Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 381
Filtrar
1.
J Alzheimers Dis ; 100(s1): S179-S185, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39093076

RESUMEN

Fortea et al.'s. (2024) recent data analysis elegantly calls attention to familial late-onset Alzheimer's disease (AD) with APOE4 homozygosity. The article by Grant (2024) reviews the factors associated with AD, particularly the APOE genotype and lifestyle, and the broad implications for prevention, both for individuals with the lifestyles associated with living in resource-rich countries and for those enduring environmental adversity in poverty settings, including high exposure to enteric pathogens and precarious access to healthcare. Grant discusses the issue of APOE genotype and its implications for the benefits of lifestyle modifications. This review highlights that bearing APOE4 could constitute an evolutionary benefit in coping with heavy enteric infections and malnutrition early in life in the critical formative first two years of brain development. However, the critical issue may be that this genotype could be a health concern under shifts in lifestyle and unhealthy diets during aging, leading to severe cognitive impairments and increased risk of AD. This commentary supports the discussions of Grant and the benefits of improving lifestyle for decreasing the risks for AD while providing further understanding and modelling of the early life benefits of APOE4 amidst adversity. This attention to the pathophysiology of AD should help further elucidate these critical, newly appreciated pathogenic pathways for developing approaches to the prevention and management in the context of the APOE genetic variations associated with AD.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E4 , Desnutrición , Plasticidad Neuronal , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/prevención & control , Apolipoproteína E4/genética , Plasticidad Neuronal/genética , Desnutrición/genética , Desnutrición/complicaciones , Homocigoto , Estilo de Vida
2.
Clin Chim Acta ; 563: 119898, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39094808

RESUMEN

BACKGROUND: The T-cell receptor (TCR)/CD3 complex plays a crucial role in T-cell development and immune regulation. CD3G gene encodes one of the CD3 subunits named CD3γ, and its deficiency can cause autoimmune disorders, immunodeficiency and recurrent infections. To date, only 13 patients with CD3G variants have been reported. CASE REPORT: We report a 10-year-old Chinese boy presented with lupus-like disease in addition to autoimmune thyroiditis, asthma, immunodeficiency and recurrent infection. Flow cytometric analysis revealed apparently decreased levels of CD3+ and CD8+ T cells but mildly decreased CD4+ T cells. However, the activation of T cells and B cells increased. RESULTS: Trio-based whole-exome sequencing revealed a homozygous pathogenic variant (c.213delA, p.Lys71fs) of CD3G gene in the proband. His parents were both heterozygous carriers of this variant. CONCLUSION: This is the first patient who met the diagnostic criteria for systemic lupus erythematosus by the Systemic Lupus International Collaborating Clinics (SLICC) group. In addition to low T cells and low Treg cells, our study further revealed T cells and B cells activation enhanced in CD3γ deficiency patient, which may play an important role in autoimmunity. We believe that our study makes a significant contribution to the literature and will provide further insight into CD3γ deficiency and monogenic lupus.


Asunto(s)
Complejo CD3 , Lupus Eritematoso Sistémico , Tiroiditis Autoinmune , Humanos , Masculino , Niño , Complejo CD3/genética , Lupus Eritematoso Sistémico/genética , Tiroiditis Autoinmune/genética , Homocigoto , Pueblo Asiatico/genética , China , Pueblos del Este de Asia
3.
Eur J Endocrinol ; 191(2): 144-155, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39097530

RESUMEN

OBJECTIVE: Adrenal cortisol production occurs through a biosynthetic pathway which depend on NADH and NADPH for energy supply. The mitochondrial respiratory chain and the reactive oxygen species (ROS) detoxification system are therefore important for steroidogenesis. Mitochondrial dysfunction leading to oxidative stress has been implicated in the pathogenesis of several adrenal conditions. Nonetheless, only very few patients with variants in one gene of the ROS detoxification system, Thioredoxin Reductase 2 (TXNRD2), have been described with variable phenotypes. DESIGN: Clinical, genetic, structural, and functional characterization of a novel, biallelic TXNRD2 splice variant. METHODS: On human biomaterial, we performed whole exome sequencing to identify and RNA analysis to characterize the specific TXNRD2 splice variant. Amino acid conservation analysis and protein structure modeling were performed in silico. Using patient's fibroblast-derived human induced pluripotent stem cells, we generated adrenal-like cells (iALC) to study the impact of wild-type (WT) and mutant TXNRD2 on adrenal steroidogenesis and ROS production. RESULTS: The patient had a complex phenotype of primary adrenal insufficiency (PAI), combined with genital, ophthalmological, and neurological features. He carried a homozygous splice variant c.1348-1G > T in TXNRD2 which leads to a shorter protein lacking the C-terminus and thereby affecting homodimerization and flavin adenine dinucleotide binding. Patient-derived iALC showed a loss of cortisol production with overall diminished adrenal steroidogenesis, while ROS production was significantly increased. CONCLUSION: Lack of TXNRD2 activity for mitochondrial ROS detoxification affects adrenal steroidogenesis and predominantly cortisol production.


Asunto(s)
Tiorredoxina Reductasa 2 , Humanos , Masculino , Tiorredoxina Reductasa 2/genética , Tiorredoxina Reductasa 2/metabolismo , Homocigoto , Especies Reactivas de Oxígeno/metabolismo , Hidrocortisona/metabolismo , Hidrocortisona/biosíntesis , Células Madre Pluripotentes Inducidas/metabolismo , Secuenciación del Exoma
4.
Transl Vis Sci Technol ; 13(8): 2, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39087930

RESUMEN

Purpose: Homozygous hypomorphic variants of the RP1 gene, including c.5797C>T, p.Arg1933Ter, have traditionally been considered non-pathogenic. This study aimed to elucidate the clinical manifestations of late-onset, slowly progressive cone/macular dystrophy in patients homozygous for p.Arg1933Ter in the RP1 gene. Methods: Five patients with biallelic p.Arg1933Ter in RP1 were retrospectively recruited, and their clinical profiles were analyzed. Copy number variation analysis and Alu insertion assessment of genes associated with inherited retinal diseases were conducted. The results of comprehensive ophthalmological examinations, multimodal imaging, and full-field electroretinogram tests were analyzed. Results: No specific sequencing errors or structural variations associated with the clinical phenotypes were identified. Alu element insertion in RP1 was not detected. The mean ± SD age at the first visit was 62.2 ± 9.8 years, with symptoms typically starting between 45 and 50 years of age. Two patients exhibited a mild form of cone/macular dystrophy, characterized by a relatively preserved fundus appearance and blurring of the ellipsoid zone on optical coherence tomography. Three patients had late-onset cone/macular dystrophy with significant atrophy. Conclusions: To our knowledge, this study is the first to report that a homozygous hypomorphic variant of RP1, previously considered non-pathogenic, leads to cone/macular dystrophy. Translational Relevance: The study introduces novel possibilities suggesting that the homozygous hypomorphic variant of RP1 may be linked to variant pathogenicity.


Asunto(s)
Electrorretinografía , Proteínas del Ojo , Tomografía de Coherencia Óptica , Humanos , Masculino , Femenino , Persona de Mediana Edad , Estudios Retrospectivos , Anciano , Proteínas del Ojo/genética , Agudeza Visual , Variaciones en el Número de Copia de ADN/genética , Progresión de la Enfermedad , Distrofia del Cono/genética , Distrofia del Cono/diagnóstico por imagen , Degeneración Macular/genética , Degeneración Macular/patología , Degeneración Macular/diagnóstico por imagen , Degeneración Macular/congénito , Linaje , Homocigoto , Fenotipo , Mutación , Adulto , Edad de Inicio , Proteínas Asociadas a Microtúbulos
5.
Mol Genet Genomic Med ; 12(8): e2507, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39132856

RESUMEN

BACKGROUND: Mucopolysaccharidosis type I (MPS-I) is a rare autosomal recessive genetic lysosomal storage disorder that is caused by pathogenic variants of the α-L-iduronidase (IDUA) gene. This study aimed to identify the genetic causes of MPS-I in a Chinese patient and construct a minigene of IDUA to analyze its variants upon splicing. METHODS: Whole-exome sequencing (WES) and Sanger sequencing were used to confirm the potential causative variants. Single-nucleotide polymorphism (SNP) array was subsequently performed to confirm uniparental disomy (UPD). Minigene assay was performed to analyze the effect on splicing of mRNA. We meanwhile explored the conservative analysis and protein homology simulation. RESULTS: A novel homozygous splicing mutation of IDUA, c.159-9T>A, was identified in an individual presenting with overlapping features of MPS-I. Interestingly, only the father and sisters, but not the mother, carried the variant in a heterozygous state. WES and SNP array analyses validated paternal UPD on chromosome 4. Minigene splicing revealed two aberrant splicing events: exon 2 skipping and intron 1 retention. Moreover, the specific structure of the mutant protein obviously changed according to the results of the homologous model. CONCLUSIONS: This study describes a rare autosomal recessive disorder with paternal UPD of chromosome 4 leading to the homozygosity of the IDUA splicing variant in patients with MPS-I for the first time. This study expands the variant spectrum of IDUA and provides insights into the splicing system, facilitating its enhanced diagnosis and treatment.


Asunto(s)
Cromosomas Humanos Par 4 , Homocigoto , Iduronidasa , Mucopolisacaridosis I , Empalme del ARN , Disomía Uniparental , Humanos , Disomía Uniparental/genética , Disomía Uniparental/patología , Iduronidasa/genética , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/patología , Masculino , Cromosomas Humanos Par 4/genética , Femenino , Polimorfismo de Nucleótido Simple , Mutación , Pueblos del Este de Asia
6.
Sci Rep ; 14(1): 18876, 2024 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143102

RESUMEN

Progressive familial intrahepatic cholestasis (PFIC) is a rare childhood manifested disease associated with impaired bile secretion with severe pruritus yellow stool, and sometimes hepatosplenomegaly. PFIC is caused by mutations in ATP8B1, ABCB11, ABCB4, TJP2, NR1H4, SLC51A, USP53, KIF12, ZFYVE19, and MYO5B genes depending on its type. ABCB11 mutations lead to PFIC2 that encodes the bile salt export pump (BSEP). Different mutations of ABCB11 have been reported in different population groups but no data available in Pakistani population being a consanguineous one. We sequenced coding exons of the ABCB11 gene along with its flanking regions in 66 unrelated Pakistani children along with parents with PFIC2 phenotype. We identified 20 variations of ABCB11: 12 in homozygous form, one compound heterozygous, and seven heterozygous. These variants include 11 missenses, two frameshifts, two nonsense mutations, and five splicing variants. Seven variants are novel candidate variants and are not detected in any of the 120 chromosomes from normal ethnically matched individuals. Insilico analysis revealed that four homozygous missense variations have high pathogenic scores. Minigene analysis of splicing variants showed exon skipping and the addition of exon. This data is a useful addition to the disease variants genomic database and would be used in the future to build a diagnostic algorithm.


Asunto(s)
Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Colestasis Intrahepática , Humanos , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/genética , Colestasis Intrahepática/genética , Pakistán , Masculino , Femenino , Niño , Preescolar , Lactante , Mutación , Exones/genética , Estudios de Cohortes , Homocigoto
7.
Animal ; 18(8): 101236, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39096602

RESUMEN

Tunchang pigs, mainly distributed throughout Hainan Province of China, are well-known for their superior meat quality, crude feed tolerance, and adaptability to high temperatures and humidity. Runs of homozygosity (ROH) can provide valuable information about the inbreeding coefficient in individuals and selection signals that may reveal candidate genes associated with key functional traits. Runs of heterozygosity (ROHet) are commonly associated with balance selection, which can help us understand the adaptive evolutionary history of domestic animals. In this study, we investigated ROHs and ROHets in 88 Tunchang pigs. We also compared the estimates of inbreeding coefficients in individuals calculated based on four methods. In summary, we detected a total of 16 ROH islands in our study, and 100 genes were found within ROH regions. These genes were correlated with economically important traits such as reproduction (e.g., SERPIND1, HIRA), meat quality (e.g., PI4KA, TBX1), immunity (e.g., ESS2, RANBP1), adaption to heat stress (TXNRD2 and DGCR8), and crude food tolerance (TRPM6). Moreover, we discovered 18 ROHet islands harbouring genes associated with reproduction (e.g., ARHGEF12, BMPR2), immune system (e.g., BRD4, DNMT3B). These findings may help us design effective breeding and conservation strategies for this unique breed.


Asunto(s)
Heterocigoto , Homocigoto , Animales , Porcinos/genética , Endogamia , China , Sus scrofa/genética , Femenino , Genoma , Masculino
8.
Turk J Gastroenterol ; 35(8): 618-624, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-39150338

RESUMEN

Familial mediterranean fever (FMF) is a genetic autoinflammatory disease typically diagnosed in childhood. In this study, we aimed to investigate the demographic, clinical, and genetic characteristics of patients aged 18 years and older who were diagnosed with FMF. Patients diagnosed with FMF between 2014 and 2022 at Karadeniz Technical University Faculty of Medicine Hospital were included in the study. Patients were divided into 2 groups based on the age of disease onset. Group I included patients with adult-onset (ages 18-40), while group II comprised patients with late onset (ages 40 and above). Subsequently, the 2 groups were compared. A total of 150 patients with a mean age of 32 (18-79) were included in the study. There were 116 patients in group I and 34 (22.7%) in group II. The most common presenting complaint was abdominal pain (91.3%), and the most prevalent complication was amyloidosis (4.7%). No significant differences were observed between age groups regarding clinical findings and symptoms. The most frequent homozygous mutations were M694V (9.3%) and R202Q (1.8%), while the heterozygous mutations were M694V (37.3%) and R202Q (35.5%), respectively. The rate of M694V gene positivity in the adult-onset group was significantly higher compared to the lateonset group (52.9% and 25%, respectively, P = .020). There does not appear to be a significant difference in clinical signs and symptoms between adult-onset and late-onset FMF patients. The higher rate of M694V gene positivity in the adult-onset group suggests that the M694V mutation may be responsible for the early expression of the disease.


Asunto(s)
Edad de Inicio , Fiebre Mediterránea Familiar , Mutación , Pirina , Humanos , Fiebre Mediterránea Familiar/genética , Adulto , Masculino , Femenino , Persona de Mediana Edad , Adulto Joven , Adolescente , Anciano , Pirina/genética , Turquía/epidemiología , Dolor Abdominal/etiología , Amiloidosis/genética , Homocigoto , Heterocigoto
9.
BMC Genomics ; 25(1): 772, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39118059

RESUMEN

BACKGROUND: The Icelandic horse and Exmoor pony are ancient, native breeds, adapted to harsh environmental conditions and they have both undergone severe historic bottlenecks. However, in modern days, the selection pressures on these breeds differ substantially. The aim of this study was to assess genetic diversity in both breeds through expected (HE) and observed heterozygosity (HO) and effective population size (Ne). Furthermore, we aimed to identify runs of homozygosity (ROH) to estimate and compare genomic inbreeding and signatures of selection in the breeds. RESULTS: HO was estimated at 0.34 and 0.33 in the Icelandic horse and Exmoor pony, respectively, aligning closely with HE of 0.34 for both breeds. Based on genomic data, the Ne for the last generation was calculated to be 125 individuals for Icelandic horses and 42 for Exmoor ponies. Genomic inbreeding coefficient (FROH) ranged from 0.08 to 0.20 for the Icelandic horse and 0.12 to 0.27 for the Exmoor pony, with the majority of inbreeding attributed to short ROHs in both breeds. Several ROH islands associated with performance were identified in the Icelandic horse, featuring target genes such as DMRT3, DOCK8, EDNRB, SLAIN1, and NEURL1. Shared ROH islands between both breeds were linked to metabolic processes (FOXO1), body size, and the immune system (CYRIB), while private ROH islands in Exmoor ponies were associated with coat colours (ASIP, TBX3, OCA2), immune system (LYG1, LYG2), and fertility (TEX14, SPO11, ADAM20). CONCLUSIONS: Evaluations of genetic diversity and inbreeding reveal insights into the evolutionary trajectories of both breeds, highlighting the consequences of population bottlenecks. While the genetic diversity in the Icelandic horse is acceptable, a critically low genetic diversity was estimated for the Exmoor pony, which requires further validation. Identified signatures of selection highlight the differences in the use of the two breeds as well as their adaptive trait similarities. The results provide insight into genomic regions under selection pressure in a gaited performance horse breed and various adaptive traits in small-sized native horse breeds. This understanding contributes to preserving genetic diversity and population health in these equine populations.


Asunto(s)
Variación Genética , Homocigoto , Endogamia , Selección Genética , Caballos/genética , Animales , Islandia , Genómica/métodos , Polimorfismo de Nucleótido Simple , Heterocigoto , Cruzamiento , Genética de Población
10.
Mol Genet Genomic Med ; 12(8): e2500, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39101447

RESUMEN

BACKGROUND: Variations in the WWOX gene have been identified as the leading cause of several central nervous system disorders. However, most previous reports have focused on the description of clinical phenotype, neglecting functional verification. Herein, we presented a case of a patient with developmental epileptic encephalopathy (DEE) caused by WWOX gene variation. CASE PRESENTATION: Our patient was a 13-month-old girl with abnormal facial features, including facial hypotonia, arched eyebrows, a broad nose, and a depressed nasal bridge. She also had sparse and yellow hair, a low anterior hairline, and a short neck. Before the age of 8 months, she was suffering from mild seizures. Her developmental delay gradually worsened, and she suffered infantile spasms. After treatment with vigabatrin, seizures subsided. WWOX gene homozygous variation c.172+1G>C was identified using whole exome sequencing. Further minigene assay confirmed that the variation site affected splicing, causing protein truncation and affecting its function. CONCLUSION: Clinical phenotype and minigene results suggest that WWOX gene homozygous variation c.172+1G>C can cause severe DEE. We also concluded that vigabatrin can effectively treat seizures.


Asunto(s)
Homocigoto , Fenotipo , Espasmos Infantiles , Oxidorreductasa que Contiene Dominios WW , Humanos , Oxidorreductasa que Contiene Dominios WW/genética , Femenino , Lactante , Espasmos Infantiles/genética , Espasmos Infantiles/patología , Anticonvulsivantes/uso terapéutico , Mutación , Vigabatrin/uso terapéutico , Proteínas Supresoras de Tumor
12.
J Clin Immunol ; 44(8): 170, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39098944

RESUMEN

Advanced genomic technologies such as whole exome or whole genome sequencing have improved diagnoses and disease outcomes for individuals with genetic diseases. Yet, variants of unknown significance (VUS) require rigorous validation to establish disease causality or modification, or to exclude them from further analysis. Here, we describe a young individual of Polynesian ancestry who in the first 13 mo of life presented with SARS-CoV-2 pneumonia, severe enterovirus meningitis and adenovirus gastroenteritis, and severe adverse reaction to MMR vaccination. Genomic analysis identified a previously reported pathogenic homozygous variant in IFNAR1 (c.1156G > T, p.Glu386* LOF), which is common in Western Polynesia. Moreover, a new and putatively deleterious canonical splice site variant in DOCK8 was also found in homozygosity (c.3234 + 2T > C). This DOCK8 variant is common in Polynesians and other under-represented ancestries in large genomic databases. Despite in silico bioinformatic predictions, extensive in vitro and ex vivo analysis revealed the DOCK8 variant likely be neutral. Thus, our study reports a novel case of IFNAR1 deficiency, but also highlights the importance of functional validation of VUS, including those predicted to be deleterious, and the pressing need to expand our knowledge of the genomic architecture and landscape of under-represented populations and ancestries.


Asunto(s)
COVID-19 , Factores de Intercambio de Guanina Nucleótido , Receptor de Interferón alfa y beta , SARS-CoV-2 , Humanos , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/deficiencia , COVID-19/genética , SARS-CoV-2/genética , Lactante , Sitios de Empalme de ARN/genética , Masculino , Femenino , Mutación/genética , Homocigoto
13.
Anim Sci J ; 95(1): e13978, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38978175

RESUMEN

Genomic prediction was conducted using 2494 Japanese Black cattle from Hiroshima Prefecture and both single-nucleotide polymorphism information and phenotype data on monounsaturated fatty acid (MUFA) and oleic acid (C18:1) analyzed with gas chromatography. We compared the prediction accuracy for four models (A, additive genetic effects; AD, as for A with dominance genetic effects; ADR, as for AD with the runs of homozygosity (ROH) effects calculated by ROH-based relationship matrix; and ADF, as for AD with the ROH-based inbreeding coefficient of the linear regression). Bayesian methods were used to estimate variance components. The narrow-sense heritability estimates for MUFA and C18:1 were 0.52-0.53 and 0.57, respectively; the corresponding proportions of dominance genetic variance were 0.04-0.07 and 0.04-0.05, and the proportion of ROH variance was 0.02. The deviance information criterion values showed slight differences among the models, and the models provided similar prediction accuracy.


Asunto(s)
Teorema de Bayes , Polimorfismo de Nucleótido Simple , Animales , Bovinos/genética , Bovinos/metabolismo , Carácter Cuantitativo Heredable , Ácidos Grasos Monoinsaturados/análisis , Ácidos Grasos Monoinsaturados/metabolismo , Fenotipo , Ácido Oléico/análisis , Homocigoto , Genómica , Modelos Genéticos , Ácidos Grasos/análisis , Ácidos Grasos/metabolismo
14.
Ann Afr Med ; 23(2): 234-236, 2024 Apr 01.
Artículo en Francés, Inglés | MEDLINE | ID: mdl-39028172

RESUMEN

Methylenetetrahydrofolate reductase (MTHFR) enzyme is one of the key enzymes involved in the metabolism of folate. Mutations in this enzyme can lead to a procoagulant state. We present a case of a 20-year-old male with no known comorbidities, who presented with fever and hemoptysis and was diagnosed as a case of pulmonary embolism. He was found to have a homozygous mutation in the MTHFR gene that was responsible for his disease state. He was started on unfractionated heparin infusion and underwent catheter-directed thrombolysis. He showed marked improvement in his condition and was discharged on oral anticoagulants with an advice to follow-up.


RésuméL'enzyme méthylènetétrahydrofolate réductase (MTHFR) est l'une des enzymes clés impliquées dans le métabolisme du folate. Les mutations de cette enzyme peuvent conduire à un état procoagulant. Nous présentons le cas d'un homme de 20 ans sans comorbidités connues, qui s'est présenté avec de la fièvre et une hémoptysie et a été diagnostiqué comme un cas d'embolie pulmonaire. Il s'est avéré qu'il présentait une mutation homozygote du gène MTHFR responsable de son état pathologique. Il a commencé une perfusion d'héparine non fractionnée et a subi une thrombolyse dirigée par cathéter. Il a montré une nette amélioration de son état et a été libéré sous anticoagulants oraux avec un conseil de suivi.


Asunto(s)
Anticoagulantes , Metilenotetrahidrofolato Reductasa (NADPH2) , Mutación , Embolia Pulmonar , Humanos , Masculino , Embolia Pulmonar/tratamiento farmacológico , Embolia Pulmonar/genética , Embolia Pulmonar/diagnóstico , Embolia Pulmonar/diagnóstico por imagen , Anticoagulantes/uso terapéutico , Adulto Joven , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Resultado del Tratamiento , Heparina/uso terapéutico , Terapia Trombolítica/métodos , Homocigoto
15.
Mol Genet Genomics ; 299(1): 69, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38992144

RESUMEN

TTC12 is a cytoplasmic and centromere-localized protein that plays a role in the proper assembly of dynein arm complexes in motile cilia in both respiratory cells and sperm flagella. This finding underscores its significance in cellular motility and function. However, the wide role of TTC12 in human spermatogenesis-associated primary ciliary dyskinesia (PCD) still needs to be elucidated. Whole-exome sequencing (WES) and Sanger sequencing were performed to identify potentially pathogenic variants causing PCD and multiple morphological abnormalities of sperm flagella (MMAF) in an infertile Pakistani man. Diagnostic imaging techniques were used for PCD screening in the patient. Real-time polymerase chain reaction (RT‒PCR) was performed to detect the effect of mutations on the mRNA abundance of the affected genes. Papanicolaou staining and scanning electron microscopy (SEM) were carried out to examine sperm morphology. Transmission electron microscopy (TEM) was performed to examine the ultrastructure of the sperm flagella, and the results were confirmed by immunofluorescence staining. Using WES and Sanger sequencing, a novel homozygous missense variant (c.C1069T; p.Arg357Trp) in TTC12 was identified in a patient from a consanguineous family. A computed tomography scan of the paranasal sinuses confirmed the symptoms of the PCD. RT-PCR showed a decrease in TTC12 mRNA in the patient's sperm sample. Papanicolaou staining, SEM, and TEM analysis revealed a significant change in shape and a disorganized axonemal structure in the sperm flagella of the patient. Immunostaining assays revealed that TTC12 is distributed throughout the flagella and is predominantly concentrated in the midpiece in normal spermatozoa. In contrast, spermatozoa from patient deficient in TTC12 showed minimal staining intensity for TTC12 or DNAH17 (outer dynein arms components). This could lead to MMAF and result in male infertility. This novel TTC12 variant not only illuminates the underlying genetic causes of male infertility but also paves the way for potential treatments targeting these genetic factors. This study represents a significant advancement in understanding the genetic basis of PCD-related infertility.


Asunto(s)
Homocigoto , Infertilidad Masculina , Mutación Missense , Cola del Espermatozoide , Humanos , Masculino , Mutación Missense/genética , Pakistán , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Cola del Espermatozoide/patología , Cola del Espermatozoide/ultraestructura , Cola del Espermatozoide/metabolismo , Adulto , Linaje , Astenozoospermia/genética , Astenozoospermia/patología , Trastornos de la Motilidad Ciliar/genética , Trastornos de la Motilidad Ciliar/patología , Secuenciación del Exoma , Oligospermia/genética , Oligospermia/patología , Síndrome de Kartagener/genética , Síndrome de Kartagener/patología
16.
BMC Genom Data ; 25(1): 68, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38982354

RESUMEN

The recent chromosome-based genome assembly and the newly developed 70K single nucleotide polymorphism (SNP) array for American mink (Neogale vison) facilitate the identification of genetic variants underlying complex traits in this species. The objective of this study was to evaluate the association between consensus runs of homozygosity (ROH) with growth and feed efficiency traits in American mink. A subsample of two mink populations (n = 2,986) were genotyped using the Affymetrix Mink 70K SNP array. The identified ROH segments were included simultaneously, concatenated into consensus regions, and the ROH-based association studies were carried out with linear mixed models considering a genomic relationship matrix for 11 growth and feed efficiency traits implemented in ASReml-R version 4. In total, 298,313 ROH were identified across all individuals, with an average length and coverage of 4.16 Mb and 414.8 Mb, respectively. After merging ROH segments, 196 consensus ROH regions were detected and used for genome-wide ROH-based association analysis. Thirteen consensus ROH regions were significantly (P < 0.01) associated with growth and feed efficiency traits. Several candidate genes within the significant regions are known for their involvement in growth and body size development, including MEF2A, ADAMTS17, POU3F2, and TYRO3. In addition, we found ten consensus ROH regions, defined as ROH islands, with frequencies over 80% of the population. These islands harbored 12 annotated genes, some of which were related to immune system processes such as DTX3L, PARP9, PARP14, CD86, and HCLS1. This is the first study to explore the associations between homozygous regions with growth and feed efficiency traits in American mink. Our findings shed the light on the effects of homozygosity in the mink genome on growth and feed efficiency traits, that can be utilized in developing a sustainable breeding program for mink.


Asunto(s)
Homocigoto , Visón , Polimorfismo de Nucleótido Simple , Animales , Visón/genética , Visón/crecimiento & desarrollo , Polimorfismo de Nucleótido Simple/genética , Estudio de Asociación del Genoma Completo/veterinaria , Alimentación Animal , Fenotipo
17.
Mol Genet Genomic Med ; 12(7): e2452, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38967264

RESUMEN

BACKGROUND: Inherited glycosylphosphatidylinositol (GPI) deficiency is an autosomal recessive disease and a set of syndromes caused by different genes involved in the biosynthesis of phosphatidylinositol characterized by severe cognitive disability, elevated serum alkaline phosphatase (ALP) levels, and distinct facial features. This report presents a patient with inherited GPI deficiency caused by a homozygous frameshift variant of PGAP3 due to uniparental isodisomy (UPiD) on chromosome 17. METHOD: Clinical characteristics of the patient were collected. Microarray analysis followed by adaptive sampling sequencing targeting chromosome 17 was used for the identification of variants. Sanger sequencing was used to confirm the variant in the target region. RESULTS: The patient was born at 38 weeks of gestation with a birthweight of 3893 g. He had a distinctive facial appearance with hypertelorism, wide nasal bridge, and cleft soft palate. Postnatal head magnetic resonance imaging revealed a Blake's pouch cyst. The serum ALP level was 940 IU/L at birth and increased to 1781 IU/L at 28 days of age. Microarray analysis revealed region of homozygosity in nearly the entire region of chromosome 17, leading to the diagnosis of UPiD. Adaptive sampling sequencing targeting chromosome 17 confirmed the homozygous variant NM_033419:c.778dupG (p.Val260Glyfs*14) in the PGAP3 gene, resulting in a diagnosis of inherited GPI deficiency. CONCLUSION: This is the first report of inherited GPI deficiency caused by UPiD. Inherited GPI deficiency must be considered in patients with unexplained hyperphosphatasemia.


Asunto(s)
Glicosilfosfatidilinositoles , Disomía Uniparental , Humanos , Masculino , Hidrolasas de Éster Carboxílico , Mutación del Sistema de Lectura , Glicosilfosfatidilinositoles/deficiencia , Glicosilfosfatidilinositoles/genética , Homocigoto , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/patología , Trastornos del Metabolismo del Fósforo/genética , Trastornos del Metabolismo del Fósforo/patología , Receptores de Superficie Celular , Convulsiones , Disomía Uniparental/genética , Disomía Uniparental/patología , Recién Nacido
18.
Artículo en Inglés | MEDLINE | ID: mdl-39008646

RESUMEN

Thromboembolic events are a common cause of morbidity and mortality with significant socioeconomic impact especially when young patients are affected. They are a rare medical event in young people and their clinical presentation can be mild or asymptomatic. The manifestation of symptoms and thrombotic events depends on both: the genetic mutations and the external risk factors that will induce the process. We present a case of a 34-year old young female, with three consecutive cerebrovascular insults in a period of ten years, and an acute myocardial infarction. There is a combination of gene mutations and polymorphism, with a predisposition to thromboembolic events. We emphasized the role of e-NOS (Endothelial nitric oxide synthase 786 T>C mutation) and the connection with smoking. The dual effect of the prolonged smoking and dysfunctional nitric oxide synthase in our young patient led to several thrombotic events. We discussed the various diagnostic tests and possible therapeutic and prophylactic strategies.


Asunto(s)
Predisposición Genética a la Enfermedad , Mutación , Óxido Nítrico Sintasa de Tipo III , Tromboembolia , Humanos , Femenino , Óxido Nítrico Sintasa de Tipo III/genética , Adulto , Tromboembolia/genética , Homocigoto , Factores de Riesgo , Fumar/efectos adversos , Infarto del Miocardio/genética , Fenotipo
19.
Hear Res ; 451: 109091, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39067415

RESUMEN

Sgms1 encodes sphingomyelin synthase 1, an enzyme in the sphingosine-1-phosphate signalling pathway, and was previously reported to underlie hearing impairment in the mouse. A new mouse allele, Sgms1tm1a, unexpectedly showed normal Auditory Brainstem Response thresholds. We found that the Sgms1tm1a mutation led to incomplete knockdown of transcript to 20 % of normal values, which was enough to support normal hearing. The Sgms1tm1b allele was generated by knocking out exon 7, leading to a complete lack of detectable transcript in the inner ear. Sgms1tm1b homozygotes showed largely normal auditory brainstem response thresholds at first, followed by progressive loss of sensitivity until they showed severe impairment at 6 months old. The endocochlear potential was consistently reduced in Sgms1tm1b mutants at 3, 4 and 8 weeks old, to around 80 mV compared with around 120 mV in control littermates. The stria vascularis showed a characteristic irregularity of marginal cell surfaces and patchy loss of Kcnq1 expression at their apical membrane, and expression analysis of the lateral wall suggested that marginal cells were the most likely initial site of dysfunction in the mutants. Finally, significant association of auditory thresholds with DNA markers within and close to the human SGMS1 gene were found in the 1958 Birth Cohort, suggesting that SGMS1 variants may play a role in the range of hearing abilities in the human population.


Asunto(s)
Potenciales Evocados Auditivos del Tronco Encefálico , Pérdida Auditiva , Estría Vascular , Transferasas (Grupos de Otros Fosfatos Sustitutos) , Animales , Femenino , Masculino , Ratones , Umbral Auditivo , Cóclea/fisiopatología , Cóclea/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Audición/genética , Pérdida Auditiva/genética , Pérdida Auditiva/fisiopatología , Homocigoto , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fenotipo , Estría Vascular/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética
20.
Genet Test Mol Biomarkers ; 28(7): 289-296, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38980801

RESUMEN

Introduction: The genetics of hereditary hemochromatosis (HH) is understudied in Iran. Here, we report the result of genetic screening of 854 individuals, referred as "suspected cases of HH," to a diagnostic laboratory in Iran over a 12-year period. Materials and Methods: From 2011 to 2012, 121 cases were screened for HH using Sanger sequencing of HFE exons. After 2012, this method was replaced by a commercial reverse hybridization assay (RHA) targeting 18 variants in the HFE, TFR2, and FPN1(SLC40A1) genes and 733 cases were screened using this method. Results: From the total studied population, HH was confirmed by genetic diagnosis in only seven cases (0.82%): two homozygotes for HFE:C282Y and five homozygotes for TFR2:AVAQ 594-597 deletion. In 254 cases (29.7%), H63D, C282Y, S65C, and four other HFE variants not targeted by RHA were identified. Although the resulting genotypes in the latter cases did not confirm HH, some of them were known modifying factors of iron overload or could cause HH in combination with a possibly undetected variant. No variant was detected in 593 cases (69.4%). Conclusion: This study showed that the spectrum of genetic variants of HH in the Iranian population includes HFE and TFR2 variants. However, HH was not confirmed in the majority (99.2%) of suspected cases. This could be explained by limitations of our genetic diagnostics and possible inaccuracies in clinical suspicion of HH. A cooperative clinical and genetic investigation is proposed as a solution to this issue.


Asunto(s)
Pruebas Genéticas , Proteína de la Hemocromatosis , Hemocromatosis , Receptores de Transferrina , Humanos , Hemocromatosis/genética , Hemocromatosis/diagnóstico , Irán/epidemiología , Proteína de la Hemocromatosis/genética , Masculino , Femenino , Pruebas Genéticas/métodos , Adulto , Persona de Mediana Edad , Receptores de Transferrina/genética , Homocigoto , Anciano , Exones/genética , Genotipo , Antígenos de Histocompatibilidad Clase I/genética , Mutación , Proteínas de Transporte de Catión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...