Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 92020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32553112

RESUMEN

Primary cilia are sensory organelles crucial for cell signaling during development and organ homeostasis. Cilia arise from centrosomes and their formation and function is governed by numerous factors. Through our studies on Townes-Brocks Syndrome (TBS), a rare disease linked to abnormal cilia formation in human fibroblasts, we uncovered the leucine-zipper protein LUZP1 as an interactor of truncated SALL1, a dominantly-acting protein causing the disease. Using TurboID proximity labeling and pulldowns, we show that LUZP1 associates with factors linked to centrosome and actin filaments. Here, we show that LUZP1 is a cilia regulator. It localizes around the centrioles and to actin cytoskeleton. Loss of LUZP1 reduces F-actin levels, facilitates ciliogenesis and alters Sonic Hedgehog signaling, pointing to a key role in cytoskeleton-cilia interdependency. Truncated SALL1 increases the ubiquitin proteasome-mediated degradation of LUZP1. Together with other factors, alterations in LUZP1 may be contributing to TBS etiology.


Primary cilia are the 'antennae' of animal cells: these small, flexible protrusions emerge from the surface of cells, where they help to sense and relay external signals. Cilia are assembled with the help of the cytoskeleton, a dynamic network of mesh-like filaments that spans the interior of the cell and controls many different biological processes. If cilia do not work properly, human diseases called ciliopathies can emerge. Townes-Brocks Syndrome (TBS) is an incurable disease that presents a range of symptoms such as malformations of the toes or fingers, hearing impairment, and kidney or heart problems. It is caused by a change in the gene that codes for a protein called SALL1, and recent work has also showed that the cells of TBS patients have defective cilia. In addition, this prior research identified a second protein that interacted with the mutant version of SALL1; called LUZP1, this protein is already known to help maintain the cytoskeleton. In this study, Bozal-Basterra et al. wanted to find out if LUZP1 caused the cilia defects in TBS. First, the protein was removed from mouse cells grown in the laboratory, which dramatically weakened the cytoskeleton. In keeping with this observation, both the number of cilia per cell and the length of the cilia were abnormal. Cells lacking LUZP1 also had defects in a signalling process that transmits signals received by cilia to different parts of the cell. All these defects were previously observed in cells isolated from TBS patients. In addition, LUZP1-deficient mouse cells showed the same problems with their cilia and cytoskeleton as the cells from individuals with TBS. Crucially, the cells from human TBS patients also had much lower levels of LUZP1 than normal, suggesting that the protein may contribute to the cilia defects present in this disease. The work by Bozal-Basterra et al. sheds light on how primary cilia depend on the cytoskeleton, while also providing new insight into TBS. In the future, this knowledge could help researchers to develop therapies for this rare and currently untreatable disease.


Asunto(s)
Anomalías Múltiples/etiología , Citoesqueleto de Actina/metabolismo , Ano Imperforado/etiología , Cilios/metabolismo , Proteínas del Citoesqueleto/fisiología , Pérdida Auditiva Sensorineural/etiología , Pulgar/anomalías , Anomalías Múltiples/metabolismo , Adulto , Animales , Ano Imperforado/metabolismo , Centrosoma/metabolismo , Proteínas del Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Humanos , Masculino , Ratones , Factores de Transcripción/metabolismo
2.
Birth Defects Res ; 109(13): 1063-1069, 2017 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-28605140

RESUMEN

BACKGROUND: The acronym VATER/VACTERL refers to the rare nonrandom association of the following component features (CF): vertebral defects (V), anorectal malformations (A), cardiac defects (C), tracheoesophageal fistula with or without esophageal atresia, renal malformations (R), and limb defects (L). Patients presenting with at least three CFs are diagnosed as having VATER/VACTERL association while patients presenting with only two CFs are diagnosed as having VATER/VACTERL-like phenotypes. Recently, rare causative copy number variations (CNVs) have been identified in patients with VATER/VACTERL association and VATER/VACTERL-like phenotypes. METHODS: To detect further causative CNVs we performed array based molecular karyotyping in 75 VATER/VACTERL and 40 VATER/VACTERL-like patients. RESULTS: Following the application of stringent filter criteria, we identified 13 microdeletions and seven microduplications in 20 unrelated patients all of which were absent in 1,307 healthy inhouse controls (n < 0.0008). Among these, microdeletion at 17q12 was confirmed to be de novo. Three microdeletions at 5q23.1, 16q23.3, 22q11.21, and one microduplication at 10q11.21 were all absent in the available parent. Microdeletion of chromosomal region 22q11.21 was previously found in VATER/VACTERL patients rendering it to be causative in our patient. The remaining 15 CNVs were inherited from a healthy parent. CONCLUSION: In two of 115 patients' causative CNVs were found (2%). The remaining identified rare CNVs represent candidates for further evaluation. Rare inherited CNVs may constitute modifiers of, or contributors to, multifactorial VATER/VACTERL or VATER/VACTERL-like phenotypes. Birth Defects Research 109:1063-1069, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Ano Imperforado/genética , Esófago/anomalías , Cardiopatías Congénitas/genética , Radio (Anatomía)/anomalías , Columna Vertebral/anomalías , Tráquea/anomalías , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Adulto , Canal Anal/anomalías , Animales , Malformaciones Anorrectales/genética , Ano Imperforado/complicaciones , Ano Imperforado/metabolismo , Variaciones en el Número de Copia de ADN , Esófago/metabolismo , Femenino , Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/metabolismo , Humanos , Cariotipo , Cariotipificación , Masculino , Fenotipo , Radio (Anatomía)/metabolismo , Columna Vertebral/metabolismo , Tráquea/metabolismo
3.
Toxicology ; 338: 77-85, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26514922

RESUMEN

Previous study have demonstrated that not only the anorectal development but also the general conditions of anorectal malformations (ARMs) male rats are severely affected by di-n-butyl phthalate (DBP) maternal exposure. However, the mechanisms underlying DBP-induced congenital defects remain elusive. Reportedly, Fgf10/Fgfr2 and androgen receptor (AR) are pivotal for the development of multiple organs. In this study, we therefore investigated the expression of Fgf10/Fgfr2 together with AR in the terminal rectum and multiple organs of ARM male rats induced by in utero exposure to DBP. DBP was administered to pregnant rats to establish the model and the incidence of ARMs in male offspring was 39.5%. On postnatal day(PND)1, the gross photograph and histopathological staining confirmed the abnormal manifestations in these organs of newborn ARMs. Decreased anogenital distance, body weight and serum testosterone level were observed in ARM male offspring. The reduced expression of Fgf10/Fgfr2 mRNA and protein was seen in terminal rectum and kidney, spleen, liver, heart in ARM male rats, whereas the reduced expression of AR was only observed in the kidney and terminal rectum. Our findings suggest the potential involvement of altered Fgf10/Fgfr2 signaling and AR in pathogenesis of local and systemic development defects in ARMs male rats induce by DBP.


Asunto(s)
Canal Anal/anomalías , Ano Imperforado/inducido químicamente , Dibutil Ftalato/toxicidad , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores Androgénicos/metabolismo , Recto/anomalías , Recto/efectos de los fármacos , Canal Anal/metabolismo , Animales , Animales Recién Nacidos , Malformaciones Anorrectales , Ano Imperforado/genética , Ano Imperforado/metabolismo , Peso Corporal , Femenino , Factor 10 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Exposición Materna/efectos adversos , Embarazo , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptores Androgénicos/genética , Recto/metabolismo , Transducción de Señal/efectos de los fármacos , Testosterona/sangre
4.
Am J Physiol Renal Physiol ; 309(10): F852-63, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26311113

RESUMEN

It has been postulated that developmental pathways are reutilized during repair and regeneration after injury, but functional analysis of many genes required for kidney formation has not been performed in the adult organ. Mutations in SALL1 cause Townes-Brocks syndrome (TBS) and nonsyndromic congenital anomalies of the kidney and urinary tract, both of which lead to childhood kidney failure. Sall1 is a transcriptional regulator that is expressed in renal progenitor cells and developing nephrons in the embryo. However, its role in the adult kidney has not been investigated. Using a mouse model of TBS (Sall1TBS), we investigated the role of Sall1 in response to acute kidney injury. Our studies revealed that Sall1 is expressed in terminally differentiated renal epithelia, including the S3 segment of the proximal tubule, in the mature kidney. Sall1TBS mice exhibited significant protection from ischemia-reperfusion injury and aristolochic acid-induced nephrotoxicity. This protection from acute injury is seen despite the presence of slowly progressive chronic kidney disease in Sall1TBS mice. Mice containing null alleles of Sall1 are not protected from acute kidney injury, indicating that expression of a truncated mutant protein from the Sall1TBS allele, while causative of congenital anomalies, protects the adult kidney from injury. Our studies further revealed that basal levels of the preconditioning factor heme oxygenase-1 are elevated in Sall1TBS kidneys, suggesting a mechanism for the relative resistance to injury in this model. Together, these studies establish a functional role for Sall1 in the response of the adult kidney to acute injury.


Asunto(s)
Anomalías Múltiples/metabolismo , Lesión Renal Aguda/metabolismo , Ano Imperforado/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Proteínas Mutantes/metabolismo , Pulgar/anomalías , Factores de Transcripción/metabolismo , Anomalías Múltiples/genética , Lesión Renal Aguda/genética , Animales , Ano Imperforado/genética , Modelos Animales de Enfermedad , Pérdida Auditiva Sensorineural/genética , Hemo-Oxigenasa 1/genética , Ratones Transgénicos , Mutación/genética , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Factores de Transcripción/genética
5.
Int J Clin Exp Pathol ; 8(4): 3727-34, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26097554

RESUMEN

The aim of this study was to determine the expression patterns of bone morphogenetic protein 7 (BMP7) during anorectal development in normal rat embryos and in embryos with anorectal malformations (ARM), and to investigate the possible role of BMP7 in the pathogenesis of ARM. ARM was induced by treating rat embryos with ethylenethiourea on the 10th gestational day (GD10). Embryos were harvested by Cesarean delivery and the spatiotemporal expression of BMP7 was evaluated in normal (n=168) and ARM embryos (n=171) from GD13 to GD16 using immunohistochemistry staining and western blot analysis. Immunohistochemical staining in normal embryos revealed that BMP7 was abundantly expressed on the epithelium of the urorectal septum (URS) and the hindgut on GD13, and BMP7-immunopositive cells were extensively detected in the URS, hindgut, and cloacal membrane by GD14. Increased positive tissue staining was noted on the fused tissue of the URS and the thin anal membrane on GD15. In ARM embryos, the epithelium of the cloaca, URS, and anorectum were negatively or only faintly immunostained for BMP7. BMP7 protein expression showed time-dependent changes in the developing hindgut according to western blotting, and reached a peak on GD15 during anus formation. BMP7 expression levels from GD14 to GD15 were significantly lower in the ARM group compared with the normal group (P<0.05). Spatiotemporal expression of BMP7 was disrupted in ARM embryos during anorectal morphogenesis from GD13 to GD16. These results suggest that downregulation of BMP7 at the time of cloacal separation into the primitive rectum and UGS might be related to the development of ARM.


Asunto(s)
Canal Anal/anomalías , Ano Imperforado/metabolismo , Proteína Morfogenética Ósea 7/metabolismo , Recto/anomalías , Canal Anal/metabolismo , Animales , Malformaciones Anorrectales , Ano Imperforado/inducido químicamente , Regulación hacia Abajo , Epitelio/metabolismo , Etilenotiourea , Femenino , Embarazo , Ratas , Ratas Wistar , Recto/metabolismo
6.
Cells Tissues Organs ; 199(2-3): 212-20, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25401498

RESUMEN

PURPOSE: The aim of this study was to determine the expression of Cdx4 (caudal-type homeobox gene-4) during anorectal development in normal and ethylenethiourea (ETU)-induced anorectal malformation (ARM) embryos with a view to establishing the possible role of Cdx4 in ARM pathogenesis. MATERIALS AND METHODS: ARM was induced by ETU on the 10th gestational day (GD10) in rat embryos. Cesarean deliveries were then performed to harvest the embryos. Spatiotemporal expression of Cdx4 was evaluated in normal rat embryos (n = 354) and ARM embryos (n = 378) from GD13 to GD16. RESULTS: Immunohistochemical staining and immunofluorescence revealed that, in normal embryos, Cdx4 expression was extensively detected on the epithelium of the cloaca on GD13. On GD14, the Cdx4-positive cells were intensively detected on the hindgut. On GD15, the anal membrane was constantly immunoreactive to Cdx4. On GD16, Cdx4-labeled cells were observed on the epithelium of the anus. In the ARM embryos, the epithelium of the cloaca, urorectal septum (URS) and anorectum was negative or faint for Cdx4. In the normal embryo group, Cdx4 protein and mRNA expression showed time-dependent changes in the developing hindgut from GD13 to GD16 on Western blot and real-time reverse transcription plus polymerase chain reaction. Once the URS divided the cloaca into the primitive rectum and urogenital sinus (UGS) on GD15, Cdx4 expression began to decrease. In addition, the expression level of Cdx4 in the ARM group from GD13 to GD15 was significantly lower than that in the normal group (p < 0.05). CONCLUSIONS: In ARM embryos, an imbalance in the spatiotemporal expression of Cdx4 was noted during anorectal morphogenesis from GD13 to GD16. This suggests that ETU may cause downregulation of Cdx4 expression. Downregulation of Cdx4 at the time of cloacal separation into the primitive rectum and UGS might thus be related to the development of ARM.


Asunto(s)
Canal Anal/metabolismo , Ano Imperforado/embriología , Ano Imperforado/metabolismo , Etilenotiourea/toxicidad , Proteínas de Homeodominio/metabolismo , Recto/metabolismo , Canal Anal/anomalías , Canal Anal/efectos de los fármacos , Animales , Malformaciones Anorrectales , Ano Imperforado/inducido químicamente , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Masculino , Morfogénesis/genética , Morfogénesis/fisiología , Embarazo , Ratas , Recto/anomalías , Recto/efectos de los fármacos
7.
Int J Clin Exp Pathol ; 7(5): 1997-2005, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24966909

RESUMEN

UNLABELLED: Fecal incontinence and constipation after procedures for anorectal malformations (ARMs) are closely related to the maldevelopment of the striated muscle complex (SMC). Previous studies have demonstrated that myogenic regulatory factors (MRFs) play a significant role in muscle development. Wnt signal pathway is extremely important for MRFs regulation. This study was designed to investigate the spatiotemporal expression pattern of Wnt5a in SMC in ARMs rat embryos. MATERIALS AND METHODS: Anorectal malformation embryos were induced by ethylene thiourea on embryonic day 10 (E10). Expression levels of protein and mRNA of Wnt5a were confirmed by immunohistochemistry staining, western blot and quantitative real-time PCR (qRT-PCR) between normal rat embryos and embryos with ARMs. RESULTS: Immunostaining revealed that, on embryonic day 17 (E17), the Wnt5a protein was initially expressed in the SMC in normal embryos. With the growth of pregnancy, the positive staining cells gradually increased. The same time-dependent changes of Wnt5a protein were detected in ARMs embryos. Besides, immunostaining showed that Wnt5a had a significant increase in normal embryos compared with ARMs embryos. Similarly, in Western blot and qRT-PCR, the higher expression of Wnt5a protein and mRNA were remarkable in normal embryos during the SMC development, relatively. CONCLUSION: Our study demonstrated that the downregulation of Wnt5a at the time of SMC development might partly be related to the dysplasia of SMC in ARMs.


Asunto(s)
Ano Imperforado/metabolismo , Desarrollo de Músculos , Músculo Estriado/metabolismo , Proteínas Wnt/metabolismo , Animales , Malformaciones Anorrectales , Ano Imperforado/inducido químicamente , Western Blotting , Modelos Animales de Enfermedad , Etilenotiourea , Femenino , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Inmunohistoquímica , Masculino , Exposición Materna , Músculo Estriado/anomalías , Embarazo , ARN Mensajero/metabolismo , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Proteínas Wnt/genética , Proteína Wnt-5a
8.
Cell Tissue Res ; 357(1): 83-90, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24744267

RESUMEN

The main aim of this study was to determine Cdx2 expression patterns during anorectal development in normal and anorectal malformation (ARM) embryos with a view to establishing the possible role of Cdx2 in ARM pathogenesis. ARM was induced with ethylenethiourea on the 10th gestational day (GD10) in rat embryos, and Cesarean deliveries were performed to harvest the embryos. The temporal and spatial expression of Cdx2 was evaluated in normal rat embryos (n = 303) and ARM embryos (n = 321) from GD13 to GD16. Immunohistochemical staining revealed that, in normal embryos, Cdx2 was mainly expressed on the epithelium of the urorectal septum (URS) and the hindgut on GD13. On GD14, Cdx2-immunopositive cells were extensively detected on the URS, hindgut, and cloacal membrane. On GD15, increased immunopositive tissue staining on the anal membrane was evident. In ARM embryos, the epithelium of the cloaca, URS, and anorectum were negative or faintly immunostaining for Cdx2. Analyses by Western blot and real-time reverse transcription plus the polymerase chain reaction revealed that, in the normal group, Cdx2 protein and mRNA expression showed time-dependent changes in the developing hindgut from GD13 to GD16. Upon the URS division of the cloaca into the primitive rectum and urogenital sinus (UGS) on GD15, Cdx2 expression began to decrease. Moreover, the Cdx2 expression level in the ARM group from GD13 to GD14 was significantly lower than that in the normal group (P < 0.05). In ARM embryos, an imbalance in the spatiotemporal expression of Cdx2 was noted during anorectal morphogenesis from GD13 to GD16. Downregulation of Cdx2 at the time of cloacal separation into the primitive rectum and UGS might thus be related to the development of ARM.


Asunto(s)
Canal Anal/anomalías , Ano Imperforado/metabolismo , Proteínas de Homeodominio/biosíntesis , Factores de Transcripción/biosíntesis , Canal Anal/efectos de los fármacos , Canal Anal/embriología , Animales , Malformaciones Anorrectales , Ano Imperforado/inducido químicamente , Ano Imperforado/genética , Factor de Transcripción CDX2 , Modelos Animales de Enfermedad , Etilenotiourea , Femenino , Proteínas de Homeodominio/genética , Masculino , Morfogénesis/genética , Embarazo , Ratas , Ratas Wistar , Factores de Transcripción/genética
9.
Birth Defects Res A Clin Mol Teratol ; 100(9): 647-57, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24740457

RESUMEN

BACKGROUND: In a recent study, high maternal periconceptional intake of vitamin E was found to be associated with risk of congenital heart defects (CHDs). To explore this association further, we investigated the association between total daily vitamin E intake and selected birth defects. METHODS: We analyzed data from 4525 controls and 8665 cases from the 1997 to 2005 National Birth Defects Prevention Study. We categorized estimated periconceptional energy-adjusted total daily vitamin E intake from diet and supplements into quartiles (referent, lowest quartile). Associations between quartiles of energy-adjusted vitamin E intake and selected birth defects were adjusted for demographic, lifestyle, and nutritional factors. RESULTS: We observed a statistically significant association with the third quartile of vitamin E intake (odds ratio [OR], 1.17; 95% confidence interval [CI], 1.01-1.35) and all CHDs combined. Among CHD sub-types, we observed associations with left ventricular outflow tract obstruction defects, and its sub-type, coarctation of the aorta and the third quartile of vitamin E intake. Among defects other than CHDs, we observed associations between anorectal atresia and the third quartile of vitamin E intake (OR, 1.66; 95% CI, 1.01-2.72) and hypospadias and the fourth quartile of vitamin E intake (OR, 1.42; 95% CI, 1.09-1.87). CONCLUSION: Selected quartiles of energy-adjusted estimated total daily vitamin E intake were associated with selected birth defects. However, because these few associations did not exhibit exposure-response patterns consistent with increasing risk associated with increasing intake of vitamin E, further studies are warranted to corroborate our findings.


Asunto(s)
Ano Imperforado/epidemiología , Suplementos Dietéticos , Cardiopatías Congénitas/epidemiología , Hipospadias/epidemiología , Efectos Tardíos de la Exposición Prenatal/epidemiología , Vitamina E/efectos adversos , Adulto , Malformaciones Anorrectales , Ano Imperforado/etiología , Ano Imperforado/metabolismo , Ano Imperforado/patología , Estudios de Casos y Controles , Metabolismo Energético , Femenino , Encuestas Epidemiológicas , Cardiopatías Congénitas/etiología , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología , Humanos , Hipospadias/etiología , Hipospadias/metabolismo , Hipospadias/patología , Recién Nacido , Estilo de Vida , Masculino , Oportunidad Relativa , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología , Factores de Riesgo , Estados Unidos/epidemiología
10.
Mamm Genome ; 24(11-12): 500-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24213835

RESUMEN

Anal atresia (i.e., anorectal malformations) is a severe disorder that occurs during the development of the distal hindgut in infants, swine, and many other mammals and has an unclear genetic background. Recently, the Shh-responsive transcription factor GLI2 has been shown as essential to the normal development of the hindgut, and QTL studies in pigs revealed that this gene may be an important candidate for anal atresia (AA). We used the pig as the model to study the contribution of GLI2 to AA. We revealed the genomic structure of the porcine GLI2 gene with 14 exons and obtained the porcine GLI2 mRNA sequence with a 4,656-bp ORF coding a 1,551-amino acid protein. We further scanned the genome-wide mutations in this gene by direct sequencing using three genomic DNA pools from the AA pigs, full-sibs of AA pigs, and unaffected pigs, respectively. Finally, 30 single nucleotide polymorphisms (SNPs) and one intronic 9-nucleotide (nt) deletion were identified. Of these SNPs, 23 are intronic, 6 are synonymous, and 1 (446 G>A) in exon 8 is nonsynonymous (365Met >Ile). NCOI-RFLP of the 446 G>A polymorphism suggested that the predominant genotypes were all GG and AG in the three pig groups. In addition, there was no significant difference among the three groups in allele frequencies, which demonstrated that this locus was not associated with AA in pigs. However, the 12 SNPs encompassing exon 4 to exon 8 showed strong linkage disequilibrium in the AA pigs, which indicated that the mutations somewhere in this region may contribute to AA in pigs. Therefore, further investigation in this region is needed to elucidate the underlying mutations involved in the porcine AA.


Asunto(s)
Ano Imperforado/genética , Estudio de Asociación del Genoma Completo , Factores de Transcripción de Tipo Kruppel/genética , Mutación , Porcinos/genética , Animales , Malformaciones Anorrectales , Ano Imperforado/metabolismo , Clonación Molecular , Modelos Animales de Enfermedad , Exones , Humanos , Desequilibrio de Ligamiento , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Proteína Gli2 con Dedos de Zinc
11.
Pediatr Surg Int ; 29(11): 1209-16, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23975021

RESUMEN

BACKGROUND: Adriamycin mouse model (AMM) is a model of VACTERL anomalies. Sonic hedgehog (Shh) pathway, sourced by the notochord, is implicated of anorectal malformations. We hypothesized hindgut anomalies observed in the AMM are the result of abnormal effect of the notochord. METHODS: Time-mated CBA/Ca mice received two intraperitoneal injections of Adriamycin (6 mg/kg) or saline as control on embryonic day (E) 7 and 8. Fetuses were harvested from E9 to E11, stained following whole mount in situ hybridization with labeled RNA probes to detect Shh and Fork head box F1(Foxf1) transcripts. Immunolocalization with endoderm marker Hnf3ß was used to visualize morphology. Embryos were scanned by OPT to obtain 3D representations of expressions. RESULTS: In AMM, the notochord was abnormally displaced ventrally with attachment to the hindgut endoderm in 71 % of the specimens. In 32 % of the treated embryos abnormal hindgut ended blindly in a cystic structure, and both of types were remarked in 29 % of treated embryos. Endodermal Shh and mesenchymal Foxf1 genes expression were preserved around the hindgut cystic malformation. CONCLUSIONS: The delamination of the developing notochord in the AMM is disrupted, which may influence signaling mechanisms from the notochord to the hindgut resulting in abnormal patterning of the hindgut.


Asunto(s)
Canal Anal/anomalías , Ano Imperforado/genética , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Notocorda/anomalías , Preñez , ARN/genética , Recto/anomalías , Canal Anal/embriología , Canal Anal/metabolismo , Animales , Malformaciones Anorrectales , Ano Imperforado/embriología , Ano Imperforado/metabolismo , Modelos Animales de Enfermedad , Doxorrubicina/toxicidad , Femenino , Factores de Transcripción Forkhead/biosíntesis , Proteínas Hedgehog/biosíntesis , Imagenología Tridimensional , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos CBA , Notocorda/embriología , Notocorda/metabolismo , Embarazo , Recto/embriología , Recto/metabolismo , Tomografía de Coherencia Óptica
12.
Int J Mol Med ; 30(6): 1459-64, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22961180

RESUMEN

Anorectal malformations (ARMs) comprise a broad spectrum of anomalies, including anal atresia, congenital anal fistula and persistence of the cloaca. Research suggests that genetic factors play an important role in ARM development. However, few genetic variants have been identified. Embryogenesis is orchestrated by crosstalk of the wingless-type MMTV integration site family (WNT) and fibroblast growth factor (FGF) signaling pathways in a process that involves several intracellular cascades. Studies in mice have implicated several genes from these pathways in the etiology of ARMs. We performed sequencing analysis of seven of these previously reported genes in 78 patients with ARMs occurring within the context of at least one additional congenital anomaly. No associations were identified with variants in WNT3A, WNT5A, WNT11, DACT1, FGF10 or the T gene. In the FGFR2 gene, three novel heterozygous nucleotide substitutions were identified. Further investigations, including the study of family members, revealed that these variants were not causally related to the phenotype in the present ARM cohort. Mutations in the seven investigated genes may nonetheless be a cause of ARMs in rare cases. However, further studies should consider genes encoding other proteins in the WNT/FGF signaling pathways as possible candidates.


Asunto(s)
Ano Imperforado/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Vía de Señalización Wnt , Proteínas Adaptadoras Transductoras de Señales/genética , Malformaciones Anorrectales , Ano Imperforado/metabolismo , Análisis Mutacional de ADN , Proteínas Fetales/genética , Factor 10 de Crecimiento de Fibroblastos/genética , Estudios de Asociación Genética , Humanos , Proteínas Nucleares/genética , Mutación Puntual , Proteínas Proto-Oncogénicas/genética , Proteínas de Dominio T Box/genética , Proteínas Wnt/genética , Proteína Wnt-5a , Proteína Wnt3A/genética
13.
J Surg Res ; 172(1): 131-6, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20888582

RESUMEN

BACKGROUND: This study was designed to investigate the expression of Notch-1 and Jagged-2 in the terminal hindgut in ethylenethiourea (ETU)-exposed rat embryos with anorectal malformations (ARMs) and its potential association with the maldevelopment of the terminal hindgut in ARMs. MATERIAL AND METHODS: ETU-exposed ARMs model was introduced to investigate the expression pattern of Notch-1 and Jagged-2 during the hindgut development using immunohistochemical staining, reverse transcriptase polymerase chain reaction (RT-PCR) and Western blot analysis. RESULTS: Immunostaining revealed that the expression of Notch-1 and Jagged-2 showed changes in the developing terminal hindgut of ARMs. The expression of Notch-1 and Jagged-2 in the terminal hindgut of ARMs rat embryos decreased at both mRNA level and protein level (P < 0.05) compared with normal tissues. CONCLUSION: This study demonstrated that the expression of Notch-1 and Jagged-2 in ARMs of ETU-exposed rat embryos was remarkably reduced, which implied its potential role in the pathogenesis of the terminal hindgut maldevelopment in ARMs.


Asunto(s)
Ano Imperforado/inducido químicamente , Ano Imperforado/metabolismo , Etilenotiourea/efectos adversos , Tracto Gastrointestinal/embriología , Tracto Gastrointestinal/metabolismo , Proteínas de la Membrana/metabolismo , Receptor Notch1/metabolismo , Animales , Malformaciones Anorrectales , Regulación del Desarrollo de la Expresión Génica , Proteína Jagged-2 , Masculino , Modelos Animales , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
14.
Eur J Pediatr Surg ; 21(4): 238-41, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21480163

RESUMEN

BACKGROUND/PURPOSE: Retinoid-mediated signal transduction plays a crucial role in the embryonic development of various organs. We previously reported that retinoic acid induced anorectal malformations (ARM) in mice. GDF11 is a TGFß superfamily molecule and is cleaved and activated by proprotein convertase subtilisin/kexin 5 (PCSK5). PCSK5 (PC5/6) mutations result in an abnormal expression of Hlxb9 and Hox genes, which include known GDF11 targets that are necessary for caudal development in vertebrate embryos. To determine a possible role of the retinoid-mediated signaling pathway in the pathogenesis of ARM, we investigated whether all-trans retinoic acid (ATRA) affected the expression patterns of PCSK5 and GDF11 in ARM-treated mouse embryos. METHODS: Pregnant ICR-Slc mice were administered 100 mg/kg ATRA by gavage on embryonic day (E) 9.0. Embryos were harvested between days E12 and E18, and mid-sagittal sections of the hindgut region were prepared for immunohistochemistry using antibodies against PCSK5 (PC5/6) and GDF11 (GDF8/11). RESULTS: Over 95% of the embryos treated with ATRA showed ARM, with rectourethral fistula or rectocloacal fistula, and a short tail. Furthermore, most of these embryos exhibited sacral malformations, tethered spinal cords, and presacral masses resembling those malformations found in caudal regression syndrome. By E14, normal mouse embryos formed a rectum and anus, and the somites behind the hindgut were positive for PC5/6 and GDF8/11. In contrast, in ARM embryos, the somites behind the hindgut were negative for PC5/6 and GDF8/11. CONCLUSION: ATRA treatment affected the caudal development in mouse embryos, resulting in anorectal, sacral, and spinal malformations, and inhibited PCSK5 and GDF11 expression in the hindgut region. These findings indicate that the expression of PCSK5 and GDF11, which plays a crucial role in the organogenesis of the hindgut, was disturbed in the hindgut region when retinoid-mediated signaling was disrupted. This study offers a new insight into the pathogenesis of ARM in mice as affected by the interaction between ATRA and PCSK5/GDF11.


Asunto(s)
Ano Imperforado/embriología , Proteínas Morfogenéticas Óseas/metabolismo , Colon/embriología , Factores de Diferenciación de Crecimiento/metabolismo , Proproteína Convertasa 5/metabolismo , Tretinoina/efectos adversos , Anomalías Múltiples/embriología , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Animales , Malformaciones Anorrectales , Ano Imperforado/metabolismo , Ano Imperforado/patología , Colon/anomalías , Colon/metabolismo , Femenino , Inmunohistoquímica , Ratones , Ratones Endogámicos ICR , Embarazo , Fístula Rectal/embriología , Fístula Rectal/metabolismo , Transducción de Señal , Cola (estructura animal)/anomalías , Cola (estructura animal)/embriología , Tretinoina/administración & dosificación , Tretinoina/metabolismo
15.
J Biochem ; 147(3): 445-50, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20053786

RESUMEN

Human SALL1 is a homologue of the Drosophila region-specific homeotic gene sal, and is also known as a causative gene for Townes-Brocks syndrome, which is characterized by multi-organ malformations. We previously demonstrated that mouse Sall1 plays a crucial role in ureteric bud invasion during kidney development, and possibly in nephron progenitor cells in the metanephric mesenchyme. To gain insights into the Sall1 functions in the kidney and other tissues, we generated R26Sall1 mice, in which Rosa26 locus stop sequences flanked by two loxP sites were located upstream of the Sall1 cDNA. This allele allowed exogenous Sall1 expression in a Cre recombinase-dependent manner. R26Sall1 mice were first crossed with CAGCre mice, which expressed Cre recombinase ubiquitously during embryogenesis. Mice expressing Sall1 ubiquitously were smaller in size compared with mice of other genotypes. We then crossed R26Sall1 mice with Six2Cre mice expressing Cre recombinase in the metanephric mesenchyme during kidney development. However, no kidney defects were observed. Taken together, overexpression of Sall1 does not affect kidney development, but does lead to a reduced body weight, suggesting that the optimal dosage of Sall1 is required for normal mouse development.


Asunto(s)
Peso Corporal , Riñón/embriología , Factores de Transcripción , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Animales , Ano Imperforado/genética , Ano Imperforado/metabolismo , Oído/anomalías , Regulación del Desarrollo de la Expresión Génica , Humanos , Riñón/citología , Riñón/metabolismo , Deformidades Congénitas de las Extremidades/genética , Deformidades Congénitas de las Extremidades/metabolismo , Ratones , Ratones Transgénicos , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...