Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 42(6): 1185-96, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26084028

ABSTRACT

The autoimmune regulator (AIRE) gene is crucial for establishing central immunological tolerance and preventing autoimmunity. Mutations in AIRE cause a rare autosomal-recessive disease, autoimmune polyendocrine syndrome type 1 (APS-1), distinguished by multi-organ autoimmunity. We have identified multiple cases and families with mono-allelic mutations in the first plant homeodomain (PHD1) zinc finger of AIRE that followed dominant inheritance, typically characterized by later onset, milder phenotypes, and reduced penetrance compared to classical APS-1. These missense PHD1 mutations suppressed gene expression driven by wild-type AIRE in a dominant-negative manner, unlike CARD or truncated AIRE mutants that lacked such dominant capacity. Exome array analysis revealed that the PHD1 dominant mutants were found with relatively high frequency (>0.0008) in mixed populations. Our results provide insight into the molecular action of AIRE and demonstrate that disease-causing mutations in the AIRE locus are more common than previously appreciated and cause more variable autoimmune phenotypes.


Subject(s)
DNA Mutational Analysis/methods , Genes, Dominant/genetics , Mutation/genetics , Polyendocrinopathies, Autoimmune/genetics , Transcription Factors/genetics , Adolescent , Adult , Amino Acid Sequence , Autoimmunity/genetics , Child , Child, Preschool , Female , Gene Frequency , Humans , Male , Microsatellite Repeats/genetics , Molecular Sequence Data , Norway , Organ Specificity/genetics , Pedigree , Penetrance , Phenotype , Russia , Young Adult , AIRE Protein
2.
Am J Hum Genet ; 104(4): 749-757, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30905398

ABSTRACT

Over a relatively short period of time, the clinical geneticist's "toolbox" has been expanded by machine-learning algorithms for image analysis, which can be applied to the task of syndrome identification on the basis of facial photographs, but these technologies harbor potential beyond the recognition of established phenotypes. Here, we comprehensively characterized two individuals with a hitherto unknown genetic disorder caused by the same de novo mutation in LEMD2 (c.1436C>T;p.Ser479Phe), the gene which encodes the nuclear envelope protein LEM domain-containing protein 2 (LEMD2). Despite different ages and ethnic backgrounds, both individuals share a progeria-like facial phenotype and a distinct combination of physical and neurologic anomalies, such as growth retardation; hypoplastic jaws crowded with multiple supernumerary, yet unerupted, teeth; and cerebellar intention tremor. Immunofluorescence analyses of patient fibroblasts revealed mutation-induced disturbance of nuclear architecture, recapitulating previously published data in LEMD2-deficient cell lines, and additional experiments suggested mislocalization of mutant LEMD2 protein within the nuclear lamina. Computational analysis of facial features with two different deep neural networks showed phenotypic proximity to other nuclear envelopathies. One of the algorithms, when trained to recognize syndromic similarity (rather than specific syndromes) in an unsupervised approach, clustered both individuals closely together, providing hypothesis-free hints for a common genetic etiology. We show that a recurrent de novo mutation in LEMD2 causes a nuclear envelopathy whose prognosis in adolescence is relatively good in comparison to that of classical Hutchinson-Gilford progeria syndrome, and we suggest that the application of artificial intelligence to the analysis of patient images can facilitate the discovery of new genetic disorders.


Subject(s)
Membrane Proteins/genetics , Mutation , Nuclear Proteins/genetics , Progeria/genetics , Adolescent , Artificial Intelligence , Cell Line, Tumor , Cell Nucleus , Child , Child, Preschool , Diagnosis, Computer-Assisted , Face , Fibroblasts/metabolism , Humans , Male , Mass Screening/methods , Medical Informatics , Phenotype , Prognosis , Syndrome
3.
Am J Hum Genet ; 100(5): 737-750, 2017 May 04.
Article in English | MEDLINE | ID: mdl-28457472

ABSTRACT

Keratolytic winter erythema (KWE) is a rare autosomal-dominant skin disorder characterized by recurrent episodes of palmoplantar erythema and epidermal peeling. KWE was previously mapped to 8p23.1-p22 (KWE critical region) in South African families. Using targeted resequencing of the KWE critical region in five South African families and SNP array and whole-genome sequencing in two Norwegian families, we identified two overlapping tandem duplications of 7.67 kb (South Africans) and 15.93 kb (Norwegians). The duplications segregated with the disease and were located upstream of CTSB, a gene encoding cathepsin B, a cysteine protease involved in keratinocyte homeostasis. Included in the 2.62 kb overlapping region of these duplications is an enhancer element that is active in epidermal keratinocytes. The activity of this enhancer correlated with CTSB expression in normal differentiating keratinocytes and other cell lines, but not with FDFT1 or NEIL2 expression. Gene expression (qPCR) analysis and immunohistochemistry of the palmar epidermis demonstrated significantly increased expression of CTSB, as well as stronger staining of cathepsin B in the stratum granulosum of affected individuals than in that of control individuals. Analysis of higher-order chromatin structure data and RNA polymerase II ChIA-PET data from MCF-7 cells did not suggest remote effects of the enhancer. In conclusion, KWE in South African and Norwegian families is caused by tandem duplications in a non-coding genomic region containing an active enhancer element for CTSB, resulting in upregulation of this gene in affected individuals.


Subject(s)
Cathepsin B/metabolism , Enhancer Elements, Genetic , Erythema/genetics , Gene Duplication , Gene Expression Regulation , Keratosis/genetics , Skin Diseases, Genetic/genetics , Case-Control Studies , Cathepsin B/genetics , Chromosome Mapping , Chromosomes, Human, Pair 8/genetics , DNA Copy Number Variations , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Epidermis/metabolism , Epigenomics , Erythema/epidemiology , Female , Genetic Markers , Humans , Keratinocytes/metabolism , Keratosis/epidemiology , MCF-7 Cells , Male , Norway/epidemiology , Pedigree , Skin Diseases, Genetic/epidemiology , South Africa/epidemiology
4.
Scand J Gastroenterol ; 55(4): 449-453, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32306784

ABSTRACT

Background: Guanylin (GN) and uroguanylin (UGN) are endogenous ligands for the intestinal receptor guanylate cyclase C (GC-C), an important regulator of intestinal fluid homeostasis. Gene expression and protein levels of GN are suppressed in inflamed intestinal tissue from patients with inflammatory bowel disease (IBD), but knowledge about plasma levels of guanylins in these conditions is sparse. We aimed to investigate the fasting plasma levels of the prohormones proGN and proUGN in patients with Crohn's Disease (CD) and relate these to levels found in persons with other diarrheal conditions, as well as persons with normal bowel habits.Methods: Plasma from patients with CD, patients with Familial GUCY2C Diarrheal Disease (FGDS), diarrhea-predominant irritable bowel syndrome (IBS-D) and healthy controls (HC) was analyzed using ELISA assays.Results: Significantly lower fasting plasma levels of proguanylins were found in CD and FGDS patients, compared to HC. In CD patients, plasma proGN levels correlated negatively with Harvey Bradshaw Index and with number of stools/24 h.Conclusion: Our data indicate that diarrhea may be a determinant for levels of proGN in plasma, and should be further explored in studies of different diarrheal disorders.


Subject(s)
Crohn Disease/blood , Diarrhea/blood , Gastrointestinal Hormones/blood , Irritable Bowel Syndrome/blood , Natriuretic Peptides/blood , Adolescent , Adult , Aged , Case-Control Studies , Child , Child, Preschool , Diarrhea/genetics , Female , Gene Expression , Humans , Irritable Bowel Syndrome/genetics , Male , Middle Aged , Plasma/chemistry , Receptors, Enterotoxin/genetics , Young Adult
5.
Epilepsia ; 59(8): 1595-1602, 2018 08.
Article in English | MEDLINE | ID: mdl-29920680

ABSTRACT

OBJECTIVE: Epilepsy is common in individuals with mutations in POLG, the gene encoding the catalytic subunit of the mitochondrial DNA polymerase gamma. Early recognition and aggressive seizure management are crucial for patient survival. Disruption of the blood-brain barrier (BBB) is implicated in various neurological disorders including epilepsy. The aim of this study was to assess whether POLG-related disease is associated with BBB dysfunction and what clinical implications this has for patients. METHODS: Our retrospective study used data from 83 patients with pathogenic POLG mutations from 4 countries--Norway, Sweden, Finland, and the United Kingdom. Data were collected using a structured questionnaire. We used the presence of raised cerebrospinal fluid (CSF) protein and a raised CSF/serum ratio of albumin (Q-alb) to evaluate the integrity of the blood-CSF barrier. RESULTS: Raised CSF protein was found in 70% of patients (n = 58/83) and appeared to be associated with the most severe phenotypes. In those in whom it was measured, the Q-alb ratio was markedly elevated (n = 18). The majority of those with epilepsy (n = 50/66, 76%) had raised CSF protein, and this preceded seizure debut in 75% (n = 15/20). The median survival time from symptom onset for those with raised CSF protein was decreased (13 months) compared to those with normal CSF protein (32 months). SIGNIFICANCE: Our results indicate that there is disruption of the BBB in POLG-related disease, as evidenced by a raised CSF protein and Q-alb ratio. We also find that raised CSF protein is a common finding in patients with POLG disease. Our data suggest that the presence of BBB dysfunction predicts a poorer outcome, and elevated CSF protein may therefore be an additional biomarker both for early diagnosis and to identify those at high risk of developing epilepsy.


Subject(s)
Blood-Brain Barrier/physiopathology , Cerebrospinal Fluid Proteins/metabolism , DNA Polymerase gamma/genetics , Epilepsy , Mutation/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Epilepsy/cerebrospinal fluid , Epilepsy/diagnosis , Epilepsy/genetics , Female , Humans , Infant , Infant, Newborn , International Cooperation , Male , Middle Aged , Retrospective Studies , Young Adult
6.
Scand J Gastroenterol ; 53(10-11): 1264-1273, 2018.
Article in English | MEDLINE | ID: mdl-30353760

ABSTRACT

OBJECTIVE: Activating mutations in the GUCY2C gene, which encodes the epithelial receptor guanylate cyclase C, cause diarrhea due to increased loss of sodium chloride to the intestinal lumen. Patients with familial GUCY2C diarrhea syndrome (FGDS) are predisposed to inflammatory bowel disease (IBD). We investigated whether genes in the guanylate cyclase C pathway are enriched for association with IBD and reversely whether genetic or transcriptional changes associated with IBD are found in FGDS patients. METHODS: (1) A set of 27 genes from the guanylate cyclase C pathway was tested for enrichment of association with IBD by Gene Set Enrichment Analysis, using genome-wide association summary statistics from 12,882 IBD patients and 21,770 controls. (2) We genotyped 163 known IBD risk loci and sequenced NOD2 in 22 patients with FGDS. Eight of them had concomitant Crohn's disease. (3) Global gene expression analysis was performed in ileal tissue from patients with FGDS, Crohn's disease and healthy individuals. RESULTS: The guanylate cyclase C gene set showed a significant enrichment of association in IBD genome-wide association data. Risk variants in NOD2 were found in 7/8 FGDS patients with concomitant Crohn's disease and in 2/14 FDGS patients without Crohn's disease. In ileal tissue, downregulation of metallothioneins characterized FGDS patients compared to healthy controls. CONCLUSIONS: Our results support a role of guanylate cyclase C signaling and disturbed electrolyte homeostasis in development of IBD. Furthermore, downregulation of metallothioneins in the ileal mucosa of FGDS patients may contribute to IBD development, possibly alongside effects from NOD2 risk variants.


Subject(s)
Diarrhea/genetics , Inflammatory Bowel Diseases/genetics , Receptors, Enterotoxin/genetics , Adult , Aged , Case-Control Studies , Diarrhea/metabolism , Down-Regulation , Family Health , Female , Gene Expression , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Ileum/pathology , Inflammatory Bowel Diseases/complications , Male , Middle Aged , Nod2 Signaling Adaptor Protein/genetics , Norway , Plasma/chemistry , Risk Assessment , Syndrome , Young Adult
7.
Bioinformatics ; 32(19): 3018-20, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27288501

ABSTRACT

MOTIVATION: The search for causative genetic variants in rare diseases of presumed monogenic inheritance has been boosted by the implementation of whole exome (WES) and whole genome (WGS) sequencing. In many cases, WGS seems to be superior to WES, but the analysis and visualization of the vast amounts of data is demanding. RESULTS: To aid this challenge, we have developed a new tool-RareVariantVis-for analysis of genome sequence data (including non-coding regions) for both germ line and somatic variants. It visualizes variants along their respective chromosomes, providing information about exact chromosomal position, zygosity and frequency, with point-and-click information regarding dbSNP IDs, gene association and variant inheritance. Rare variants as well as de novo variants can be flagged in different colors. We show the performance of the RareVariantVis tool in the Genome in a Bottle WGS data set. AVAILABILITY AND IMPLEMENTATION: https://www.bioconductor.org/packages/3.3/bioc/html/RareVariantVis.html CONTACT: tomasz.stokowy@k2.uib.no SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Exome , Genome, Human , Rare Diseases/genetics , Sequence Analysis, DNA/methods , Genetic Variation , Humans
8.
J Inherit Metab Dis ; 40(6): 861-866, 2017 11.
Article in English | MEDLINE | ID: mdl-28865037

ABSTRACT

BACKGROUND: Mitochondria play an important role in iron metabolism and haematopoietic cell homeostasis. Recent studies in mice showed that a mutation in the catalytic subunit of polymerase gamma (POLG) was associated with haematopoietic dysfunction including anaemia. The aim of this study was to analyse the frequency of anaemia in a large cohort of patients with POLG related disease. METHODS: We conducted a multi-national, retrospective study of 61 patients with confirmed, pathogenic biallelic POLG mutations from six centres, four in Norway and two in the United Kingdom. Clinical, laboratory and genetic data were collected using a structured questionnaire. Anaemia was defined as an abnormally low haemoglobin value adjusted for age and sex. Univariate survival analysis was performed using log-rank test to compare differences in survival time between categories. RESULTS: Anaemia occurred in 67% (41/61) of patients and in 23% (14/61) it was already present at clinical presentation. The frequency of anaemia in patients with early onset disease including Alpers syndrome and myocerebrohepatopathy spectrum (MCHS) was high (72%) and 35% (8/23) of these had anaemia at presentation. Survival analysis showed that the presence of anaemia was associated with a significantly worse survival (P = 0.004). CONCLUSION: Our study reveals that anaemia can be a feature of POLG-related disease. Further, we show that its presence is associated with significantly worse prognosis either because anaemia itself is impacting survival or because it reflects the presence of more serious disease. In either case, our data suggests anaemia is a marker for negative prognosis.


Subject(s)
Anemia/etiology , Anemia/genetics , DNA Polymerase gamma/genetics , Adolescent , Child , Child, Preschool , Diffuse Cerebral Sclerosis of Schilder/genetics , Female , Humans , Infant , Infant, Newborn , Male , Mutation/genetics , Pilot Projects , Retrospective Studies , United Kingdom
9.
Am J Gastroenterol ; 111(10): 1467-1475, 2016 10.
Article in English | MEDLINE | ID: mdl-27527747

ABSTRACT

OBJECTIVES: The objective of this study was to study the prevalence of gastrointestinal (GI) symptoms and histopathology in patients with common variable immunodeficiency (CVID) as well as linking the findings to GI infections and markers of systemic immune activation. METHODS: In this cross-sectional study, we addressed GI symptoms in 103 patients and GI histopathological findings in 53 patients who underwent upper and lower endoscopic examination. The most frequent histopathological findings were linked to GI symptoms, B-cell phenotype, and markers of systemic immune activation (soluble (s)CD14, sCD25, and sCD163). Microarray analysis compared "celiac-like disease" in CVID to celiac disease. Screening for selected bacterial and viral infections in fecal samples and gut mucosal biopsies was performed. RESULTS: The main findings of this study were as follows: most common GI symptoms were bloating (34%), pain (30%), and diarrhea (26%). The most frequent histopathological findings were increased intraepithelial lymphocytes in the descending part of the duodenum, i.e., "celiac-like disease" (46% of patients), decreased numbers of plasma cells in GI tract mucosa (62%), and lymphoid hyperplasia (38%), none of which were associated with GI symptoms. Reduced plasma cells in GI mucosa were associated with B-cell phenotypic characteristics of CVID, and increased serum levels of sCD14 (P=0.025), sCD25 (P=0.01), and sCD163 (P=0.04). Microarray analyses distinguished between CVID patients with "celiac-like disease" and celiac disease. Positive tests for bacterial and viral infections were scarce both in fecal samples and gut mucosal biopsies, including PCR test for norovirus in biopsy specimens (0 positive tests). CONCLUSIONS: In conclusion, GI pathology is common in CVID, but does not necessarily cause symptoms. However, reduced plasma cells in GI mucosa were linked to systemic immune activation, "celiac-like disease" in CVID and true celiac disease appear to be different disease entities, as assessed by gene expression, and infections (including norovirus) are rarely a cause of the CVID enteropathy.


Subject(s)
Common Variable Immunodeficiency/epidemiology , Gastrointestinal Diseases/epidemiology , Abdominal Pain/epidemiology , Abdominal Pain/immunology , Abdominal Pain/pathology , Adult , Aged , Aged, 80 and over , B-Lymphocytes/immunology , Celiac Disease/epidemiology , Celiac Disease/genetics , Celiac Disease/immunology , Celiac Disease/pathology , Colonoscopy , Common Variable Immunodeficiency/immunology , Constipation/epidemiology , Constipation/immunology , Constipation/pathology , Cross-Sectional Studies , Diarrhea/epidemiology , Diarrhea/immunology , Diarrhea/pathology , Duodenum/pathology , Endoscopy, Digestive System , Esophageal Mucosa/pathology , Female , Gastric Mucosa/pathology , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/pathology , Gastrointestinal Tract/pathology , Humans , Intestinal Mucosa/pathology , Lymphocytes/pathology , Male , Middle Aged , Plasma Cells/pathology , Prevalence , Transcriptome , Young Adult
10.
Scand J Gastroenterol ; 51(11): 1308-15, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27338166

ABSTRACT

OBJECTIVE: Familial GUCY2C diarrhoea syndrome (FGDS) is caused by an activating mutation in the GUCY2C gene encoding the receptor guanylate cyclase C in enterocytes. Activation leads to increased secretion of fluid into the intestinal lumen. Twenty percent of the patients have increased risk of Crohn's disease and intestinal obstruction (CD, 20%) and the condition resembles irritable bowel syndrome with diarrhoea. We aimed to describe fluid content, contractility, peristaltic activity and bowel wall thickness in the intestine in fasting FGDS patients, using ultrasound, with healthy volunteers serving as controls. METHODS: Twenty-three patients with FGDS and 22 healthy controls (HC) were examined with a Logiq E9 scanner in a fasting state. Bowel wall thickness was measured and fluid-filled small bowel loops were counted using three-dimensional (3D) magnetic positioning navigation. The HC ingested 500 ml PEG solution, an electrolyte balanced, non-absorbable solution, in order to investigate the contractions of the small bowel. RESULTS: The fasting 23 FGDS patients had significantly higher number of fluid-filled small bowel segments compared to 22 fasting HC, p < 0.001. A high number of non-occlusive contractions in the ileum was observed, which was significant when compared to HC after ingesting PEG solution, p < 0.016. An increase in intestinal wall thickness or other signs of CD were not observed. CONCLUSIONS: FGDS is characterised by multiple, fluid-filled small bowel loops with incomplete contractions and fluid stagnation in fasting state. These findings may play a role in the increased risk of bowel obstruction as well as IBS-like symptoms observed in these patients.


Subject(s)
Diarrhea/physiopathology , Ileum/physiopathology , Intestinal Diseases/genetics , Intestine, Small/physiopathology , Peristalsis , Receptors, Guanylate Cyclase-Coupled/genetics , Receptors, Peptide/genetics , Adult , Aged , Case-Control Studies , Crohn Disease/diagnostic imaging , Diarrhea/etiology , Female , Humans , Ileum/diagnostic imaging , Intestinal Diseases/diagnostic imaging , Intestinal Obstruction/diagnostic imaging , Intestine, Small/diagnostic imaging , Linear Models , Male , Middle Aged , Mutation , Receptors, Enterotoxin , Ultrasonography , Young Adult
11.
N Engl J Med ; 366(17): 1586-95, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22436048

ABSTRACT

BACKGROUND: Familial diarrhea disorders are, in most cases, severe and caused by recessive mutations. We describe the cause of a novel dominant disease in 32 members of a Norwegian family. The affected members have chronic diarrhea that is of early onset, is relatively mild, and is associated with increased susceptibility to inflammatory bowel disease, small-bowel obstruction, and esophagitis. METHODS: We used linkage analysis, based on arrays with single-nucleotide polymorphisms, to identify a candidate region on chromosome 12 and then sequenced GUCY2C, encoding guanylate cyclase C (GC-C), an intestinal receptor for bacterial heat-stable enterotoxins. We performed exome sequencing of the entire candidate region from three affected family members, to exclude the possibility that mutations in genes other than GUCY2C could cause or contribute to susceptibility to the disease. We carried out functional studies of mutant GC-C using HEK293T cells. RESULTS: We identified a heterozygous missense mutation (c.2519G→T) in GUCY2C in all affected family members and observed no other rare variants in the exons of genes in the candidate region. Exposure of the mutant receptor to its ligands resulted in markedly increased production of cyclic guanosine monophosphate (cGMP). This may cause hyperactivation of the cystic fibrosis transmembrane regulator (CFTR), leading to increased chloride and water secretion from the enterocytes, and may thus explain the chronic diarrhea in the affected family members. CONCLUSIONS: Increased GC-C signaling disturbs normal bowel function and appears to have a proinflammatory effect, either through increased chloride secretion or additional effects of elevated cellular cGMP. Further investigation of the relevance of genetic variants affecting the GC-C-CFTR pathway to conditions such as Crohn's disease is warranted. (Funded by Helse Vest [Western Norway Regional Health Authority] and the Department of Science and Technology, Government of India.).


Subject(s)
Diarrhea/genetics , Mutation, Missense , Receptors, Guanylate Cyclase-Coupled/genetics , Receptors, Peptide/genetics , Chronic Disease , Cyclic GMP/biosynthesis , Diarrhea/metabolism , Female , Genetic Linkage , Heterozygote , Humans , Male , Pedigree , Polymorphism, Single Nucleotide , Receptors, Enterotoxin , Receptors, Guanylate Cyclase-Coupled/metabolism , Receptors, Peptide/metabolism , Signal Transduction
12.
Am J Hum Genet ; 89(5): 634-43, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-22019273

ABSTRACT

A subset of ciliopathies, including Sensenbrenner, Jeune, and short-rib polydactyly syndromes are characterized by skeletal anomalies accompanied by multiorgan defects such as chronic renal failure and retinitis pigmentosa. Through exome sequencing we identified compound heterozygous mutations in WDR19 in a Norwegian family with Sensenbrenner syndrome. In a Dutch family with the clinically overlapping Jeune syndrome, a homozygous missense mutation in the same gene was found. Both families displayed a nephronophthisis-like nephropathy. Independently, we also identified compound heterozygous WDR19 mutations by exome sequencing in a Moroccan family with isolated nephronophthisis. WDR19 encodes IFT144, a member of the intraflagellar transport (IFT) complex A that drives retrograde ciliary transport. We show that IFT144 is absent from the cilia of fibroblasts from one of the Sensenbrenner patients and that ciliary abundance and morphology is perturbed, demonstrating the ciliary pathogenesis. Our results suggest that isolated nephronophthisis, Jeune, and Sensenbrenner syndromes are clinically overlapping disorders that can result from a similar molecular cause.


Subject(s)
Cilia , Ectodermal Dysplasia/genetics , Mutation, Missense , Polycystic Kidney Diseases/genetics , Proteins/genetics , Short Rib-Polydactyly Syndrome/genetics , Thoracic Diseases/genetics , Adolescent , Adult , Child , Cilia/genetics , Cilia/pathology , Craniofacial Abnormalities/genetics , Cytoskeletal Proteins , Exome/genetics , Female , Fibroblasts/metabolism , Flagella/genetics , Flagella/pathology , Humans , Intracellular Signaling Peptides and Proteins , Male , Molecular Sequence Data , Morocco , Netherlands , Norway , Oligonucleotide Array Sequence Analysis , Pedigree , Polycystic Kidney Diseases/congenital , Young Adult
13.
Am J Hum Genet ; 87(3): 410-7, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20797687

ABSTRACT

Polyneuropathy, hearing loss, ataxia, retinitis pigmentosa, and cataract (PHARC) is a neurodegenerative disease marked by early-onset cataract and hearing loss, retinitis pigmentosa, and involvement of both the central and peripheral nervous systems, including demyelinating sensorimotor polyneuropathy and cerebellar ataxia. Previously, we mapped this Refsum-like disorder to a 16 Mb region on chromosome 20. Here we report that mutations in the ABHD12 gene cause PHARC disease and we describe the clinical manifestations in a total of 19 patients from four different countries. The ABHD12 enzyme was recently shown to hydrolyze 2-arachidonoyl glycerol (2-AG), the main endocannabinoid lipid transmitter that acts on cannabinoid receptors CB1 and CB2. Our data therefore represent an example of an inherited disorder related to endocannabinoid metabolism. The endocannabinoid system is involved in a wide range of physiological processes including neurotransmission, mood, appetite, pain appreciation, addiction behavior, and inflammation, and several potential drugs targeting these pathways are in development for clinical applications. Our findings show that ABHD12 performs essential functions in both the central and peripheral nervous systems and the eye. Any future drug-mediated interference with this enzyme should consider the potential risk of long-term adverse effects.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Metabolism, Inborn Errors/genetics , Monoacylglycerol Lipases/genetics , Mutation/genetics , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/genetics , Abnormalities, Multiple/enzymology , Abnormalities, Multiple/genetics , Adolescent , Animals , Child , Female , Gene Expression Profiling , Gene Expression Regulation , Genotype , Humans , Male , Metabolism, Inborn Errors/enzymology , Mice , Middle Aged , Monoacylglycerol Lipases/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Syndrome , Young Adult
15.
Am J Med Genet A ; 155A(10): 2397-408, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22043478

ABSTRACT

Osteopathia striata with cranial sclerosis (OSCS) is an X-linked disease caused by truncating mutations in WTX. Females exhibit sclerotic striations on the long bones, cranial sclerosis, and craniofacial dysmorphism. Males with OSCS have significant skeletal sclerosis, do not have striations but do display a more severe phenotype commonly associated with gross structural malformations, patterning defects, and significant pre- and postnatal lethality. The recent description of mutations in WTX underlying OSCS has led to the identification of a milder, survivable phenotype in males. Individuals with this presentation can have, in addition to skeletal sclerosis, Hirschsprung disease, joint contractures, cardiomyopathy, and neuromuscular anomalies. A diagnosis of OSCS should be considered in males with macrocephaly, skeletal sclerosis that is most marked in the cranium and the absence of metaphyseal striations. The observation of striations in males may be indicative of a WTX mutation in a mosaic state supporting the contention that this sign in females is indicative of the differential lyonization of cells in the osteoblastic lineage.


Subject(s)
Genetic Diseases, X-Linked/pathology , Osteosclerosis/pathology , Phenotype , Adaptor Proteins, Signal Transducing/genetics , Bone and Bones/pathology , DNA Mutational Analysis , DNA Primers/genetics , Female , Genetic Diseases, X-Linked/genetics , Humans , Luciferases , Male , Megalencephaly/pathology , Osteosclerosis/genetics , Tumor Suppressor Proteins/genetics
16.
Tidsskr Nor Laegeforen ; 131(15): 1420-3, 2011 Aug 09.
Article in Norwegian | MEDLINE | ID: mdl-21844942

ABSTRACT

BACKGROUND: The trichorhinophalangeal syndrome (TRPS) is a hereditary, skeletal dysplasia which has a characteristic clinical presentation and is classified in types 1, 2 and 3, based on phenotype and genotype. Typical findings may be mild and many patients probably remain undiagnosed. MATERIAL AND METHODS: The paper is based on four case reports and provides a short review of the condition. RESULTS: Our four patients all have typical facial features, such as a large nose and thin upper lip, thin hair and short curved fingers with characteristic radiological findings. The condition is autosomal dominant and caused by a mutation in the TRPS1 gene, which codes a gene-regulating protein involved in development of hair and modulation of chondrocytes. The diagnosis can be based on clinical findings, but DNA-analysis can be of help in unclear situations. Two of our patients were diagnosed from clinical and radiological findings, but for the two others genetic examinations were done as well. There is no causal treatment, but the diagnosis can give patients an explanation of their problems, and genetic counseling for the patient and family can be offered. Orthopedic surgery and cosmetic aids are valuable for many. INTERPRETATION: In an increasingly technified medical daily life, the clinical view is still the most important tool in diagnosing patients with this condition.


Subject(s)
Abnormalities, Multiple , Craniofacial Abnormalities , Hair Diseases , Hand Deformities, Congenital , Hypotrichosis , Langer-Giedion Syndrome , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Adolescent , Child , Child, Preschool , Craniofacial Abnormalities/diagnosis , Craniofacial Abnormalities/genetics , Female , Fingers/abnormalities , Genetic Counseling , Hair Diseases/diagnosis , Hair Diseases/genetics , Hand Deformities, Congenital/diagnosis , Hand Deformities, Congenital/genetics , Humans , Hypotrichosis/diagnosis , Hypotrichosis/genetics , Langer-Giedion Syndrome/diagnosis , Langer-Giedion Syndrome/genetics , Nose/abnormalities
18.
Eur J Hum Genet ; 28(8): 1078-1086, 2020 08.
Article in English | MEDLINE | ID: mdl-32203205

ABSTRACT

Rare sequence variants in the non-coding part of the BRCA genes are often reported as variants of uncertain significance (VUS), which leave patients and doctors in a challenging position. The aim of this study was to determine the pathogenicity of the BRCA1 c.5407-25T>A variant found in 20 families from Norway, France and United States with suspected hereditary breast and ovarian cancer. This was done by combining clinical and family information with allele frequency data, and assessment of the variant's effect on mRNA splicing. Mean age at breast (n = 12) and ovarian (n = 11) cancer diagnosis in female carriers was 49.9 and 60.4 years, respectively. The mean Manchester score in the 20 families was 16.4. The allele frequency of BRCA1 c.5407-25T>A was 1/64,566 in non-Finnish Europeans (gnomAD database v2.1.1). We found the variant in 1/400 anonymous Norwegian blood donors and 0/784 in-house exomes. Sequencing of patient-derived cDNA from blood, normal breast and ovarian tissue showed that BRCA1 c.5407-25T>A leads to skipping of exon 23, resulting in frameshift and protein truncation: p.(Gly1803GlnfsTer11). Western blot analysis of transiently expressed BRCA1 proteins in HeLa cells showed a reduced amount of the truncated protein compared with wild type. Noteworthily, we found that a small amount of full-length transcript was also generated from the c.5407-25T>A allele, potentially explaining the intermediate cancer burden in families carrying this variant. In summary, our results show that BRCA1 c.5407-25T>A leads to partial skipping of exon 23, and could represent a likely pathogenic variant with reduced penetrance.


Subject(s)
BRCA1 Protein/genetics , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Penetrance , Point Mutation , Adult , BRCA1 Protein/metabolism , Economics , Female , Gene Frequency , HeLa Cells , Hereditary Breast and Ovarian Cancer Syndrome/pathology , Heterozygote , Humans , Middle Aged , RNA Splicing
19.
Pract Lab Med ; 17: e00136, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31649988

ABSTRACT

BACKGROUND: Reported concentrations of serotonin in platelet-poor plasma (PPP) in healthy subjects vary widely due to different pre-analytical procedures. AIM: To examine how different pre-analytical conditions affect the measured concentration of serotonin in PPP. METHOD: Six pre-analytical protocols were compared for preparation of PPP from EDTA whole blood for quantification of serotonin from nine healthy individuals. Three combinations of centrifugation with a mild centrifugation of gel-free EDTA tubes followed by a stronger centrifugation were compared to single-stage centrifugation of EDTA tubes with separator gel and heat shock treatment of blood prior to centrifugation. All samples were analysed using the same enzyme linked immunosorbent assay (ELISA) method. RESULTS: Findings show that two consecutive centrifugations; first a mild centrifugation at 100 or 200×g followed by centrifugation at 4500 or 14500×g resulted in the lowest serotonin concentration in PPP. CONCLUSION: Two successive centrifugations to produce PPP for serotonin analysis; first a mild centrifugation to avoid mechanical stress on the platelets, and next a stronger centrifugation to remove platelets, is superior to the use of gel tubes and heat shock treatment.

SELECTION OF CITATIONS
SEARCH DETAIL