Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 266
Filtrar
1.
Cells ; 12(13)2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37443734

RESUMEN

Neural tube defects (NTDs), including anencephaly and spina bifida, are common major malformations of fetal development resulting from incomplete closure of the neural tube. These conditions lead to either universal death (anencephaly) or severe lifelong complications (spina bifida). Despite hundreds of genetic mouse models of neural tube defect phenotypes, the genetics of human NTDs are poorly understood. Furthermore, pharmaceuticals, such as antiseizure medications, have been found clinically to increase the risk of NTDs when administered during pregnancy. Therefore, a model that recapitulates human neurodevelopment would be of immense benefit to understand the genetics underlying NTDs and identify teratogenic mechanisms. Using our self-organizing single rosette cortical organoid (SOSR-COs) system, we have developed a high-throughput image analysis pipeline for evaluating the SOSR-CO structure for NTD-like phenotypes. Similar to small molecule inhibition of apical constriction, the antiseizure medication valproic acid (VPA), a known cause of NTDs, increases the apical lumen size and apical cell surface area in a dose-responsive manner. GSK3ß and HDAC inhibitors caused similar lumen expansion; however, RNA sequencing suggests VPA does not inhibit GSK3ß at these concentrations. The knockout of SHROOM3, a well-known NTD-related gene, also caused expansion of the lumen, as well as reduced f-actin polarization. The increased lumen sizes were caused by reduced cell apical constriction, suggesting that impingement of this process is a shared mechanism for VPA treatment and SHROOM3-KO, two well-known causes of NTDs. Our system allows the rapid identification of NTD-like phenotypes for both compounds and genetic variants and should prove useful for understanding specific NTD mechanisms and predicting drug teratogenicity.


Asunto(s)
Anencefalia , Defectos del Tubo Neural , Disrafia Espinal , Embarazo , Femenino , Humanos , Ratones , Animales , Ácido Valproico/farmacología , Anencefalia/complicaciones , Anencefalia/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Ratones Noqueados , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/genética , Disrafia Espinal/genética , Encéfalo/patología , Proteínas de Microfilamentos
2.
Arch Gynecol Obstet ; 307(1): 293-299, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35554661

RESUMEN

OBJECTIVES: To analyse additional structural and genetic anomalies in fetuses with acrania/exencephaly/anencephaly sequence (AEAS). METHODS: A retrospective analysis of 139 fetuses with AEAS diagnosed between 2006 and 2020 in a single tertiary referral ultrasound department. RESULTS: The median gestational age at diagnosis decreased from 15 weeks in 2006 to 13 weeks in 2020 (- 0.21 per each year; p = 0.009). In 103 fetuses, the defects were limited to the neural tube (NTD) (74.1%), in 36 fetuses (25.9%), there were additional structural non-NTD anomalies. The most common were ventral body wall defects present in 17.8% (23/139), followed by anomalies of the limbs (7.2%; 10/139), face (6.5%; 9/139) and heart (6.5%; 9/139). Genetic anomalies were diagnosed in 7 of the 74 conclusive results (9.5%; 7/74; trisomy 18, n = 5; triploidy, n = 1; duplication of Xq, n = 1). In univariate logistic regression models, male sex, limb anomalies and ventral body wall defects significantly increased the risk of genetic anomalies (OR 12.3; p = 0.024; OR 16.5; p = 0.002 and OR 10.4; p = 0.009, respectively). CONCLUSIONS: A significant number of fetuses with AEAS have additional structural non-NTD anomalies, which are mostly consistent with limb body wall complex. Genetic abnormalities are diagnosed in almost 10% of affected fetuses and trisomy 18 is the most common aberration. Factors that significantly increased the odds of genetic anomalies in fetuses with AEAS comprise male sex, limb anomalies and ventral body wall defects.


Asunto(s)
Anencefalia , Defectos del Tubo Neural , Embarazo , Femenino , Masculino , Humanos , Lactante , Anencefalia/diagnóstico por imagen , Anencefalia/epidemiología , Anencefalia/genética , Síndrome de la Trisomía 18 , Estudios Retrospectivos , Defectos del Tubo Neural/diagnóstico por imagen , Defectos del Tubo Neural/epidemiología , Diagnóstico Prenatal
3.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33941697

RESUMEN

Gap closure is a common morphogenetic process. In mammals, failure to close the embryonic hindbrain neuropore (HNP) gap causes fatal anencephaly. We observed that surface ectoderm cells surrounding the mouse HNP assemble high-tension actomyosin purse strings at their leading edge and establish the initial contacts across the embryonic midline. Fibronectin and laminin are present, and tensin 1 accumulates in focal adhesion-like puncta at this leading edge. The HNP gap closes asymmetrically, faster from its rostral than caudal end, while maintaining an elongated aspect ratio. Cell-based physical modeling identifies two closure mechanisms sufficient to account for tissue-level HNP closure dynamics: purse-string contraction and directional cell motion implemented through active crawling. Combining both closure mechanisms hastens gap closure and produces a constant rate of gap shortening. Purse-string contraction reduces, whereas crawling increases gap aspect ratio, and their combination maintains it. Closure rate asymmetry can be explained by asymmetric embryo tissue geometry, namely a narrower rostral gap apex, whereas biomechanical tension inferred from laser ablation is equivalent at the gaps' rostral and caudal closure points. At the cellular level, the physical model predicts rearrangements of cells at the HNP rostral and caudal extremes as the gap shortens. These behaviors are reproducibly live imaged in mouse embryos. Thus, mammalian embryos coordinate cellular- and tissue-level mechanics to achieve this critical gap closure event.


Asunto(s)
Embrión de Mamíferos/metabolismo , Cresta Neural/metabolismo , Tubo Neural/metabolismo , Rombencéfalo/metabolismo , Anencefalia/embriología , Anencefalia/genética , Anencefalia/metabolismo , Animales , Cadherinas/metabolismo , Embrión de Mamíferos/embriología , Femenino , Fibronectinas/metabolismo , Laminina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal/métodos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Cresta Neural/embriología , Tubo Neural/embriología , Rombencéfalo/embriología , Imagen de Lapso de Tiempo/métodos
5.
J Exp Med ; 217(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-32845958

RESUMEN

Failure of neural tube closure during embryonic development can result in anencephaly, one of the most common birth defects in humans. A family with recurrent anencephalic fetuses was investigated to understand its etiology and pathogenesis. Exome sequencing revealed a recessive germline 21-bp in-frame deletion in NUAK2 segregating with the disease. In vitro kinase assays demonstrated that the 7-amino acid truncation in NUAK2, a serine/threonine kinase, completely abrogated its catalytic activity. Patient-derived disease models including neural progenitor cells and cerebral organoids showed that loss of NUAK2 activity led to decreased Hippo signaling via cytoplasmic YAP retention. In neural tube-like structures, endogenous NUAK2 colocalized apically with the actomyosin network, which was disrupted in patient cells, causing impaired nucleokinesis and apical constriction. Our results establish NUAK2 as an indispensable kinase for brain development in humans and suggest that a NUAK2-Hippo signaling axis regulates cytoskeletal processes that govern cell shape during neural tube closure.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anencefalia/genética , Mutación con Pérdida de Función/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Agregación Celular , Consanguinidad , Regulación hacia Abajo/genética , Femenino , Feto/patología , Genes Recesivos , Vía de Señalización Hippo , Humanos , Masculino , Células-Madre Neurales/metabolismo , Tubo Neural/patología , Organoides/patología , Linaje , Dominios Proteicos , Proteínas Serina-Treonina Quinasas/química , Transducción de Señal , Transcripción Genética , Turquía , Proteínas Señalizadoras YAP
6.
Clin Genet ; 98(3): 299-302, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32621286

RESUMEN

Neural tube defects (NTD) are among the most common congenital anomalies, affecting about 1:1000 births. In most cases, the etiology of NTD is multifactorial and the genetic variants associated with them remain largely unknown. There is extensive evidence from animal models over the past two decades implicating SHROOM3 in neural tube formation; however, its exact role in human disease has remained elusive. In this report, we present the first case of a human fetus with a homozygous loss of function variant in SHROOM3. The fetus presents with anencephaly and cleft lip and palate, similar to previously described Shroom3 mouse mutants and is suggestive of a novel monogenic cause of NTD. Our case provides clarification on the contribution of SHROOM3 to human development after decades of model organism research.


Asunto(s)
Anencefalia/genética , Labio Leporino/genética , Fisura del Paladar/genética , Proteínas de Microfilamentos/genética , Anencefalia/complicaciones , Anencefalia/patología , Labio Leporino/complicaciones , Labio Leporino/patología , Fisura del Paladar/complicaciones , Fisura del Paladar/patología , Femenino , Feto , Homocigoto , Humanos , Mutación con Pérdida de Función/genética , Análisis por Micromatrices , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología
7.
J Hum Genet ; 65(11): 985-993, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32576942

RESUMEN

Neural tube defects (NTDs) are debilitating human congenital abnormalities due to failure of neural tube closure. Sonic Hedgehog (SHH) signaling is required for dorsal-ventral patterning of the neural tube. The loss of activation in SHH signaling normally causes holoprosencephaly while the loss of inhibition causes exencephaly due to failure in neural tube closure. WDR34 is a dynein intermedia chain component which is required for SHH activation. However, Wdr34 knockout mouse exhibit exencephaly. Here we screened mutations in WDR34 gene in 100 anencephaly patients of Chinese Han population. Compared to 1000 Genome Project data, two potentially disease causing missense mutations of WDR34 gene (c.1177G>A; p.G393S and c.1310A>G; p.Y437C) were identified in anencephaly patients. These two mutations did not affect the protein expression level of WDR34. Luciferase reporter and endogenous target gene expression level showed that both mutations are lose-of-function mutations in SHH signaling. Surprisingly, WDR34 could promote planar cell polarity (PCP) signaling and the G393S lost this promoting effect on PCP signaling. Morpholino knockdown of wdr34 in zebrafish caused severe convergent extension defects and pericardial abnormalities. The G393S mutant has less rescuing effects than both WT and Y437C WDR34 in zebrafish. Our results suggested that mutation in WDR34 could contribute to human NTDs by affecting both SHH and PCP signaling.


Asunto(s)
Anencefalia/genética , Proteínas Portadoras/genética , Proteínas Hedgehog/genética , Defectos del Tubo Neural/genética , Adulto , Anencefalia/patología , Animales , Polaridad Celular/genética , Anomalías Congénitas/genética , Anomalías Congénitas/patología , Femenino , Regulación del Desarrollo de la Expresión Génica , Genoma Humano/genética , Humanos , Masculino , Defectos del Tubo Neural/patología , Adulto Joven , Pez Cebra/genética
8.
Epigenetics Chromatin ; 12(1): 22, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30992047

RESUMEN

BACKGROUND: Neural tube defects (NTDs) are common birth defects involving the central nervous system. Recent studies on the etiology of human NTDs have raised the possibility that epigenetic regulation could be involved in determining susceptibility to them. RESULTS: Here, we show that the H2AK119ub1 E3 ligase CUL4B is required for the activation of retinoic acid (RA)-inducible developmentally critical homeobox (HOX) genes in NT2/D1 embryonal carcinoma cells. RA treatment led to attenuation of H2AK119ub1 due to decrease in CUL4B, further affecting HOX gene regulation. Furthermore, we found that CUL4B interacted directly with RORγ and negatively regulated its transcriptional activity. Interestingly, knockdown of RORγ decreased the expression of HOX genes along with increased H2AK119ub1 occupancy levels, at HOX gene sites in N2/D1 cells. In addition, upregulation of HOX genes was observed along with lower levels of CUL4B-mediated H2AK119ub1 in both mouse and human anencephaly NTD cases. Notably, the expression of HOXA10 genes was negatively correlated with CUL4B levels in human anencephaly NTD cases. CONCLUSIONS: Our results indicate that abnormal HOX gene expression induced by aberrant CUL4B-mediated H2AK119ub1 levels may be a risk factor for NTDs, and highlight the need for further analysis of genome-wide epigenetic modifications in NTDs.


Asunto(s)
Anencefalia/genética , Proteínas Cullin/genética , Código de Histonas , Histonas/metabolismo , Proteínas de Homeodominio/genética , Ubiquitinación , Animales , Línea Celular , Línea Celular Tumoral , Proteínas Cullin/metabolismo , Histonas/genética , Proteínas Homeobox A10 , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Unión Proteica
9.
Med Sci Monit ; 24: 5015-5026, 2018 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-30022792

RESUMEN

BACKGROUND DNA Base Excision Repair Gene-DNA LigaseIII (LIG3) is an important repair gene in the repair pathway and plays an important role in maintaining the integrity of mitochondria. Rs1052536 and rs3135967 polymorphisms of the gene are associated with lung cancer, keratoconus, and Fuchs endothelial corneal dystrophy. There is no previously published report on the relationship between the polymorphisms and neural tube defects (NTDs). MATERIAL AND METHODS Mass ARRAY iPLEX was used to determine the distribution of the polymorphisms in the case group of 108 NTD pregnant women and a control group of 233 normal healthy pregnant women to examine the relevance of their polymorphisms and NTD occurrence. RESULTS The homozygotes of rs1052536 TT were associated with an increased risk for NTDs than CC (P=0.014, OR=2.31, 95%CI [1.17-4.54]), and variants of rs1052536 T were associated with an increased risk of NTDs (P=0.024, OR=1.50, 95%CI [1.06-2.13]). The stratified analysis showed that TT genotype of rs1052536 increased the risk of anencephaly (P=0.016, OR=2.69, 95%CI [1.18-6.10]) and the T allele significantly increased the risk of cranial NTDs (P=0.033, OR=1.56, 95%CI [1.04-2.35]). CONCLUSIONS Rs1052536 in LIG3 gene might be a potential genetic risk factor in a high-risk area of NTDs in China.


Asunto(s)
ADN Ligasa (ATP)/genética , Defectos del Tubo Neural/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Adulto , Alelos , Anencefalia/genética , Estudios de Casos y Controles , China/epidemiología , ADN Ligasa (ATP)/fisiología , Reparación del ADN/genética , Femenino , Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad , Humanos , Proteínas de Unión a Poli-ADP-Ribosa/fisiología , Polimorfismo de Nucleótido Simple/genética , Embarazo , Factores de Riesgo , Adulto Joven
10.
Biomarkers ; 23(7): 640-646, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29708443

RESUMEN

OBJECTIVE: Previous studies have not used family-based methods to evaluate maternal-paternal genetic effects of the folate metabolizing enzyme, dihydro folate reductase (DHFR) essential during embryogenesis. Present study focuses on evaluating the association and influence of parental genetic effects of DHFR 19 bp deletion in the development of foetal neural tube defects (NTDs) using family-based triad approach. MATERIALS AND METHODS: The study population (n = 924) including 124 NTD case-parent trios (n = 124 × 3 = 372) and 184 healthy control-parent trios (n = 184 × 3 = 552) from Telangana, India, was genotyped for DHFR 19 bp deletion. Statistical analysis was used by SPSS and parent-of-origin effects (POE). RESULTS: Foetuses with deletion genotype (DD) were at risk of developing anencephaly (OR = 3.26, p = 0.020). Among parents, increased maternal risk of having an anencephaly foetus (OR = 2.66, p = 0.028) was observed in mothers with DD genotype. In addition, POE analysis also demonstrated higher risk of maternal transmission of the deletion allele to anencephaly foetus compared with paternal transmission (OR = 6.00, p = 0.016). Interestingly, maternal-paternal-offspring genotype incompatibility revealed maternal deletion genotype (DD) in association with paternal heterozygous deletion genotype (WD) significantly increased risk for NTDs (OR = 5.29, p = 0.013). CONCLUSIONS: This study, using family-based case-parent and control-parent triad approach, is the first to report influence of maternal transmission of DHFR 19 bp deletion in the development of anencephaly in the foetus.


Asunto(s)
Anencefalia/genética , Predisposición Genética a la Enfermedad , Herencia Materna/genética , Eliminación de Secuencia , Tetrahidrofolato Deshidrogenasa/genética , Familia , Femenino , Desarrollo Fetal/genética , Genotipo , Humanos , Masculino , Factores de Riesgo
11.
Hum Genet ; 137(3): 195-202, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29423651

RESUMEN

Neural tube defects (NTDs), which include spina bifida and anencephaly, are the second most common form of human structural congenital malformations. While it is well established that SHROOM3 plays a pivotal role in the complex morphogenetic processes involved in neural tube closure (NTC), the underlying genetic contributions of SHROOM gene family members in the etiology of human NTDs remain poorly understood. Herein, we systematically investigated the mutation patterns of SHROOM1-4 in a Chinese population composed of 343 NTD cases and 206 controls, using targeted next-generation sequencing. Functional variants were further confirmed by western blot and the mammalian two-hybrid assays. Loss of function (LoF) variants were identified in SHROOM3. We observed 1.56 times as many rare [minor allele frequency (MAF) < 0.01] coding variants (p = 2.9 × 10-3) in SHROOM genes, and 4.5 times as many rare D-Mis (deleterious missense) variants in SHROOM2 genes in the NTD cases compared with the controls. D-Mis variants of SHROOM2 (p.A1331S; p.R1557H) were confirmed by Sanger sequencing, and these variants were determined to have profound effects on gene function that disrupted their binding with ROCK1 in vitro. These findings provide genetic and molecular insights into the effects of rare damaging variants in SHROOM2, indicating that such variants of SHROOM2 might contribute to the risk of human NTDs. This research enhances our understanding of the genetic contribution of the SHROOM gene family to the etiology of human NTDs.


Asunto(s)
Anencefalia/genética , Proteínas de la Membrana/genética , Proteínas de Microfilamentos/genética , Defectos del Tubo Neural/genética , Feto Abortado , Anencefalia/fisiopatología , China , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Humanos , Mutación con Pérdida de Función/genética , Masculino , Proteínas de la Membrana/química , Proteínas de Microfilamentos/química , Defectos del Tubo Neural/fisiopatología , Disrafia Espinal/genética , Disrafia Espinal/fisiopatología
12.
Clin Genet ; 93(4): 870-879, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29205322

RESUMEN

Neural tube defects (NTDs) affecting the brain (anencephaly) are lethal before or at birth, whereas lower spinal defects (spina bifida) may lead to lifelong neurological handicap. Collectively, NTDs rank among the most common birth defects worldwide. This study focuses on anencephaly, which despite having a similar frequency to spina bifida and being the most common type of NTD observed in mouse models, has had more limited inclusion in genetic studies. A genetic influence is strongly implicated in determining risk of NTDs and a molecular diagnosis is of fundamental importance to families both in terms of understanding the origin of the condition and for managing future pregnancies. Here we used a custom panel of 191 NTD candidate genes to screen 90 patients with cranial NTDs (n = 85 anencephaly and n = 5 craniorachischisis) with a targeted exome sequencing platform. After filtering and comparing to our in-house control exome database (N = 509), we identified 397 rare variants (minor allele frequency, MAF < 1%), 21 of which were previously unreported and predicted damaging. This included 1 frameshift (PDGFRA), 2 stop-gained (MAT1A; NOS2) and 18 missense variations. Together with evidence for oligogenic inheritance, this study provides new information on the possible genetic causation of anencephaly.


Asunto(s)
Anencefalia/genética , Epistasis Genética , Defectos del Tubo Neural/genética , Disrafia Espinal/genética , Anencefalia/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Estudios de Asociación Genética , Humanos , Masculino , Ratones , Mutación , Defectos del Tubo Neural/fisiopatología , Fenotipo , Embarazo , Cráneo/anomalías , Cráneo/fisiopatología , Disrafia Espinal/fisiopatología , Secuenciación del Exoma
13.
Taiwan J Obstet Gynecol ; 56(4): 550-553, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28805617

RESUMEN

OBJECTIVE: We present molecular cytogenetic characterization of a duplication of 15q24.2-q26.2 associated with anencephaly and neural tube defect (NTD). CASE REPORT: A 35-year-old pregnant woman was found to have a fetus with anencephaly by prenatal ultrasound at 12 weeks of gestation. The pregnancy was subsequently terminated, and a malformed fetus was delivered with anencephaly. Cytogenetic analysis of the cultured placental tissues revealed a karyotype of 46,XX,dup(15) (q24.2q26.2). Parental karyotypes were normal. Array comparative genomic hybridization analysis of the placental tissues revealed a 20.36-Mb duplication of 15q24.2-q26.2 encompassing 100 Online Mendelian Inheritance of in Man (OMIM) genes including LINGO1, MTHFS, KIF7 and CHD2. Metaphase fluorescence in situ hybridization analysis using 15q25.1-specidic probe confirmed a duplication of 15q25.1. Polymorphic DNA marker analysis showed a maternal origin of the duplication. CONCLUSION: A duplication of chromosome 15q24.2-q26.2 can be associated with NTD.


Asunto(s)
Anencefalia/genética , Defectos del Tubo Neural/genética , Trisomía/genética , Aborto Inducido , Adulto , Anencefalia/embriología , Cromosomas Humanos Par 15/genética , Hibridación Genómica Comparativa , Análisis Citogenético , Femenino , Humanos , Hibridación Fluorescente in Situ , Cariotipo , Cariotipificación , Defectos del Tubo Neural/embriología , Embarazo , Ultrasonografía Prenatal
14.
Hum Mol Genet ; 26(6): 1104-1114, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28087737

RESUMEN

Anencephaly (APH) is characterized by the absence of brain tissues and cranium. During primary neurulation stage of the embryo, the rostral part of the neural pore fails to close, leading to APH. APH shows a heterogeneous etiology, ranging from environmental to genetic causes. The autosomal recessive inheritance of APH has been reported in several populations. In this study, we employed whole-exome sequencing and identified a homozygous missense mutation c.1522C > A (p.Pro508Thr) in the TRIM36 gene as the cause of autosomal recessive APH in an Indian family. The TRIM36 gene is expressed in the developing brain, suggesting a role in neurogenesis. In silico analysis showed that proline at codon position 508 is highly conserved in 26 vertebrate species, and the mutation is predicted to affect the conformation of the B30.2/SPRY domain of TRIM36. Both in vitro and in vivo results showed that the mutation renders the TRIM36 protein less stable. TRIM36 is known to associate with microtubules. Transient expression of the mutant TRIM36 in HeLa and LN229 cells resulted in microtubule disruption, disorganized spindles, loosely arranged chromosomes, multiple spindles, abnormal cytokinesis, reduced cell proliferation and increased apoptosis as compared with cells transfected with its wild-type counterpart. The siRNA knock down of TRIM36 in HeLa and LN229 cells also led to reduced cell proliferation and increased apoptosis. We suggest that microtubule disruption and disorganized spindles mediated by mutant TRIM36 affect neural cell proliferation during neural tube formation, leading to APH.


Asunto(s)
Anencefalia/epidemiología , Anencefalia/genética , Proteínas Portadoras/genética , Mutación/genética , Anencefalia/fisiopatología , Exoma/genética , Femenino , Feto , Homocigoto , Humanos , India/epidemiología , Masculino , Linaje
15.
Nutrients ; 8(11)2016 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-27886045

RESUMEN

Neural tube defects (NTDs) are a group of severe congenital malformations, induced by the combined effects of genes and the environment. The most valuable finding so far has been the protective effect of folic acid supplementation against NTDs. However, many women do not take folic acid supplements until they are pregnant, which is too late to prevent NTDs effectively. Long-term intake of folic acid-fortified food is a good choice to solve this problem, and mandatory folic acid fortification should be further promoted, especially in Europe, Asia and Africa. Vitamin B2, vitamin B-6, vitamin B-12, choline, betaine and n-3 polyunsaturated fatty acids (PUFAs) can also reduce the NTD risk by interacting with the one-carbon metabolism pathway. This suggest that multivitamin B combined with choline, betaine and n-3 PUFAs supplementation may have a better protective effect against NTDs than folic acid alone. Genetic polymorphisms involved in one-carbon metabolism are associated with NTD risk, and gene screening for women of childbearing age prior to pregnancy may help prevent NTDs induced by the risk allele. In addition, the consumption of alcohol, tea and coffee, and low intakes of fruit and vegetable are also associated with the increased risk of NTDs, and should be avoided by women of childbearing age.


Asunto(s)
Anencefalia/metabolismo , Anencefalia/prevención & control , Carbono/metabolismo , Suplementos Dietéticos , Deficiencia de Ácido Fólico/terapia , Ácido Fólico/administración & dosificación , Fenómenos Fisiologicos Nutricionales Maternos , Estado Nutricional , Anencefalia/genética , Anencefalia/fisiopatología , Animales , Femenino , Deficiencia de Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/fisiopatología , Alimentos Fortificados , Interacción Gen-Ambiente , Humanos , Valor Nutritivo , Embarazo , Ingesta Diaria Recomendada , Factores de Riesgo
16.
Birth Defects Res A Clin Mol Teratol ; 106(8): 685-95, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27166990

RESUMEN

BACKGROUND: After years of periconceptional folic acid supplementation, the prevalence of neural tube defects (NTDs) remains stable following the remarkable reduction observed immediately after the fortification practice. There is accumulating evidence that folate receptor (FR) autoimmunity may play a role in the etiology of folate-sensitive NTDs. METHODS: From 2011 to 2013, 118 NTD cases and 242 healthy controls were recruited from a population-based birth defects surveillance system in Northern China. Enzyme-linked immunosorbent assay was used to measure FR autoantibodies in maternal and cord blood. Logistic regression models were used to estimate the odds ratios (OR) and 95% confidence intervals (95% CI). RESULTS: Plasma FR autoantibodies levels were significantly elevated in mothers of infants with NTDs compared with mothers of healthy controls. Using the lowest tertile as the referent group, 2.20-fold (95% CI, 0.71-6.80) and 5.53-fold increased odds (95% CI, 1.90-16.08) of NTDs were observed for the second and third tertile of immunoglobulin G (IgG), respectively, and the odds of NTDs for each successive tertile of IgM was 0.98 (95% CI, 0.35-2.75) and 3.49 (95% CI, 1.45-8.39), respectively. A dose-response relationship was found between FR autoantibodies levels and risk of NTDs (P < 0.001 for IgG, P = 0.002 for IgM). The same pattern was observed in both subtypes of spina bifida and anencephaly. No significant difference in levels of cord blood FR autoantibodies was observed. CONCLUSION: Higher levels of FR autoimmunity in maternal plasma are associated with elevated risk of NTDs in a dose-response manner. Birth Defects Research (Part A) 106:685-695, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Anencefalia/inmunología , Autoanticuerpos/sangre , Autoinmunidad/efectos de los fármacos , Receptor 1 de Folato/antagonistas & inhibidores , Ácido Fólico/inmunología , Inmunoglobulina G/sangre , Adulto , Anencefalia/diagnóstico , Anencefalia/genética , Anencefalia/patología , Estudios de Casos y Controles , China , Relación Dosis-Respuesta Inmunológica , Tolerancia a Medicamentos/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Receptor 1 de Folato/genética , Receptor 1 de Folato/inmunología , Ácido Fólico/administración & dosificación , Expresión Génica , Humanos , Lactante , Modelos Logísticos , Masculino , Oportunidad Relativa , Riesgo
17.
Am J Nurs ; 116(3): 60-6, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26914056

RESUMEN

: In the spring of 2012, a nurse in Washington State detected a cluster of babies born with anencephaly-a fatal condition in which infants are born without parts of the brain or skull. The resulting investigation initially confirmed a rate of anencephaly between January 2010 and January 2013 of 8.4 per 10,000 live births-more than four times the national average. As of November 2015, cases of anencephaly in Washington State have continued to increase, with the current rate estimated at 9.5 per 10,000 live births. While no distinct cause has yet been determined, neural tube defects-including anencephaly-are known to have multiple causes, including folic acid deficit, genetic variants in the folate pathway, and exposure to a variety of environmental and occupational toxins. This article describes many of these risk factors and explores the findings of Washington's ongoing investigation.


Asunto(s)
Anencefalia/diagnóstico , Anencefalia/genética , Deficiencia de Ácido Fólico/fisiopatología , Hispánicos o Latinos/estadística & datos numéricos , Población Blanca/estadística & datos numéricos , Anencefalia/epidemiología , Femenino , Humanos , Incidencia , Vigilancia de la Población , Prevalencia , Washingtón/epidemiología
18.
Am J Med Genet A ; 167A(10): 2244-50, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26108864

RESUMEN

Neural tube defects (NTDs) are the most common of the severe malformations of the brain and spinal cord. Increased maternal intake of folic acid (FA) during the periconceptional period is known to reduce NTD risk. Data from 1046 NTD cases in South Carolina were gathered over 20 years of surveillance. It was possible to determine maternal periconceptional FA use in 615 NTD-affected pregnancies. In 163 occurrent (26.9%) and two recurrent (22%) NTD cases, the mothers reported periconceptional FA use. These women were older and more likely to be white. Maternal periconceptional FA usage was reported in 40.4% of cases of spina bifida with other anomalies but in only 25.2% of isolated spina bifida cases (P = 0.02). This enrichment for associated anomalies was not noted among cases of anencephaly or of encephalocele. Among the 563 subsequent pregnancies to mothers with previous NTD-affected pregnancies, those taking FA had a 0.4% NTD recurrence rate, but the recurrence without FA was 8.5%. NTDs with other associated findings were less likely to be prevented by FA, suggesting there is a background NTD rate that cannot be further reduced by FA. Nonetheless, the majority (73.9%) of NTDs in pregnancies in which the mothers reported periconceptional FA use were isolated NTDs of usual types. Cases in which FA failed in prevention of NTDs provide potential areas for further study into the causation of NTDs. The measures and techniques implemented in South Carolina can serve as an effective and successful model for prevention of NTD occurrence and recurrence.


Asunto(s)
Anencefalia/diagnóstico , Suplementos Dietéticos , Encefalocele/diagnóstico , Ácido Fólico/administración & dosificación , Disrafia Espinal/diagnóstico , Adulto , Negro o Afroamericano , Anencefalia/etnología , Anencefalia/genética , Anencefalia/prevención & control , Encefalocele/etnología , Encefalocele/genética , Encefalocele/prevención & control , Femenino , Fertilización , Hispánicos o Latinos , Humanos , Masculino , Vigilancia de la Población , Embarazo , Diagnóstico Prenatal , Recurrencia , Riesgo , South Carolina/epidemiología , Disrafia Espinal/etnología , Disrafia Espinal/genética , Disrafia Espinal/prevención & control , Población Blanca
20.
Mol Cell Biochem ; 407(1-2): 51-6, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26045171

RESUMEN

Congenital malformations, such as neural tube defects (NTDs) and congenital heart disease (CHD), cause significant fetal mortality and childhood morbidity. NTDs are a common congenital anomaly, and are typically induced by higher maternal homocysteine (Hcy) levels and abnormal folate metabolism. The gene encoding methionine synthase reductase (MTRR) is essential for adequate remethylation of Hcy. Previous studies have focused on the coding region of genes involved in one-carbon metabolism, but recent research demonstrates that an allelic change in a non-coding region of MTRR (rs326119) increases the risk of CHD. We hypothesized that this variant might contribute to the etiology of NTDs as well, based on a common role during early embryogenesis. In the present study, 244 neural tube defect cases and 407 controls from northern China were analyzed to determine any association (by χ (2) test) between rs326119 and disease phenotypes. Significant increased risk of anencephaly was seen in MTRR variant rs326119 heterozygote (het) and homozygote (hom) individuals [odds ratios (OR)het = 1.81; ORhom = 2.05)]. Furthermore, this variant was also a risk factor for congenital malformations of the adrenal gland (OR = 1.85), likely due to multiple systemic malformations in the NTDs case population. Our present data indicate that the rs326119 non-coding variant of MTRR has a pleiotropic effect on the development of multiple tissues, especially during early stages in utero. This suggests the allelic state of MTRR is a significant clinical factor affecting Hcy levels and optimal folic supplementation.


Asunto(s)
Anencefalia/genética , Pueblo Asiatico/genética , Ferredoxina-NADP Reductasa/genética , Defectos del Tubo Neural/genética , Polimorfismo de Nucleótido Simple , China , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Cardiopatías Congénitas/genética , Humanos , Intrones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...