore, the functional characterization of the SCN5A BrS rare variants still represents a major hurdle and is fundamental to confirming their pathogenic effect. Human cardiomyocytes (CMs) differentiated from pluripotent stem cells (PSCs) have been extensively demonstrated to be reliable platforms for investigating cardiac diseases, being able to recapitulate specific traits of disease, including arrhythmic events and conduction abnormalities. Based on this, in this study, we performed a functional analysis of the BrS familial rare variant NM_198056.2:c.3673G>A (NP_932173.1:p.Glu1225Lys), which has been never functionally characterized before in a cardiac-relevant context, as the human cardiomyocyte. Using a specific lentiviral vector encoding a GFP-tagged SCN5A gene carrying the specific c.3673G>A variant and CMs differentiated from control PSCs (PSC-CMs), we demonstrated an impairment of the mutated Nav1.5, thus suggesting the pathogenicity of the rare BrS detected variant. More broadly, our work supports the application of PSC-CMs for the assessment of the pathogenicity of gene variants, the identification of which is increasing exponentially due to the advances in next-generation sequencing methods and their massive use in genetic testing.


Subject(s)
Brugada Syndrome , Pluripotent Stem Cells , Humans , Brugada Syndrome/metabolism , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Mutation , Pluripotent Stem Cells/metabolism
3.
Hum Mol Genet ; 29(2): 177-188, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31868880

ABSTRACT

Mitochondria undergo continuous cycles of fusion and fission in response to physiopathological stimuli. The key player in mitochondrial fission is dynamin-related protein 1 (DRP1), a cytosolic protein encoded by dynamin 1-like (DNM1L) gene, which relocalizes to the outer mitochondrial membrane, where it assembles, oligomerizes and drives mitochondrial division upon guanosine-5'-triphosphate (GTP) hydrolysis. Few DRP1 mutations have been described so far, with patients showing complex and variable phenotype ranging from early death to encephalopathy and/or optic atrophy. The disease is the consequence of defective mitochondrial fission due to faulty DRP1 function. However, the underlying molecular mechanisms and the functional consequences at mitochondrial and cellular level remain elusive. Here we report on a 5-year-old girl presenting psychomotor developmental delay, global hypotonia and severe ataxia due to axonal sensory neuropathy harboring a novel de novo heterozygous missense mutation in the GTPase domain of DRP1 (NM_012062.3:c.436G>A, NP_036192.2: p.D146N variant in DNM1L). Patient's fibroblasts show hyperfused/balloon-like giant mitochondria, highlighting the importance of D146 residue for DRP1 function. This dramatic mitochondrial rearrangement phenocopies what observed overexpressing DRP1-K38A, a well-known experimental dominant negative version of DRP1. In addition, we demonstrated that p.D146N mutation has great impact on peroxisomal shape and function. The p.D146N mutation compromises the GTPase activity without perturbing DRP1 recruitment or assembly, causing decreased mitochondrial and peroxisomal turnover. In conclusion, our findings highlight the importance of sensory neuropathy in the clinical spectrum of DRP1 variants and, for the first time, the impact of DRP1 mutations on mitochondrial turnover and peroxisomal functionality.


Subject(s)
Dynamins/genetics , Fibroblasts/ultrastructure , Mitochondria/genetics , Mitochondria/ultrastructure , Mitochondrial Dynamics/genetics , Peripheral Nervous System Diseases/genetics , Autophagy/genetics , Child, Preschool , Dynamins/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Heterozygote , Humans , Mitochondria/metabolism , Mitochondria/pathology , Mutation , Pedigree , Peripheral Nervous System Diseases/enzymology , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Peroxisomes/metabolism , Reactive Oxygen Species/metabolism , Exome Sequencing
4.
Eur Heart J ; 42(11): 1082-1090, 2021 03 14.
Article in English | MEDLINE | ID: mdl-33221895

ABSTRACT

AIMS: Brugada syndrome (BrS) is associated with an increased risk of sudden cardiac death due to ventricular tachycardia/fibrillation (VT/VF) in young, otherwise healthy individuals. Despite SCN5A being the most commonly known mutated gene to date, the genotype-phenotype relationship is poorly understood and remains uncertain. This study aimed to elucidate the genotype-phenotype correlation in BrS. METHODS AND RESULTS: Brugada syndrome probands deemed at high risk of future arrhythmic events underwent genetic testing and phenotype characterization by the means of epicardial arrhythmogenic substrate (AS) mapping, and were divided into two groups according to the presence or absence of SCN5A mutation. Two-hundred probands (160 males, 80%; mean age 42.6 ± 12.2 years) were included in this study. Patients harbouring SCN5A mutations exhibited a spontaneous type 1 pattern and experienced aborted cardiac arrest or spontaneous VT/VF more frequently than the other subjects. SCN5A-positive patients exhibited a larger epicardial AS area, more prolonged electrograms and more frequently observed non-invasive late potentials. The presence of an SCN5A mutation explained >26% of the variation in the epicardial AS area and was the strongest predictor of a large epicardial area. CONCLUSION: In BrS, the genetic background is the main determinant for the extent of the electrophysiological abnormalities. SCN5A mutation carriers exhibit more pronounced epicardial electrical abnormalities and a more aggressive clinical presentation. These results contribute to the understanding of the genetic determinants of the BrS phenotypic expression and provide possible explanations for the varying degrees of disease expression.


Subject(s)
Brugada Syndrome , Tachycardia, Ventricular , Adult , Brugada Syndrome/genetics , Electrocardiography , Epicardial Mapping , Humans , Male , Middle Aged , NAV1.5 Voltage-Gated Sodium Channel/genetics , Phenotype , Tachycardia, Ventricular/genetics , Ventricular Fibrillation
5.
Int J Mol Sci ; 23(21)2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36362347

ABSTRACT

GLUT1 deficiency syndrome (GLUT1DS1; OMIM #606777) is a rare genetic metabolic disease, characterized by infantile-onset epileptic encephalopathy, global developmental delay, progressive microcephaly, and movement disorders (e.g., spasticity and dystonia). It is caused by heterozygous mutations in the SLC2A1 gene, which encodes the GLUT1 protein, a glucose transporter across the blood-brain barrier (BBB). Most commonly, these variants arise de novo resulting in sporadic cases, although several familial cases with AD inheritance pattern have been described. Twenty-seven Italian pediatric patients, clinically suspect of GLUT1DS from both sporadic and familial cases, have been enrolled. We detected by trios sequencing analysis 25 different variants causing GLUT1DS. Of these, 40% of the identified variants (10 out of 25) had never been reported before, including missense, frameshift, and splice site variants. Their structural mapping on the X-ray structure of GLUT1 strongly suggested the potential pathogenic effects of these novel disease-related mutations, broadening the genotypic spectrum heterogeneity found in the SLC2A1 gene. Moreover, 24% is located in a vulnerable region of the GLUT1 protein that involves transmembrane 4 and 5 helices encoded by exon 4, confirming a mutational hotspot in the SLC2A1 gene. Lastly, we investigated possible correlations between mutation type and clinical and biochemical data observed in our GLUT1DS cohort, revealing that splice site and frameshift variants are related to a more severe phenotype and low CSF parameters.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Humans , Glucose Transporter Type 1/genetics , Carbohydrate Metabolism, Inborn Errors/genetics , Monosaccharide Transport Proteins/genetics , Mutation , Molecular Biology
6.
Pacing Clin Electrophysiol ; 44(3): 552-556, 2021 03.
Article in English | MEDLINE | ID: mdl-33372694

ABSTRACT

We present, to our knowledge, the first case of immunosuppressive therapy (IST) application in a 12-year-old child with arrhythmogenic inflammatory cardiomyopathy resulting from the overlap between autoimmune myocarditis and primary arrhythmogenic cardiomyopathy. Indication to off-lable IST was compelling, because of recurrent drug-refractory ventricular arrhythmias (VAs). We show that IST was feasible, safe, and effective on multiple clinical endpoints, including symptoms, VA recurrences, and T-troponin release. Remarkably, all diagnostic and therapeutic strategies were worked out by a dedicated multidisciplinary team, including specialized pediatric immunologists.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/drug therapy , Arrhythmogenic Right Ventricular Dysplasia/immunology , Immunosuppression Therapy , Azathioprine/therapeutic use , Biomarkers/blood , Child , Echocardiography , Electrocardiography , Humans , Magnetic Resonance Imaging , Male , Myocarditis/drug therapy , Myocarditis/immunology , Prednisone/therapeutic use , Recurrence , Risk Factors
7.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946750

ABSTRACT

Genetic testing in Brugada syndrome (BrS) is still not considered to be useful for clinical management of patients in the majority of cases, due to the current lack of understanding about the effect of specific variants. Additionally, family history of sudden death is generally not considered useful for arrhythmic risk stratification. We sought to demonstrate the usefulness of genetic testing and family history in diagnosis and risk stratification. The family history was collected for a proband who presented with a personal history of aborted cardiac arrest and in whom a novel variant in the SCN5A gene was found. Living family members underwent ajmaline testing, electrophysiological study, and genetic testing to determine genotype-phenotype segregation, if any. Patch-clamp experiments on transfected human embryonic kidney 293 cells enabled the functional characterization of the SCN5A novel variant in vitro. In this study, we provide crucial human data on the novel heterozygous variant NM_198056.2:c.5000T>A (p.Val1667Asp) in the SCN5A gene, and demonstrate its segregation with a severe form of BrS and multiple sudden deaths. Functional data revealed a loss of function of the protein affected by the variant. These results provide the first disease association with this variant and demonstrate the usefulness of genetic testing for diagnosis and risk stratification in certain patients. This study also demonstrates the usefulness of collecting the family history, which can assist in understanding the severity of the disease in certain situations and confirm the importance of the functional studies to distinguish between pathogenic mutations and harmless genetic variants.


Subject(s)
Brugada Syndrome/genetics , Mutation, Missense , NAV1.5 Voltage-Gated Sodium Channel/genetics , Adolescent , Adult , Aged , Ajmaline/pharmacology , Amino Acid Substitution , Brugada Syndrome/complications , Brugada Syndrome/metabolism , Death, Sudden, Cardiac/etiology , Electrocardiography , Female , Genetic Testing , HEK293 Cells , Heterozygote , Humans , Loss of Function Mutation , Male , Middle Aged , Mutant Proteins/genetics , Mutant Proteins/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Patch-Clamp Techniques , Pedigree , Polymorphism, Single Nucleotide , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
8.
Europace ; 22(12): 1864-1872, 2020 12 23.
Article in English | MEDLINE | ID: mdl-32995851

ABSTRACT

AIMS: We aimed at addressing the role of late gadolinium enhancement (LGE) in arrhythmic risk stratification of LMNA-associated cardiomyopathy (CMP). METHODS AND RESULTS: We present data from a multicentre national cohort of patients with LMNA mutations. Of 164 screened cases, we finally enrolled patients with baseline cardiac magnetic resonance (CMR) including LGE sequences [n = 41, age 35 ± 17 years, 51% males, mean left ventricular ejection fraction (LVEF) by echocardiogram 56%]. The primary endpoint of the study was follow-up (FU) occurrence of malignant ventricular arrhythmias [MVA, including sustained ventricular tachycardia (VT), ventricular fibrillation, and appropriate implantable cardioverter-defibrillator (ICD) therapy]. At baseline CMR, 25 subjects (61%) had LGE, with non-ischaemic pattern in all of the cases. Overall, 23 patients (56%) underwent ICD implant. By 10 ± 3 years FU, eight patients (20%) experienced MVA, consisting of appropriate ICD shocks in all of the cases. In particular, the occurrence of MVA in LGE+ vs. LGE- groups was 8/25 vs. 0/16 (P = 0.014). Of note, no significant differences between LGE+ and LGE- patients were found in currently recognized risk factors for sudden cardiac death (male gender, non-missense mutations, baseline LVEF <45% and non-sustained VT), all P-value >0.05. CONCLUSIONS: In LMNA-CMP patients, LGE at baseline CMR is significantly associated with MVA. In particular, as suggested by this preliminary experience, the absence of LGE allowed to rule-out MVA at 10 years mean FU.


Subject(s)
Cardiomyopathies , Defibrillators, Implantable , Adolescent , Adult , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/genetics , Contrast Media , Female , Follow-Up Studies , Gadolinium , Humans , Lamin Type A/genetics , Male , Middle Aged , Predictive Value of Tests , Risk Assessment , Risk Factors , Stroke Volume , Ventricular Function, Left , Young Adult
9.
Ann Intern Med ; 171(7): 458-463, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31476771

ABSTRACT

Background: Mutations in the LMNA (lamin A/C) gene have been associated with neuromuscular and cardiac manifestations, but the clinical implications of these signs are not well understood. Objective: To learn more about the natural history of LMNA-related disease. Design: Observational study. Setting: 13 clinical centers in Italy from 2000 through 2018. Patients: 164 carriers of an LMNA mutation. Measurements: Detailed cardiologic and neurologic evaluation at study enrollment and for a median of 10 years of follow-up. Results: The median age at enrollment was 38 years, and 51% of participants were female. Neuromuscular manifestations preceded cardiac signs by a median of 11 years, but by the end of follow-up, 90% of the patients had electrical heart disease followed by structural heart disease. Overall, 10 patients (6%) died, 14 (9%) received a heart transplant, and 32 (20%) had malignant ventricular arrhythmias. Fifteen patients had gait loss, and 6 had respiratory failure. Atrial fibrillation and second- and third-degree atrioventricular block were observed, respectively, in 56% and 51% of patients with combined cardiac and neuromuscular manifestations and 37% and 33% of those with heart disease only. Limitations: Some of the data were collected retrospectively. Neuromuscular manifestations were more frequent in this analysis than in previous studies. Conclusion: Many patients with an LMNA mutation have neurologic symptoms by their 30s and develop progressive cardiac manifestations during the next decade. A substantial proportion of these patients will have life-threatening neurologic or cardiologic conditions. Primary Funding Source: None.


Subject(s)
Cardiomyopathies/epidemiology , Cardiomyopathies/genetics , Lamin Type A/genetics , Muscular Dystrophies/epidemiology , Mutation , Adult , Arrhythmias, Cardiac/epidemiology , Arrhythmias, Cardiac/genetics , Atrial Fibrillation/epidemiology , Atrial Fibrillation/genetics , Atrioventricular Block/epidemiology , Atrioventricular Block/genetics , Disease Progression , Female , Follow-Up Studies , Gait Disorders, Neurologic/epidemiology , Gait Disorders, Neurologic/genetics , Heart Failure/genetics , Heart Failure/mortality , Heart Transplantation/statistics & numerical data , Humans , Italy/epidemiology , Male , Middle Aged , Muscular Dystrophies/genetics , Prospective Studies , Respiratory Insufficiency/epidemiology , Respiratory Insufficiency/genetics
10.
Int J Mol Sci ; 21(16)2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32824506

ABSTRACT

Brugada syndrome (BrS) is diagnosed by the presence of an elevated ST-segment and can result in sudden cardiac death. The most commonly found mutated gene is SCN5A, which some argue is the only gene that has been definitively confirmed to cause BrS, while the potential causative effect of other genes is still under debate. While the issue of BrS genetics is currently a hot topic, current knowledge is not able to result in molecular confirmation of over half of BrS cases. Therefore, it is difficult to develop research models with wide potential. Instead, the clinical genetics first need to be better understood. In this study, we provide crucial human data on the novel heterozygous variant NM_198056.2:c.4285G>A (p.Val1429Met) in the SCN5A gene, and demonstrate its segregation with BrS, suggesting a pathogenic effect. These results provide the first disease association with this variant and are crucial clinical data to communicate to basic scientists, who could perform functional studies to better understand the molecular effects of this clinically-relevant variant in BrS.


Subject(s)
Brugada Syndrome/genetics , Mutation , NAV1.5 Voltage-Gated Sodium Channel/genetics , Adult , Aged , Brugada Syndrome/diagnosis , Female , Heterozygote , Humans , Male , Middle Aged , Pedigree
11.
Europace ; 21(10): 1550-1558, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31292628

ABSTRACT

AIMS: The Brugada syndrome (BrS) is an inherited disease associated with an increased risk of sudden cardiac death. Often, the genetic cause remains undetected. Perhaps due at least in part because the NaV1.8 protein is expressed more in both the central and peripheral nervous systems than in the heart, the SCN10A gene is not included in diagnostic arrhythmia/sudden death panels in the vast majority of cardiogenetics centres. METHODS AND RESULTS: Clinical characteristics were assessed in patients harboring either SCN5A or novel SCN10A variants. Genetic testing was performed using Next Generation Sequencing on genomic DNA. Clinical characteristics, including the arrhythmogenic substrate, in BrS patients harboring novel SCN10A variants and SCN5A variants are comparable. Clinical characteristics, including gender, age, personal history of cardiac arrest/syncope, spontaneous BrS electrocardiogram pattern, family history of sudden death, and arrhythmic substrate are not significantly different between probands harboring SCN10A or SCN5A variants. CONCLUSION: Future studies are warranted to further characterize the role of these specific SCN10A variants.


Subject(s)
Brugada Syndrome/genetics , DNA/genetics , Genetic Predisposition to Disease , Mutation, Missense , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/genetics , Adolescent , Adult , Aged , Brugada Syndrome/diagnosis , Brugada Syndrome/metabolism , DNA Mutational Analysis , Electrocardiography , Female , Genetic Testing , Humans , Male , Middle Aged , NAV1.5 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Young Adult
12.
Adv Exp Med Biol ; 1147: 319-344, 2019.
Article in English | MEDLINE | ID: mdl-31147885

ABSTRACT

The muscular dystrophies are an heterogeneous group of inherited myopathies characterised by the progressive wasting of skeletal muscle tissue. Pericytes have been shown to make muscle in vitro and to contribute to skeletal muscle regeneration in several animal models, although recent data has shown this to be controversial. In fact, some pericyte subpopulations have been shown to contribute to fibrosis and adipose deposition in muscle. In this chapter, we explore the identity and the multifaceted role of pericytes in dystrophic muscle, potential therapeutic applications and the current need to overcome the hurdles of characterisation (both to identify pericyte subpopulations and track cell fate), to prevent deleterious differentiation towards myogenic-inhibiting subpopulations, and to improve cell proliferation and engraftment efficacy.


Subject(s)
Muscular Dystrophies , Pericytes , Animals , Cell Differentiation , Muscle Development , Muscle, Skeletal , Regeneration
13.
Int J Mol Sci ; 20(19)2019 10 04.
Article in English | MEDLINE | ID: mdl-31590245

ABSTRACT

Brugada syndrome (BrS) is marked by an elevated ST-segment elevation and increased risk of sudden cardiac death. Variants in the SCN5A gene are considered to be molecular confirmation of the syndrome in about one third of cases, while the genetics remain a mystery in about half of the cases, with the remaining cases being attributed to variants in any of a number of genes. Before research models can be developed, it is imperative to understand the genetics in patients. Even data from humans is complicated, since variants in the most common gene in BrS, SCN5A, are associated with a number of pathologies, or could even be considered benign, depending on the variant. Here, we provide crucial human data on a novel NM_198056.2:c.2091G>A (p.Trp697X) point-nonsense heterozygous variant in the SCN5A gene, as well as its segregation with BrS. The results herein suggest a pathogenic effect of this variant. These results could be used as a stepping stone for functional studies to better understand the molecular effects of this variant in BrS.


Subject(s)
Brugada Syndrome/genetics , Codon, Nonsense , NAV1.5 Voltage-Gated Sodium Channel/genetics , Adult , Brugada Syndrome/pathology , Female , Humans , Male , Middle Aged , Pedigree
14.
Prenat Diagn ; 2018 Apr 12.
Article in English | MEDLINE | ID: mdl-29644715

ABSTRACT

OBJECTIVES: To compare the prognostic value of fetal serum biochemistry and fetal urine biochemistry in predicting renal outcome in lower urinary tract obstruction (LUTO). METHODS: We retrospectively studied renal outcome following a prenatal diagnosis of LUTO in cases for which both fetal blood and fetal urine were sampled. We classified the renal outcome as either "favorable," when postnatal renal function was normal, or "adverse," in the case of postnatal chronic renal failure or when renal histological lesions were present at autopsy in the case of termination of pregnancy. A prognostic model was constructed for urine and serum separately. ß2-Microglobulin was the only remaining independent predictor in fetal urine. ß2-Microglobulin in serum and urine were compared by using receiver operating characteristic curves. RESULTS: In the 50 cases included, the rate of adverse outcome was 34 of 50(68%): autopsy confirmed severity of renal disease in all 27 cases who underwent termination of pregnancy, and among the 23 live born children, 7 developed renal failure. Fetal serum and urine markers were all significantly associated with renal outcome (P < .01). The receiver operating characteristic curves for fetal serum and fetal urinary ß2-microglobulin were similar (area under the curve = 0.908 versus 0.909, P = .96). CONCLUSION: Fetal serum biochemistry and fetal urine biochemistry are of similar prognostic value in predicting postnatal renal outcome in fetuses with LUTO.

15.
Hum Mol Genet ; 24(20): 5828-35, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26220970

ABSTRACT

Brugada syndrome (BrS) is an inherited cardiac arrhythmic disorder that can lead to sudden death, with a prevalence of 1:5000 in Caucasian population and affecting mainly male patients in their third to fourth decade of life. BrS is inherited as an autosomal dominant trait; however, to date genetic bases have been only partially understood. Indeed most mutations are located in the SCN5A gene, encoding the alpha-subunit of the Na(+) cardiac channel, but >70% BrS patients still remain genetically undiagnosed. Although 21 other genes have been associated with BrS susceptibility, their pathogenic role is still unclear. A recent next-generation sequencing study investigated the contribution of 45 arrhythmia susceptibility genes in BrS pathogenesis, observing a significant enrichment only for SCN5A. In our study, we evaluated the distribution of putative functional variants in a wider panel of 158 genes previously associated with arrhythmic and cardiac defects in a cohort of 91 SCN5A-negative BrS patients. In addition, to identify genes significantly enriched in BrS, we performed a mutation burden test by using as control dataset European individuals selected from the 1000Genomes project. We confirmed BrS genetic heterogeneity and identified new potential BrS candidates such as DSG2 and MYH7, suggesting a possible genetic overlap between different cardiac disorders.


Subject(s)
Brugada Syndrome/genetics , Genetic Predisposition to Disease , Adult , Aged , Brugada Syndrome/metabolism , Cardiac Myosins/genetics , DNA Mutational Analysis , Desmoglein 2/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Mutation , Myosin Heavy Chains/genetics , White People/genetics
16.
Radiology ; 276(1): 207-18, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25611737

ABSTRACT

PURPOSE: To investigate whether specific patterns of brain gray matter (GM) regional volumes and white matter (WM) microstructural abnormalities and spinal cord atrophy occur in patients with pure and complicated hereditary spastic paraplegias (HSPs). Relationships between clinical and cognitive features of patients with HSP who had brain and cervical cord damage were also investigated. MATERIALS AND METHODS: This study was approved by the local ethical committees on human studies, and written informed consent from all subjects was obtained prior to enrollment. Forty-four patients with HSP (20 genetically defined cases and 24 without genetic diagnosis) and 19 healthy control subjects underwent clinical, neuropsychological, and advanced magnetic resonance (MR) imaging evaluations. Patterns of GM atrophy and WM microstructural damage obtained by using structural and diffusion-tensor MR imaging were compared between groups. Cervical cord atrophy was also assessed by using an active surface method. Correlations between clinical, cognitive, and diffusion-tensor MR imaging measures were evaluated. RESULTS: Clinical data showed that spastic paraplegia is accompanied by a number of other features, including sensory disturbances, and verbal and spatial memory deficits, not only in complicated HSP but also in pure HSP. MR imaging demonstrated a similar involvement of motor, association, and cerebellar WM pathways (P < .05, family-wise error corrected for multiple comparisons) and cervical cord (P < .001) in patients with HSP relative to healthy control subjects, regardless of their clinical picture. The severity of WM damage correlated with the degree of spasticity (P < .05, family-wise error corrected) and cognitive impairment (r values, -0.39 to 0.51; P values, .001-.05) in both pure and complicated HSP. CONCLUSION: The detection of a distributed pattern of central nervous system damage in patients with pure and complicated HSP suggests that the "primary" corticospinal tract involvement known to occur in these patients may be associated with a neurodegenerative process, which spreads out to extramotor regions, likely via anatomic connections. This observation is in line with emerging pieces of evidence that, independent of the clinical phenotype, there is a common neurodegenerative cascade shared by different neurologic disorders.


Subject(s)
Central Nervous System/pathology , Magnetic Resonance Imaging , Spastic Paraplegia, Hereditary/pathology , Adult , Aged , Cognition , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Phenotype , Spastic Paraplegia, Hereditary/diagnosis , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/psychology
17.
Clin Sci (Lond) ; 129(12): 1225-36, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26561594

ABSTRACT

Kidney glomeruli ultrafilter blood to generate urine and they are dysfunctional in a variety of kidney diseases. There are two key vascular growth factor families implicated in glomerular biology and function, namely the vascular endothelial growth factors (VEGFs) and the angiopoietins (Angpt). We present examples showing not only how these molecules help generate and maintain healthy glomeruli but also how they drive disease when their expression is dysregulated. Finally, we review how manipulating VEGF and Angpt signalling may be used to treat glomerular disease.


Subject(s)
Angiopoietins/metabolism , Glomerular Filtration Rate , Kidney Diseases/metabolism , Kidney Glomerulus/blood supply , Kidney Glomerulus/metabolism , Signal Transduction , Vascular Endothelial Growth Factors/metabolism , Angiopoietins/genetics , Animals , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/physiopathology , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Humans , Kidney Diseases/genetics , Kidney Diseases/physiopathology , Kidney Diseases/therapy , Kidney Glomerulus/physiopathology , Vascular Endothelial Growth Factors/genetics
18.
Circ Res ; 113(4): 389-98, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23753573

ABSTRACT

RATIONALE: A cell-based biological pacemaker is based on the differentiation of stem cells and the selection of a population displaying the molecular and functional properties of native sinoatrial node (SAN) cardiomyocytes. So far, such selection has been hampered by the lack of proper markers. CD166 is specifically but transiently expressed in the mouse heart tube and sinus venosus, the prospective SAN. OBJECTIVE: We have explored the possibility of using CD166 expression for isolating SAN progenitors from differentiating embryonic stem cells. METHODS AND RESULTS: We found that in embryonic day 10.5 mouse hearts, CD166 and HCN4, markers of the pacemaker tissue, are coexpressed. Sorting embryonic stem cells for CD166 expression at differentiation day 8 selects a population of pacemaker precursors. CD166+ cells express high levels of genes involved in SAN development (Tbx18, Tbx3, Isl-1, Shox2) and function (Cx30.2, HCN4, HCN1, CaV1.3) and low levels of ventricular genes (Cx43, Kv4.2, HCN2, Nkx2.5). In culture, CD166+ cells form an autorhythmic syncytium composed of cells morphologically similar to and with the electrophysiological properties of murine SAN myocytes. Isoproterenol increases (+57%) and acetylcholine decreases (-23%) the beating rate of CD166-selected cells, which express the ß-adrenergic and muscarinic receptors. In cocultures, CD166-selected cells are able to pace neonatal ventricular myocytes at a rate faster than their own. Furthermore, CD166+ cells have lost pluripotency genes and do not form teratomas in vivo. CONCLUSIONS: We demonstrated for the first time the isolation of a nonteratogenic population of cardiac precursors able to mature and form a fully functional SAN-like tissue.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , Sinoatrial Node/cytology , Stem Cells/cytology , Acetylcholine/pharmacology , Animals , Biomarkers/metabolism , Cardiotonic Agents/pharmacology , Cell Differentiation/physiology , Cell Line , Cell Proliferation , Coculture Techniques , Embryonic Stem Cells/drug effects , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Isoproterenol/pharmacology , Mice , Models, Animal , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Sinoatrial Node/drug effects , Sinoatrial Node/metabolism , Stem Cells/drug effects , Stem Cells/metabolism
20.
Clin Chim Acta ; 552: 117684, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38016628

ABSTRACT

Atherosclerotic lesions are present even in very young individuals and therefore, risk stratification for cardiovascular (CV) disease should be implemented in childhood to promote early prevention strategies. In this review we critically appraise clinical, biochemical and genetic biomarkers available for pediatric clinical practice, which might be integrated to build effective algorithms to identify children at risk of future CV events. The first critical issue is to characterize in children aged 2-5 years, those CV risk factors/clinical conditions associated with dramatically accelerated atherosclerosis. Presence of clinical conditions such as obesity, diabetes mellitus, Kawasaki disease, etc., or positive family history for premature CV disease should be evaluated. Subsequently, a complete lipid profile and Lipoprotein(a) determination are recommended for children with increased baseline CV risk. Individuals with altered lipid profile could then undergo genetic testing for monogenic dyslipidemias to identify children with high CV genetic risk, who will be directed to appropriate therapeutic options. In perspective, calculation of a polygenic risk score, based on the analysis of several common single-nucleotide polymorphisms, could be integrated for better risk assessment. We here emphasize the importance of a "holistic" strategy integrating biochemical, anamnestic and genetic data to stratify CV risk in early childhood.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Humans , Child, Preschool , Child , Risk Factors , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/genetics , Cardiovascular Diseases/prevention & control , Atherosclerosis/diagnosis , Atherosclerosis/genetics , Risk Assessment , Heart Disease Risk Factors , Lipids
SELECTION OF CITATIONS
SEARCH DETAIL