Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
1.
Development ; 151(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38587174

ABSTRACT

The gastrointestinal (GI) tract is complex and consists of multiple organs with unique functions. Rare gene variants can cause congenital malformations of the human GI tract, although the molecular basis of these has been poorly studied. We identified a patient with compound-heterozygous variants in RFX6 presenting with duodenal malrotation and atresia, implicating RFX6 in development of the proximal intestine. To identify how mutations in RFX6 impact intestinal patterning and function, we derived induced pluripotent stem cells from this patient to generate human intestinal organoids (HIOs). We identified that the duodenal HIOs and human tissues had mixed regional identity, with gastric and ileal features. CRISPR-mediated correction of RFX6 restored duodenal identity. We then used gain- and loss-of-function and transcriptomic approaches in HIOs and Xenopus embryos to identify that PDX1 is a downstream transcriptional target of RFX6 required for duodenal development. However, RFX6 had additional PDX1-independent transcriptional targets involving multiple components of signaling pathways that are required for establishing early regional identity in the GI tract. In summary, we have identified RFX6 as a key regulator in intestinal patterning that acts by regulating transcriptional and signaling pathways.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins , Organoids , Regulatory Factor X Transcription Factors , Trans-Activators , Humans , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/metabolism , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Organoids/metabolism , Organoids/embryology , Duodenum/metabolism , Duodenum/embryology , Intestines/embryology , Intestinal Atresia/genetics , Induced Pluripotent Stem Cells/metabolism , Body Patterning/genetics , Signal Transduction/genetics , Mutation/genetics
2.
Genes (Basel) ; 14(11)2023 Oct 24.
Article in English | MEDLINE | ID: mdl-38002928

ABSTRACT

Strømme syndrome is an ultra-rare primary ciliopathy with clinical variability. The syndrome is caused by bi-allelic variants in CENPF, a protein with key roles in both chromosomal segregation and ciliogenesis. We report three unrelated patients with Strømme syndrome and, using high-throughput sequencing approaches, we identified novel pathogenic variants in CENPF, including one structural variant, giving a genetic diagnosis to the patients. Patient 1 was a premature baby who died at 26 days with congenital malformations affecting many organs including the brain, eyes, and intestine. She was homozygous for a donor splice variant in CENPF, NM_016343.3:c.1068+1G>A, causing skipping of exon 7, resulting in a frameshift. Patient 2 was a female with intestinal atresia, microcephaly, and a Peters anomaly. She had normal developmental milestones at the age of 7 years. She is compound heterozygous for CENPF NM_016343.3:c.5920dup and c.8991del, both frameshift. Patient 3 was a male with anomalies of the brain, eye, intestine, and kidneys. He was compound heterozygous for CENPF p.(Glu298Ter), and a 5323 bp deletion covering exon 1. CENPF exon 1 is flanked by repetitive sequences that may represent a site of a recurrent structural variation, which should be a focus in patients with Strømme syndrome of unknown etiology.


Subject(s)
Intestinal Atresia , Microcephaly , Child , Female , Humans , Infant , Male , Anterior Eye Segment , Intestinal Atresia/genetics , Microcephaly/genetics , Mutation
3.
Anim Genet ; 54(2): 104-112, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36639915

ABSTRACT

Intestinal atresia is an under-diagnosed congenital defect in cattle. It results in complete occlusion of the intestinal lumen and, unless surgically corrected, results in death or euthanasia of the affected calf. There is limited information on the incidence of this condition or on risk factors, including predisposing alleles, associated with the defect. In this study, active surveillance of 39 dairy farms over 8 years identified 197 cases of intestinal atresia among 56 454 calves born, an incidence of 0.35%. The majority of cases (83%) had occlusion of the jejunum, although cases with blockage of the colon (14%) or anus (4%) were also identified. The defect was twice as common in male as in female calves (p < 0.0001), and was more common in progeny of older cows than in progeny of first or second lactation cows (p < 0.001). Year and farm of birth were also significantly associated with incidence (p < 0.05). The incidence of intestinal atresia was highest among the progeny of three related Jersey sires, suggesting that a gene for intestinal atresia was segregating within this family. Linkage analysis utilising 28 affected progeny of two half-sib putative carrier sires identified two putative quantitative trait loci associated with the defect, on chromosomes 14 and 26, although no clear candidate genes were identified. There was no evidence of a sire-effect among the progeny of Holstein-Friesian sires. However, a case-control genome-wide association study involving 91 cases and 375 healthy controls, identified 31 SNP in 18 loci as associated with the defect in this breed. These data suggest that intestinal atresia in dairy calves is not a simple Mendelian trait as previously reported but a complex multigenic disorder.


Subject(s)
Intestinal Atresia , Pregnancy , Animals , Cattle/genetics , Female , Male , Intestinal Atresia/genetics , Intestinal Atresia/veterinary , Genome-Wide Association Study , Parturition , Risk Factors , Lactation
4.
Genes (Basel) ; 13(5)2022 05 04.
Article in English | MEDLINE | ID: mdl-35627206

ABSTRACT

Multiple intestinal atresia with combined immune deficiency (MIA-CID) is an autosomal recessive syndrome due to mutations in the TTC7A gene implicated in the polarization of intestinal and thymic epithelial cells. MIA-CID is lethal in the first year of life in the majority of patients. Dermatological manifestations have been reported in a few cases. We describe a child affected with MIA-CID due to a previously unreported TTC7A homozygous missense mutation. Surgery for bowel occlusion was performed in the first days of life. The patient was totally dependent on parenteral nutrition since birth and presented severe diarrhea and recurrent infections. He underwent hematopoietic stem cell transplantation at 17 months with complete donor engraftment and partial immunity improvement. In the second year of life, he progressively developed diffuse papular follicular keratoses on ichthyosiform skin, nail clubbing, and subungual hyperkeratosis. Histopathology showed hyperkeratosis with follicular plugging and scattered apoptotic keratinocytes, visualized at an ultrastructural examination. Our findings expand the spectrum of dermatological manifestations which can develop in MIA-CID patients. Examination of further patients will allow defining whether keratinocyte apoptosis is also a disease feature.


Subject(s)
Darier Disease , Intestinal Atresia , Primary Immunodeficiency Diseases , Abnormalities, Multiple , Child , Eyebrows/abnormalities , Humans , Intestinal Atresia/genetics , Intestinal Atresia/pathology , Male , Mutation , Proteins/genetics
5.
Am J Med Genet C Semin Med Genet ; 190(1): 102-108, 2022 03.
Article in English | MEDLINE | ID: mdl-35488810

ABSTRACT

Biallelic loss-of-function (LoF) variants in CENPF gene are responsible for Strømme syndrome, a condition presenting with intestinal atresia, anterior ocular chamber anomalies, and microcephaly. Through an international collaboration, four individuals (three males and one female) carrying CENPF biallelic variants, including two missense variants in homozygous state and four LoF variants, were identified by exome sequencing. All individuals had variable degree of developmental delay/intellectual disability and microcephaly (ranging from -2.9 SDS to -5.6 SDS) and a recognizable pattern of dysmorphic facial features including inverted-V shaped interrupted eyebrows, epicanthal fold, depressed nasal bridge, and pointed chin. Although one of the cases had duodenal atresia, all four individuals did not have the combination of internal organ malformations of Strømme syndrome (intestinal atresia and anterior eye segment abnormalities). Immunofluorescence analysis on skin fibroblasts on one of the four cases with the antibody for ARL13B that decorates primary cilia revealed shorter primary cilia that are consistent with a ciliary defect. This case-series of individuals with biallelic CENPF variants suggests the spectrum of clinical manifestations of the disorder that may be related to CENPF variants is broad and can include phenotypes lacking the cardinal features of Strømme syndrome.


Subject(s)
Chromosomal Proteins, Non-Histone , Intellectual Disability , Intestinal Atresia , Microcephaly , Microfilament Proteins , Chromosomal Proteins, Non-Histone/genetics , Eye Abnormalities , Female , Humans , Intestinal Atresia/genetics , Male , Microcephaly/genetics , Microfilament Proteins/genetics , Mutation/genetics , Phenotype
6.
Pediatr Transplant ; 26(5): e14270, 2022 08.
Article in English | MEDLINE | ID: mdl-35307919

ABSTRACT

BACKGROUND: MRS/MFS is a rare multisystem disorder with a poor prognosis. The high mortality rate of this syndrome is related to the severity of the associated gastrointestinal, pancreatic, and hepatobiliary conditions, as most of them are not amenable to conventional medical and surgical treatments. METHODS: We report the case of a Romani girl with all the key clinical features of MRS/MFS, and a review of cases reported in the literature. Our patient is a newborn from consanguineous parents who presented duodenal atresia, hypoplastic pancreas, gallbladder agenesis, and neonatal diabetes. Given the clinical suspicion of MRS/MFS, a genetic analysis was performed which revealed the presence of a homozygous variant in the RFX6 gene. During the course of the disease, the patient presented intractable secretory diarrhea and severe intestinal failure. RESULTS: At 2 years of age, she underwent MVT of the stomach, duodenum, small intestine, colon, liver, and pancreas. There were no surgical complications. Histologic evaluation of the small bowel showed extensive patches of gastric heterotopia. After more than 10 years of follow-up, she had presented with normal gastrointestinal, hepatic, and pancreatic function. She has one of the longest survival periods in the literature. CONCLUSIONS: Our experience suggests that multivisceral transplantation may be a promising option in select cases of MRS/MFS.


Subject(s)
Diabetes Mellitus , Gallbladder Diseases , Intestinal Atresia , Diabetes Mellitus/genetics , Female , Gallbladder Diseases/genetics , Gallbladder Diseases/pathology , Humans , Infant, Newborn , Intestinal Atresia/genetics , Intestinal Atresia/pathology , Intestinal Atresia/surgery , Tracheoesophageal Fistula
7.
Am J Surg Pathol ; 46(6): 846-853, 2022 06 01.
Article in English | MEDLINE | ID: mdl-34985046

ABSTRACT

Mutations in the tetratricopeptide repeat domain 7A (TTC7A) gene are a rare cause of congenital enteropathy that can result in significant morbidity. TTC7A deficiency leads to disruption of the intestinal epithelium. The histopathology of this condition has been partly described in case reports and clinical studies. This manuscript describes an in-depth investigation of the pediatric gastrointestinal pathology of the largest histologically examined cohort with confirmed TTC7A mutations reported to date and, for the first time, compared the findings to age-matched and sex-matched control patients with intestinal atresia not thought to be associated with TTC7A mutations. Hematoxylin and eosin-stained slides of endoscopically obtained mucosal biopsies and surgical resection specimens from 7 patients with known TTC7A mutations were examined retrospectively. The microscopic findings were found to be on a spectrum from atresia-predominant to those with predominantly epithelial abnormalities. Several unique histopathologic characteristics were observed when compared with controls. These included neutrophilic colitis and prominent lamina propria eosinophilia throughout the gastrointestinal tract. Striking architectural abnormalities of the epithelium were observed in 4 of the 7 patients. The 5 patients with intestinal atresia demonstrated hypertrophy and disorganization of the colonic muscularis mucosae accompanied by bland spindle cell nodules within the intestinal wall. The components of the latter were further elucidated using immunohistochemistry, and we subsequently hypothesize that they represent obliterated mucosa with remnants of the muscularis mucosae. Finally, atrophic gastritis was noted in 4 patients. In conclusion, the unique histopathologic characteristics of TTC7A mutation-associated enteropathy described herein more fully describe this novel disease entity in infants who present with congenital enteropathy or enterocolitis.


Subject(s)
Germ-Line Mutation , Intestinal Atresia , Proteins , Severe Combined Immunodeficiency , Child , Humans , Infant , Intestinal Atresia/genetics , Intestinal Mucosa/pathology , Intestines/abnormalities , Proteins/genetics , Retrospective Studies , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/pathology
8.
Am J Med Genet A ; 188(4): 1299-1306, 2022 04.
Article in English | MEDLINE | ID: mdl-34970864

ABSTRACT

The beta-actin gene (ACTB) encodes a ubiquitous cytoskeletal protein, essential for embryonic development in humans. De novo heterozygous missense variants in the ACTB are implicated in causing Baraitser-Winter cerebrofrontofacial syndrome (BWCFFS; MIM#243310). ACTB pathogenic variants are rarely associated with intestinal malformations. We report on a rare case of monozygotic twins presenting with proximal small bowel atresia and hydrops in one, and apple-peel bowel atresia and laryngeal dysgenesis in the other. The twin with hydrops could not be resuscitated. Intensive and surgical care was provided to the surviving twin. Rapid trio genome sequencing identified a de novo missense variant in ACTB (NM_00101.3:c.1043C>T; p.(Ser348Leu)) that guided the care plan. The identical variant subsequently was identified in the demised twin. To characterize the functional effect, the variant was recreated as a pseudoheterozygote in a haploid wild-type S. cerevisiae strain. There was an obvious growth defect of the yACT1S348L/WT pseudoheterozygote compared to a yACT1WT/WT strain when grown at 22°C but not when grown at 30°C, consistent with the yACT1 S348L variant having a functional defect that is dominant over the wild-type allele. The functional results provide supporting evidence that the Ser348Leu variant is likely to be a pathogenic variant, including being associated with intestinal malformations in BWCFFS, and can demonstrate variable expressivity within monozygotic twins.


Subject(s)
Intestinal Atresia , Twins, Monozygotic , Actins/genetics , Actins/metabolism , Biological Variation, Population , Craniofacial Abnormalities , Edema , Epilepsy , Facies , Humans , Intellectual Disability , Intestinal Atresia/diagnosis , Intestinal Atresia/genetics , Lissencephaly , Saccharomyces cerevisiae/metabolism , Twins, Monozygotic/genetics
9.
Orphanet J Rare Dis ; 16(1): 455, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34715892

ABSTRACT

BACKGROUND: Homozygous mutations in the transcription factor RFX6 are the cause of the Mitchell-Riley syndrome (MRS) associating neonatal diabetes, congenital digestive system, such as biliary atresia, pancreatic hypoplasia, duodenal and/or jejunal atresia, intestinal malrotation, gallbladder aplasia, cholestasis. A constitutive inactivation of RFX6 leads also to gastric heterotopia. Application of RNA-seq in human diseases may help to better understand pathogenic mechanism of diseases and to predict the risk of developing chronic disorders and personalizing their prevention and treatment. We evaluated oncogenic patterns and cancer predisposition using the transcriptomic profile in a case of MRS with neonatal diabetes, duodenal atresia, and extensive intestinal tract gastric heterotopia. RESULTS: We signalled the interactors of RFX6 with other up and downregulated genes, that may be interested in severity of diabetic condition, in multi-organs impairment and cancer predisposition. Furthermore, several dysregulated genes are involved in biological processes that can lead to promote cancer including "Evading apoptosis" (BAD, BBC3, EGF, FGFR2, FLT3LG, HMOX1, HRAS, IFNAR2, IGF1R, IL12RB1, IL13RA1, IL15, IL2RB, IL2RG, IL6R, KEAP1, MGST1, PDGFA, PDGFRB, PIK3R3, RALB, RALGDS, RASSF1, SOS1, TGFA, TXNRD3), "Proliferation" (APC, BRAF, CCND2, CCND3, CCNE2, FGFR2, FLT3LG, FZD1, FZD6, HMOX1, HRAS, IGF1R, KEAP1, LRP6, MAPK3, MGST1, PDGFA, PDGFB, PDGFRB, RB1, SOS1, TGFA, TXNRD3, WNT10B), "Sustained angiogenesis" (BRAF, FGFR2, FLT3LG, HRAS, IGF1R, JAG1, MAPK3, NOTCH2, PDGFA, PDGFB, PDGFRB, SOS1, TGFA, TGFB1), "Genomic instability" (BAD, BBC3) and "Insensitivity to anti-growth signals" (SMAD2, TGFB1). We also inspected the signalings and their related genes in cancer, such as "PI3K signaling", "ERK signaling", "JAK-STAT signaling", "Calcium signaling", "Other RAS signaling", "WNT signaling". CONCLUSIONS: In our MRS patient, we signaled the interactors of RFX6 with other up- and downregulated genes that may be related to severe diabetic condition, multi-organ impairment, and cancer predisposition. Notably, many dysregulated genes may lead to triggering carcinogenesis. The possibility of the patient developing cancer degeneration in heterotopic gastric mucosa and/or additional long-term tumoral sequelae is not excluded. Personalized prevention and treatment strategies should be proposed.


Subject(s)
Diabetes Mellitus , Intestinal Atresia , Neoplasms , Carcinogenesis , Duodenal Obstruction , Gallbladder Diseases , Gastric Mucosa/metabolism , Humans , Infant, Newborn , Intestinal Atresia/genetics , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Phosphatidylinositol 3-Kinases , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/metabolism , Transcriptome
10.
Brain ; 144(12): 3597-3610, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34415310

ABSTRACT

Phosphatidylinositol 4-kinase IIIα (PI4KIIIα/PI4KA/OMIM:600286) is a lipid kinase generating phosphatidylinositol 4-phosphate (PI4P), a membrane phospholipid with critical roles in the physiology of multiple cell types. PI4KIIIα's role in PI4P generation requires its assembly into a heterotetrameric complex with EFR3, TTC7 and FAM126. Sequence alterations in two of these molecular partners, TTC7 (encoded by TTC7A or TCC7B) and FAM126, have been associated with a heterogeneous group of either neurological (FAM126A) or intestinal and immunological (TTC7A) conditions. Here we show that biallelic PI4KA sequence alterations in humans are associated with neurological disease, in particular hypomyelinating leukodystrophy. In addition, affected individuals may present with inflammatory bowel disease, multiple intestinal atresia and combined immunodeficiency. Our cellular, biochemical and structural modelling studies indicate that PI4KA-associated phenotypical outcomes probably stem from impairment of PI4KIIIα-TTC7-FAM126's organ-specific functions, due to defective catalytic activity or altered intra-complex functional interactions. Together, these data define PI4KA gene alteration as a cause of a variable phenotypical spectrum and provide fundamental new insight into the combinatorial biology of the PI4KIIIα-FAM126-TTC7-EFR3 molecular complex.


Subject(s)
Hereditary Central Nervous System Demyelinating Diseases/genetics , Intestinal Atresia/genetics , Minor Histocompatibility Antigens/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Primary Immunodeficiency Diseases/genetics , Female , Humans , Male , Pedigree , Polymorphism, Single Nucleotide
11.
Front Immunol ; 12: 759308, 2021.
Article in English | MEDLINE | ID: mdl-34975848

ABSTRACT

Rare autosomal-recessive variants in tetratricopeptide repeat domain 7A (TTC7A) gene have been shown to cause intestinal and immune disorders of variable severity. Missense mutations in TTC7A gene, usually retaining most of the functional motifs, is associated with relative milder clinical presentations. In this study, we reported a patient who was suffering from severe multiple intestinal atresia (MIA) with combined immunodeficiency (CID) that led to the pyloric diaphragm, ileum atresia, colon stenosis, and multiple episodes of sepsis. In spite of several surgeries and supportive treatment, the patient died of severe sepsis and multiple organ failure at age of 3 months. The whole exome sequencing (WES) of peripheral blood samples identified a novel homozygous TTC7A missense mutation (c. 206T>C, p. L69P), inherited from his parents with consanguineous marriage. In silico analysis revealed that a hydrogen bond present between Gly65 and Leu69 in the wild-type TTC7A was disrupted by the Leu69Pro mutation. Moreover, this homozygous missense mutation led to a reduced TTC7A expression in lymphocytes and intestinal tissues, accompanied by impeded lymphocyte development. Further studies demonstrated that the PI4K-FAM126A-EFR3A pathway was impaired in colon tissues. Our data strongly support the linkage of severe MIA-CID with the missense mutation in TTC7A gene. More knowledge of the TTC7A protein functions will have important therapeutic implications for patients with MIA-CID.


Subject(s)
Intestinal Atresia/genetics , Mutation, Missense/genetics , Proteins/genetics , Severe Combined Immunodeficiency/genetics , Child , Humans , Intestinal Atresia/immunology , Male , Mutation, Missense/immunology , Proteins/immunology , Severe Combined Immunodeficiency/immunology
12.
Development ; 147(21)2020 11 05.
Article in English | MEDLINE | ID: mdl-33033118

ABSTRACT

Mitchell-Riley syndrome (MRS) is caused by recessive mutations in the regulatory factor X6 gene (RFX6) and is characterised by pancreatic hypoplasia and neonatal diabetes. To determine why individuals with MRS specifically lack pancreatic endocrine cells, we micro-CT imaged a 12-week-old foetus homozygous for the nonsense mutation RFX6 c.1129C>T, which revealed loss of the pancreas body and tail. From this foetus, we derived iPSCs and show that differentiation of these cells in vitro proceeds normally until generation of pancreatic endoderm, which is significantly reduced. We additionally generated an RFX6HA reporter allele by gene targeting in wild-type H9 cells to precisely define RFX6 expression and in parallel performed in situ hybridisation for RFX6 in the dorsal pancreatic bud of a Carnegie stage 14 human embryo. Both in vitro and in vivo, we find that RFX6 specifically labels a subset of PDX1-expressing pancreatic endoderm. In summary, RFX6 is essential for efficient differentiation of pancreatic endoderm, and its absence in individuals with MRS specifically impairs formation of endocrine cells of the pancreas head and tail.


Subject(s)
Cell Differentiation , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Endoderm/embryology , Gallbladder Diseases/genetics , Gallbladder Diseases/pathology , Induced Pluripotent Stem Cells/pathology , Intestinal Atresia/genetics , Intestinal Atresia/pathology , Mutation/genetics , Pancreas/embryology , Regulatory Factor X Transcription Factors/genetics , Alleles , Base Sequence , Cell Differentiation/genetics , Chromatin/metabolism , Consanguinity , Diabetes Mellitus/diagnostic imaging , Embryo, Mammalian/metabolism , Embryonic Development , Family , Female , Gallbladder Diseases/diagnostic imaging , Genome, Human , Humans , Induced Pluripotent Stem Cells/metabolism , Intestinal Atresia/diagnostic imaging , Male , Pedigree , Transcription, Genetic , Transcriptome/genetics , X-Ray Microtomography
13.
PLoS One ; 15(4): e0232023, 2020.
Article in English | MEDLINE | ID: mdl-32352981

ABSTRACT

INTRODUCTION: Intestinal atresia is a rare congenital affliction that is often associated with severe bacterial infections despite adequate neonatal surgery. Previous studies have focused on enteric nervous system variations. We hypothesized that epithelial systems (ES) may also be involved in the pathophysiology of postnatal disorders. MATERIALS AND METHODS: Global gene expression was measured by transcriptomic analysis in a rat model of induced intestinal atresia. The analyses then focused on genes involved in ES (enterocytes and goblet cells). Rat fetus small intestines at various stages of development (ED15, ED17, ED19, and ED21, n = 22), were used as non-operated controls and compared to the upper and lower segments of rat fetus small intestines with an induced atresia (n = 14; ligature at ED18). The pattern of gene expression was then confirmed by histochemistry, electron microscopy, and RT-qPCR. RESULTS: From ED15 to ED21, the expression of several genes exhibited a physiological increase of ES markers, with a significant increase at the end of gestation. The operated embryos exhibited significantly higher variations of gene expression in the proximal segment than in the distal segment in terms of absorption and the epithelial barrier. An increase in goblet cells and markers was observed in the proximal segment compared to the controls. CONCLUSION: Fetal intestinal obstruction accelerates maturation in the proximal segment and disrupts the intestinal wall in the distal segment, with a decrease in the number of mucosal cells. Moreover, the epithelial cells underwent significant changes, supporting the notion that intestinal disorders involve more than the ENS.


Subject(s)
Intestinal Atresia/genetics , Intestinal Atresia/physiopathology , Intestinal Mucosa/physiopathology , Animals , Disease Models, Animal , Enteric Nervous System , Enterocytes/metabolism , Epithelial Cells/metabolism , Female , Fetus , Gastrointestinal Motility/physiology , Gene Expression Profiling/methods , Goblet Cells/metabolism , Intestinal Obstruction/physiopathology , Intestines/physiopathology , Pregnancy , Rats , Rats, Wistar , Transcriptome/genetics
14.
BMC Med Genet ; 21(1): 24, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32028929

ABSTRACT

BACKGROUND: Maturity-onset diabetes of the young (MODY) is a genetically and clinically heterogeneous group of hereditary diabetes, generally caused by one abnormal gene. MODY5 is caused by mutations of the hepatocyte nuclear factor 1 homeobox ß gene (HNF1ß), always as a part of Chr17q12 deletion, whereas heterozygous mutation in B lymphocyte kinase (BLK) gene is responsible for MODY11. CASE PRESENTATION: We report a patient who developed diabetes with a 1.58-Mb Chr17q12 microdeletion and BLK gene c.211G > A mutation using the cytoscan high-density array and whole-exome sequencing analysis. The patient received the surgery at five days after birth for the duodenal atresia and had normal growth postoperatively. Mild elevated liver enzymes were found along with the normal renal function. Quantitative analysis of ß-cell function markers, including fasting insulin (< 0.2 mIU/L), fasting C-peptide (0.02 µg/L), postprandial-2 h insulin (< 0.2 mIU/L), and postprandial-2 h C-peptide (0.03 µg/L) suggested a severe loss of insulin secreting capacity. Meanwhile, islet autoantibodies (GADA, IA-2, ICA, and IAA) in the patient's blood appeared negative. Neither dysplasia in other tissues nor abnormality in development and behavior was found. CONCLUSION: To date, gastrointestinal malformations were extremely rarely reported in patients with MODY. Our clinical report further expands the clinical presentation and variability of MODY5.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Duodenal Obstruction/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Intestinal Atresia/genetics , src-Family Kinases/genetics , Diabetes Mellitus, Type 2/pathology , Duodenal Obstruction/pathology , Female , Humans , Infant, Newborn , Insulin/genetics , Intestinal Atresia/pathology , Male , Mutation/genetics , Phenotype
15.
Eur J Med Genet ; 63(5): 103844, 2020 May.
Article in English | MEDLINE | ID: mdl-31953238

ABSTRACT

This report describes siblings with Stromme syndrome, a rare genetic condition that primarily presents with a triad of intestinal atresia, cranial and ocular malformations, and other organ systems could be involved. This clinical triad was initially named after the first person to describe it in 1993. Here, we report a family with two siblings who presented with unusual intestinal atresia and ocular and CNS abnormalities. The first patient is a 6-year-old-boy with apple peel duodeno-jejunal atresia, unilateral microphthalmia and microcephaly. The second patient, a younger brother, presented with intestinal atresia, corneal opacity and alobar holoprosencephaly and passed away at the age of 3 months. Exome sequencing showed a novel homozygous variant in the CENPF gene, NM_016343.3: c.1195-2 A > G that was detected in both of the affected siblings. This is a report and literature review of CENPF-related ciliopathy, which may result in Stromme syndrome. As this is the fourth report linking the CENPF gene variant with Stromme syndrome and first reported case presented with holoprosencephaly, it will expand the current knowledge on the genotype and the phenotype of Stromme syndrome.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Eye Abnormalities/genetics , Intestinal Atresia/genetics , Microcephaly/genetics , Microfilament Proteins/genetics , Phenotype , Child , Eye Abnormalities/pathology , Homozygote , Humans , Infant , Intestinal Atresia/pathology , Male , Microcephaly/pathology , Mutation , Pedigree
16.
Fetal Diagn Ther ; 47(2): 98-103, 2020.
Article in English | MEDLINE | ID: mdl-31167209

ABSTRACT

BACKGROUND: The "double bubble" sign is an ultrasonographic finding that commonly represents duodenal atresia and is associated with trisomy 21. OBJECTIVES: We sought to evaluate the positive predictive value of a prenatally identified double bubble sign for duodenal atresia and the genetic etiologies associated with it. METHODS: We examined a retrospective cohort with prenatal double bubble sign between January 1, 2008, and June 30, 2017. Postnatal diagnoses were determined by review of operative reports and additional postnatal evaluation including cytogenetic analysis, molecular analysis, and/or clinical genetic evaluation. RESULTS: All live births at our institution with a prenatal double bubble sign had confirmed duodenal atresia. Additional anatomic anomalies and/or genetic abnormalities were identified in 62% of cases. Out of 21 cases, 6 had trisomy 21. Of the remaining 15 cases, 8 were nonisolated duodenal atresia, 3 of which had a heterotaxy syndrome. In the 7 isolated cases, 1 likely pathogenic chromosomal microdeletion was identified. CONCLUSIONS: Prenatal double bubble sign is a reliable predictor of duodenal atresia. In addition to trisomy 21, heterotaxy may be encountered. ZIC3 mutations as well as microdeletion of 4q22.3 may be underlying genetic etiologies to be considered in the diagnostic evaluation of a prenatal double bubble sign.


Subject(s)
Duodenal Obstruction/diagnostic imaging , Intestinal Atresia/diagnostic imaging , Ultrasonography, Prenatal , Chromosome Deletion , Chromosomes, Human, Pair 4 , Down Syndrome/genetics , Duodenal Obstruction/genetics , Genetic Predisposition to Disease , Gestational Age , Heterotaxy Syndrome/genetics , Humans , Intestinal Atresia/genetics , Predictive Value of Tests , Retrospective Studies , Risk Factors
17.
Eur J Hum Genet ; 28(1): 132-136, 2020 01.
Article in English | MEDLINE | ID: mdl-31488893

ABSTRACT

NAME OF THE DISEASE (SYNONYMS): Stromme syndrome.Jejunal atresia with microcephaly and ocular anomalies.Apple peel syndrome with microcephaly and ocular anomalies.Ciliopathy phenotype.Primary microcephaly and intellectual disability.OMIM# of the disease 243605.Name of the analysed genes or DNA/chromosome segments CENPF.OMIM# of the gene(s) 600236.Review of the analytical and clinical validity as well as of the clinical utility of DNA-based testing for mutations in CENPF genes in diagnostic, prenatal settings, and for risk assessment in relatives.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Eye Abnormalities/genetics , Genetic Testing/methods , Intestinal Atresia/genetics , Microcephaly/genetics , Microfilament Proteins/genetics , Eye Abnormalities/diagnosis , Genetic Testing/standards , Humans , Intestinal Atresia/diagnosis , Microcephaly/diagnosis , Mutation , Phenotype , Sensitivity and Specificity
18.
BMJ Case Rep ; 12(8)2019 Aug 26.
Article in English | MEDLINE | ID: mdl-31451461

ABSTRACT

This is a case report of a neonate who was antenatally diagnosed with jejunal atresia which turned out to be duodenal atresia with apple peel syndrome. A previous sibling, who also had apple peel but with jejunal atresia, succumbed to sepsis after surgery. The first sibling had jejunal stenosis and had died of sepsis following surgery. Combination of duodenal atresia with apple peel is extremely rare. This coupled with a familial condition is rarer still. This case was challenging due to the short length of the gut and prolonged need for total parenteral nutrition and sepsis in postoperative period.


Subject(s)
Digestive System Surgical Procedures/methods , Duodenal Obstruction , Intestinal Atresia , Jejunostomy/methods , Neonatal Sepsis , Adult , Diagnosis, Differential , Duodenal Obstruction/diagnosis , Duodenal Obstruction/etiology , Duodenal Obstruction/physiopathology , Duodenal Obstruction/surgery , Duodenum/abnormalities , Duodenum/diagnostic imaging , Duodenum/surgery , Female , Humans , Infant, Newborn , Intestinal Atresia/diagnosis , Intestinal Atresia/genetics , Intestinal Atresia/physiopathology , Intestinal Atresia/surgery , Jejunum/abnormalities , Jejunum/diagnostic imaging , Jejunum/surgery , Medical History Taking , Neonatal Sepsis/diagnosis , Neonatal Sepsis/etiology , Neonatal Sepsis/therapy , Parenteral Nutrition, Total/methods , Pregnancy , Prenatal Diagnosis/methods , Rare Diseases/diagnosis , Siblings , Treatment Outcome
20.
Am J Med Genet A ; 179(8): 1426-1431, 2019 08.
Article in English | MEDLINE | ID: mdl-31183986

ABSTRACT

Down syndrome is the most common human chromosomal disorder. Among clinical findings, one constant concern is the high prevalence of gastrointestinal system alterations. The aim of this study was to determine the prevalence of gastrointestinal disorders at a Down syndrome outpatient clinic during a 10-year follow-up period. Data from medical files were retrospectively reviewed from 1,207 patients. Gastrointestinal changes occurred in 612 (50.7%). The most prevalent disorder was chronic intestinal constipation. Intestinal parasite occurred in 22% (mainly giardiasis), gastroesophageal reflux disease in 14%, digestive tract malformations occurred in 5%: 13 cases of duodenal atresia, 8 of imperforate anus, 4 annular pancreases, 2 congenital megacolon, 2 esophageal atresias, 2 esophageal compression by anomalous subclavian and 1 case of duodenal membrane. We had 38/1,207 (3.1%) patients with difficulty in sucking and only three with dysphagia that resolved before the second year of life. Peptic ulcer disease, celiac disease, and biliary lithiasis were less prevalent with 3% each. Awareness of the high prevalence of gastrointestinal disorders promotes outstanding clinical follow-up as well as adequate development and greater quality of life for patients with Down syndrome and their families.


Subject(s)
Anus, Imperforate/complications , Constipation/complications , Down Syndrome/complications , Duodenal Obstruction/complications , Esophageal Atresia/complications , Gastroesophageal Reflux/complications , Giardiasis/complications , Hirschsprung Disease/complications , Intestinal Atresia/complications , Adolescent , Adult , Anus, Imperforate/diagnosis , Anus, Imperforate/genetics , Anus, Imperforate/pathology , Brazil , Child , Child, Preschool , Constipation/diagnosis , Constipation/genetics , Constipation/pathology , Cross-Sectional Studies , Down Syndrome/diagnosis , Down Syndrome/genetics , Down Syndrome/pathology , Duodenal Obstruction/diagnosis , Duodenal Obstruction/genetics , Duodenal Obstruction/pathology , Esophageal Atresia/diagnosis , Esophageal Atresia/genetics , Esophageal Atresia/pathology , Female , Gastroesophageal Reflux/diagnosis , Gastroesophageal Reflux/genetics , Gastroesophageal Reflux/pathology , Gastrointestinal Tract/abnormalities , Gastrointestinal Tract/metabolism , Giardiasis/diagnosis , Giardiasis/genetics , Giardiasis/pathology , Hirschsprung Disease/diagnosis , Hirschsprung Disease/genetics , Hirschsprung Disease/pathology , Humans , Infant , Infant, Newborn , Intestinal Atresia/diagnosis , Intestinal Atresia/genetics , Intestinal Atresia/pathology , Male , Quality of Life/psychology , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...