Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Publication year range
1.
Nature ; 592(7852): 93-98, 2021 04.
Article in English | MEDLINE | ID: mdl-33568816

ABSTRACT

Long non-coding RNAs (lncRNAs) can be important components in gene-regulatory networks1, but the exact nature and extent of their involvement in human Mendelian disease is largely unknown. Here we show that genetic ablation of a lncRNA locus on human chromosome 2 causes a severe congenital limb malformation. We identified homozygous 27-63-kilobase deletions located 300 kilobases upstream of the engrailed-1 gene (EN1) in patients with a complex limb malformation featuring mesomelic shortening, syndactyly and ventral nails (dorsal dimelia). Re-engineering of the human deletions in mice resulted in a complete loss of En1 expression in the limb and a double dorsal-limb phenotype that recapitulates the human disease phenotype. Genome-wide transcriptome analysis in the developing mouse limb revealed a four-exon-long non-coding transcript within the deleted region, which we named Maenli. Functional dissection of the Maenli locus showed that its transcriptional activity is required for limb-specific En1 activation in cis, thereby fine-tuning the gene-regulatory networks controlling dorso-ventral polarity in the developing limb bud. Its loss results in the En1-related dorsal ventral limb phenotype, a subset of the full En1-associated phenotype. Our findings demonstrate that mutations involving lncRNA loci can result in human Mendelian disease.


Subject(s)
Extremities , Homeodomain Proteins/genetics , Limb Deformities, Congenital/genetics , RNA, Long Noncoding/genetics , Sequence Deletion/genetics , Transcription, Genetic , Transcriptional Activation/genetics , Animals , Cell Line , Chromatin/genetics , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic
2.
J Clin Immunol ; 43(7): 1566-1580, 2023 10.
Article in English | MEDLINE | ID: mdl-37273120

ABSTRACT

Patients with autosomal dominant (AD) hyper-IgE syndrome (HIES) suffer from a constellation of manifestations including recurrent bacterial and fungal infections, severe atopy, and skeletal abnormalities. This condition is typically caused by monoallelic dominant-negative (DN) STAT3 variants. In 2020, we described 12 patients from eight kindreds with DN IL6ST variants resulting in a new form of AD HIES. These variants encoded truncated GP130 receptors, with intact extracellular and transmembrane domains, but lacking the intracellular recycling motif and the four STAT3-binding residues, resulting in an inability to recycle and activate STAT3. We report here two new DN variants of IL6ST in three unrelated families with HIES-AD. The biochemical and clinical impacts of these variants are different from those of the previously reported variants. The p.(Ser731Valfs*8) variant, identified in seven patients from two families, lacks the recycling motif and all the STAT3-binding residues, but its levels on the cell surface are only slightly increased and it underlies mild biological phenotypes with variable clinical expressivity. The p.(Arg768*) variant, identified in a single patient, lacks the recycling motif and the three most distal STAT3-binding residues. This variant accumulates at the cell surface and underlies severe biological and clinical phenotypes. The p.(Ser731Valfs*8) variant shows that a DN GP130 expressed at near normal levels on the cell surface can underlie heterogeneous clinical presentations, ranging from mild to severe. The p.(Arg768*) variant demonstrates that a truncated GP130 protein retaining one STAT3-binding residue can underlie severe HIES.


Subject(s)
Hypersensitivity, Immediate , Job Syndrome , Humans , Job Syndrome/diagnosis , Job Syndrome/genetics , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Phenotype , STAT3 Transcription Factor , Hypersensitivity, Immediate/complications , Mutation/genetics
3.
Hum Mutat ; 43(2): 266-282, 2022 02.
Article in English | MEDLINE | ID: mdl-34859529

ABSTRACT

De novo variants in QRICH1 (Glutamine-rich protein 1) has recently been reported in 11 individuals with intellectual disability (ID). The function of QRICH1 is largely unknown but it is likely to play a key role in the unfolded response of endoplasmic reticulum stress through transcriptional control of proteostasis. In this study, we present 27 additional individuals and delineate the clinical and molecular spectrum of the individuals (n = 38) with QRICH1 variants. The main clinical features were mild to moderate developmental delay/ID (71%), nonspecific facial dysmorphism (92%) and hypotonia (39%). Additional findings included poor weight gain (29%), short stature (29%), autism spectrum disorder (29%), seizures (24%) and scoliosis (18%). Minor structural brain abnormalities were reported in 52% of the individuals with brain imaging. Truncating or splice variants were found in 28 individuals and 10 had missense variants. Four variants were inherited from mildly affected parents. This study confirms that heterozygous QRICH1 variants cause a neurodevelopmental disorder including short stature and expands the phenotypic spectrum to include poor weight gain, scoliosis, hypotonia, minor structural brain anomalies, and seizures. Inherited variants from mildly affected parents are reported for the first time, suggesting variable expressivity.


Subject(s)
Autism Spectrum Disorder , Dwarfism , Intellectual Disability , Neurodevelopmental Disorders , Scoliosis , Autism Spectrum Disorder/genetics , Humans , Intellectual Disability/genetics , Muscle Hypotonia , Neurodevelopmental Disorders/genetics , Seizures , Weight Gain
4.
Am J Med Genet A ; 188(1): 319-325, 2022 01.
Article in English | MEDLINE | ID: mdl-34580982

ABSTRACT

Cranio-lenticulo-sutural dysplasia (CLSD; MIM 607812) is a rare or underdiagnosed condition, as only two families have been reported. The original family (Boyadjiev et al., Human Genetics, 2003, 113, 1-9 and Boyadjiev et al., Nature Genetics, 2006, 38, 1192-1197) showed recessive inheritance of the condition with a biallelic SEC23A missense variant in affected individuals. In contrast, another child with sporadic CLSD had a monoallelic SEC23A variant inherited from the reportedly unaffected father (Boyadjiev et al., Clinical Genetics, 2011, 80, 169-176), raising questions on possible digenism. Here, we report a 2-month-old boy seen because of large fontanels with wide cranial sutures, a large forehead, hypertelorism, a thin nose, a high arched palate, and micrognathia. His mother was clinically unremarkable, while his father had a history of large fontanels in infancy who had closed only around age 10 years; he also had a large forehead, hypertelorism, a thin, beaked nose and was operated for bilateral glaucoma with exfoliation of the lens capsule. Trio genome sequencing and familial segregation revealed a monoallelic c.1795G > A transition in SEC23A that was de novo in the father and transmitted to the proband. The variant predicts a nonconservative substitution (p.E599K) in an ultra-conserved residue that is seen in 3D models of yeast SEC23 to be involved in direct binding between SEC23 and SAR1 subunits of the coat protein complex II coat. This observation confirms the link between SEC23A variants and CLSD but suggests that in addition to the recessive inheritance described in the original family, SEC23A variants may result in dominant inheritance of CLSD, possibly by a dominant-negative disruptive effect on the SEC23 multimer.


Subject(s)
Mutation, Missense , Vesicular Transport Proteins , Base Sequence , Child , Humans , Infant , Male , Mutation, Missense/genetics , Vesicular Transport Proteins/genetics
5.
Clin Genet ; 99(6): 780-788, 2021 06.
Article in English | MEDLINE | ID: mdl-33586135

ABSTRACT

Four individuals from two families presented with a multisystemic condition of suspected genetic origin that was diagnosed only after genome analysis. The main phenotypic features were immune system dysregulation (severe immunodeficiency with autoimmunity) and intellectual disability. The four individuals were found to be homozygous for a 4.4 Kb deletion removing exons 20-23 (NM_003291.4) of the TPP2 gene, predicting a frameshift with premature termination of the protein. The deletion was located on a shared chromosome 13 haplotype indicating a Swiss founder mutation. Tripeptidyl peptidase 2 (TPP2) is a protease involved in HLA/antigen complex processing and amino acid homeostasis. Biallelic variants in TPP2 have been described in 10 individuals with variable features including immune deficiency, autoimmune cytopenias, and intellectual disability or chronic sterile brain inflammation mimicking multiple sclerosis. Our observations further delineate this severe condition not yet included in the OMIM catalog. Timely recognition of TPP2 deficiency is crucial since (1) immune surveillance is needed and hematopoietic stem cell transplantation may be necessary, and (2) for provision of genetic counselling. Additionally, enzyme replacement therapy, as already established for TPP1 deficiency, might be an option in the future.


Subject(s)
Aminopeptidases/genetics , Autoimmune Diseases/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Frameshift Mutation/genetics , Immunologic Deficiency Syndromes/genetics , Serine Endopeptidases/genetics , Adult , Child , Child, Preschool , Exons/genetics , Female , Humans , Male , Young Adult
6.
Am J Med Genet A ; 185(2): 517-527, 2021 02.
Article in English | MEDLINE | ID: mdl-33398909

ABSTRACT

Bone dysplasias (osteochondrodysplasias) are a large group of conditions associated with short stature, skeletal disproportion, and radiographic abnormalities of skeletal elements. Nearly all are genetic in origin. We report a series of seven children with similar findings of chondrodysplasia and growth failure following early hematopoietic stem cell transplantation (HSCT) for pediatric non-oncologic disease: hemophagocytic lymphohistiocytosis or HLH (five children, three with biallelic HLH-associated variants [in PRF1 and UNC13D] and one with HLH secondary to visceral Leishmaniasis), one child with severe combined immunodeficiency and one with Omenn syndrome (both children had biallelic RAG1 pathogenic variants). All children had normal growth and no sign of chondrodysplasia at birth and prior to their primary disease. After HSCT, all children developed growth failure, with standard deviation scores for height at or below -3. Radiographically, all children had changes in the spine, metaphyses and epiphyses, compatible with a spondyloepimetaphyseal dysplasia. Genomic sequencing failed to detect pathogenic variants in genes associated with osteochondrodysplasias. We propose that such chondrodysplasia with growth failure is a novel, rare, but clinically important complication following early HSCT for non-oncologic pediatric diseases. The pathogenesis is unknown but could possibly involve loss or perturbation of the cartilage-bone stem cell population.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Lymphohistiocytosis, Hemophagocytic/genetics , Osteochondrodysplasias/genetics , Child , Child, Preschool , Female , Humans , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/therapy , Male , Membrane Proteins/genetics , Osteochondrodysplasias/complications , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/therapy , Perforin/genetics , Treatment Outcome
7.
BMC Endocr Disord ; 21(1): 172, 2021 Aug 21.
Article in English | MEDLINE | ID: mdl-34419042

ABSTRACT

BACKGROUND: The Mauriac syndrome was described in 1930 as a peculiar combination of poorly controlled diabetes mellitus type 1, stunted growth and glycogenic hepatopathy. More recently, lactic acidosis was recognized as an additional feature, often induced by insulin treatment. CASE PRESENTATION: A 17-year old girl known for diabetes type 1A and Mauriac syndrome was admitted to the emergency room with hyperglycemia of > 41 mmol/l without ketoacidosis. Under a standard insulin regimen, hyperglycemia was rapidly corrected but marked hyperlactatemia occurred. CONCLUSIONS: The mechanism of impaired glucose utilization and lactate elevation independent of ketoacidosis in Mauriac syndrome is intriguing. The rarity of Mauriac syndrome and its resemblance to glycogen storage diseases suggest the presence of a specific metabolic or genetic predisposition that remains to be identified.


Subject(s)
Diabetes Complications/pathology , Diabetes Mellitus, Type 1/complications , Hepatomegaly/pathology , Hyperlactatemia/pathology , Lactates/metabolism , Adolescent , Diabetes Complications/etiology , Diabetes Complications/metabolism , Female , Hepatomegaly/etiology , Humans , Hyperlactatemia/etiology , Hyperlactatemia/metabolism , Prognosis
8.
N Engl J Med ; 374(26): 2553-2562, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27355534

ABSTRACT

BACKGROUND: Cortical-bone fragility is a common feature in osteoporosis that is linked to nonvertebral fractures. Regulation of cortical-bone homeostasis has proved elusive. The study of genetic disorders of the skeleton can yield insights that fuel experimental therapeutic approaches to the treatment of rare disorders and common skeletal ailments. METHODS: We evaluated four patients with Pyle's disease, a genetic disorder that is characterized by cortical-bone thinning, limb deformity, and fractures; two patients were examined by means of exome sequencing, and two were examined by means of Sanger sequencing. After a candidate gene was identified, we generated a knockout mouse model that manifested the phenotype and studied the mechanisms responsible for altered bone architecture. RESULTS: In all affected patients, we found biallelic truncating mutations in SFRP4, the gene encoding secreted frizzled-related protein 4, a soluble Wnt inhibitor. Mice deficient in Sfrp4, like persons with Pyle's disease, have increased amounts of trabecular bone and unusually thin cortical bone, as a result of differential regulation of Wnt and bone morphogenetic protein (BMP) signaling in these two bone compartments. Treatment of Sfrp4-deficient mice with a soluble Bmp2 receptor (RAP-661) or with antibodies to sclerostin corrected the cortical-bone defect. CONCLUSIONS: Our study showed that Pyle's disease was caused by a deficiency of sFRP4, that cortical-bone and trabecular-bone homeostasis were governed by different mechanisms, and that sFRP4-mediated cross-regulation between Wnt and BMP signaling was critical for achieving proper cortical-bone thickness and stability. (Funded by the Swiss National Foundation and the National Institutes of Health.).


Subject(s)
Bone Density/genetics , Bone Remodeling/genetics , Osteochondrodysplasias/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Adolescent , Animals , Biomarkers/blood , Bone Morphogenetic Proteins/metabolism , Bone Remodeling/physiology , Bone and Bones/pathology , Bone and Bones/physiology , Child, Preschool , Disease Models, Animal , Female , Gene Deletion , Homeostasis , Humans , Male , Mice , Mice, Knockout , Middle Aged , Osteochondrodysplasias/physiopathology , Sequence Analysis, DNA , Signal Transduction , Wnt Proteins/metabolism
9.
Genet Med ; 21(12): 2734-2743, 2019 12.
Article in English | MEDLINE | ID: mdl-31263216

ABSTRACT

PURPOSE: We observed four individuals in two unrelated but consanguineous families from Portugal and Brazil affected by early-onset retinal degeneration, sensorineural hearing loss, microcephaly, intellectual disability, and skeletal dysplasia with scoliosis and short stature. The phenotype precisely matched that of an individual of Azorean descent published in 1986 by Liberfarb and coworkers. METHODS: Patients underwent specialized clinical examinations (including ophthalmological, audiological, orthopedic, radiological, and developmental assessment). Exome and targeted sequencing was performed on selected individuals. Minigene constructs were assessed by quantitative polymerase chain reaction (qPCR) and Sanger sequencing. RESULTS: Affected individuals shared a 3.36-Mb region of autozygosity on chromosome 22q12.2, including a 10-bp deletion (NM_014338.3:c.904-12_904-3delCTATCACCAC), immediately upstream of the last exon of the PISD (phosphatidylserine decarboxylase) gene. Sequencing of PISD from paraffin-embedded tissue from the 1986 case revealed the identical homozygous variant. In HEK293T cells, this variant led to aberrant splicing of PISD transcripts. CONCLUSION: We have identified the genetic etiology of the Liberfarb syndrome, affecting brain, eye, ear, bone, and connective tissue. Our work documents the migration of a rare Portuguese founder variant to two continents and highlights the link between phospholipid metabolism and bone formation, sensory defects, and cerebral development, while raising the possibility of therapeutic phospholipid replacement.


Subject(s)
Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Adolescent , Adult , Brazil , Exome/genetics , Female , Genotype , HEK293 Cells , Hearing Loss, Sensorineural/genetics , Humans , Intellectual Disability/genetics , Male , Microcephaly/genetics , Musculoskeletal Abnormalities/genetics , Osteochondrodysplasias/genetics , Pedigree , Phenotype , Portugal , Retinal Degeneration/genetics , Syndrome , Young Adult
10.
J Hum Genet ; 64(3): 261-264, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30531807

ABSTRACT

Spondylocostal dysostosis (SCDO) is a heterogeneous group of skeletal disorders characterized by multiple segmentation defects involving vertebrae and ribs. Seven disease genes have been reported as causal genes for SCDO: DLL3, MESP2, TBX6, HES7, RIPPLY2, DMRT2, and LFNG. Here we report a Japanese SCDO case with multiple severe vertebral anomalies from cervical to sacral spine. The patient was a compound heterozygote for c.372delG (p.K124Nfs*) and c.601G>A (p.D201N) variants of LFNG, which encodes a glycosyltransferase (O-fucosylpeptide 3-beta-N-acetylglucosaminyltransferase). The missense variant was in the DxD motif, an active-site motif of the glycosyltransferase, and its loss of the enzyme function was confirmed by an in vitro enzyme assay. This is the second report of LFNG mutations in SCDO.


Subject(s)
Abnormalities, Multiple/genetics , Glycosyltransferases/genetics , Hernia, Diaphragmatic/genetics , Hexosyltransferases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mutation , Abnormalities, Multiple/pathology , Amino Acid Sequence , Glucosyltransferases , Hernia, Diaphragmatic/pathology , Humans , Infant , Male , Prognosis , Sequence Homology
11.
J Bone Miner Metab ; 37(2): 378-383, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29948344

ABSTRACT

Data from exome sequencing show that a proportion of individuals in whom a genetic disorder is suspected turn out to have not one, but two to four distinct ones. This may require an evolution in our diagnostic attitude towards individuals with complex disorders. We report a patient with splenomegaly, pneumopathy, bone changes and fronto-temporal dementia (FTD). "Sea-blue histiocytes" in his bone marrow pointed to a lysosomal storage disease. Homozygosity for a pathogenic mutation in the SMPD1 gene confirmed Niemann-Pick disease type B (NPD-B). Mild cognitive impairment and abnormal brain FDG PET were consistent with FTD. We initially tried to fit the skeletal and neurologic phenotype into the NPD-B diagnosis. However, additional studies revealed a pathogenic mutation in the SQSTM1 gene. Thus, our patient had two distinct diseases; NPD-B, and Paget's disease of bone with FTD. The subsequent finding of a mutation in SQSTM1 gene ended our struggle to explain the combination of findings by a singular "unifying" diagnosis and allowed us to make specific therapeutic decisions. SQSTM1 mutations have been reported in association with FTD, possibly because of defective autophagy. Bisphosphonates may be beneficial for PDB, but since they are known to inhibit acid sphingomyelinase activity, we refrained from using them in this patient. While the principle of looking for unifying diagnosis remains valid, physicians should consider the possibility of co-existing multiple diagnoses when clinical features are difficult to explain by a single one. Accurate diagnostic work-up can guide genetic counseling but also lead to better medical management.


Subject(s)
Bone and Bones/pathology , Frontotemporal Dementia/complications , Hepatomegaly/complications , Niemann-Pick Disease, Type B/complications , Osteitis Deformans/complications , Sequestosome-1 Protein/genetics , Splenomegaly/complications , Bone Marrow/pathology , Humans , Male , Middle Aged , Niemann-Pick Disease, Type B/diagnostic imaging , Osteitis Deformans/diagnostic imaging , Tomography, X-Ray Computed
12.
Rheumatol Int ; 39(3): 441-452, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30327864

ABSTRACT

Progressive pseudorheumatoid dysplasia (PPRD) is a genetic bone disorder characterised by the progressive degeneration of articular cartilage that leads to pain, stiffness and joint enlargement. As PPRD is a rare disease, available literature is mainly represented by single case reports and only a few larger case series. Our aim is to review the literature concerning clinical, laboratory and radiological features of PPRD. PPRD is due to a mutation in Wnt1-inducible signalling protein 3 (WISP3) gene, which encodes a signalling factor involved in cartilage homeostasis. The disease onset in childhood and skeletal changes progresses over time leading to significant disability. PPRD is a rare condition that should be suspected if a child develops symmetrical polyarticular involvement without systemic inflammation, knobbly interphalangeal joints of the hands, and gait abnormalities. A full skeletal survey, or at least a lateral radiograph of the spine, can direct towards a correct diagnosis that can be confirmed molecularly. More than 70 WISP3 mutations have so far been reported. Genetic testing should start with the study of genomic DNA extracted from blood leucocytes, but intronic mutations in WISP3 causing splicing aberrations can only be detected by analysing WISP3 mRNA, which can be extracted from cultured skin fibroblasts. A skin biopsy is, therefore, indicated in patients with typical PPRD findings and negative mutation screening of genomic DNA.


Subject(s)
Joint Diseases/congenital , Alternative Splicing , CCN Intercellular Signaling Proteins/genetics , Child , Child, Preschool , Humans , Introns , Joint Diseases/diagnostic imaging , Joint Diseases/genetics , Joint Diseases/physiopathology , Mutation , RNA, Messenger/metabolism , Radiography , Sequence Analysis, DNA , Sequence Analysis, RNA , Skin/cytology
14.
Am J Hum Genet ; 92(6): 990-5, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23684011

ABSTRACT

Kenny-Caffey syndrome (KCS) and the similar but more severe osteocraniostenosis (OCS) are genetic conditions characterized by impaired skeletal development with small and dense bones, short stature, and primary hypoparathyroidism with hypocalcemia. We studied five individuals with KCS and five with OCS and found that all of them had heterozygous mutations in FAM111A. One mutation was identified in four unrelated individuals with KCS, and another one was identified in two unrelated individuals with OCS; all occurred de novo. Thus, OCS and KCS are allelic disorders of different severity. FAM111A codes for a 611 amino acid protein with homology to trypsin-like peptidases. Although FAM111A has been found to bind to the large T-antigen of SV40 and restrict viral replication, its native function is unknown. Molecular modeling of FAM111A shows that residues affected by KCS and OCS mutations do not map close to the active site but are clustered on a segment of the protein and are at, or close to, its outer surface, suggesting that the pathogenesis involves the interaction with as yet unidentified partner proteins rather than impaired catalysis. FAM111A appears to be crucial to a pathway that governs parathyroid hormone production, calcium homeostasis, and skeletal development and growth.


Subject(s)
Abnormalities, Multiple/genetics , Bone Diseases, Developmental/genetics , Craniofacial Abnormalities/genetics , Dwarfism/genetics , Hyperostosis, Cortical, Congenital/genetics , Hypocalcemia/genetics , Hypoparathyroidism/genetics , Receptors, Virus/genetics , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/mortality , Abnormalities, Multiple/pathology , Adolescent , Adult , Bone Diseases, Developmental/mortality , Bone Diseases, Developmental/pathology , Child , Craniofacial Abnormalities/mortality , Craniofacial Abnormalities/pathology , Dwarfism/diagnostic imaging , Dwarfism/mortality , Genetic Association Studies , Heterozygote , Humans , Hyperostosis, Cortical, Congenital/diagnostic imaging , Hyperostosis, Cortical, Congenital/mortality , Hypocalcemia/diagnostic imaging , Hypocalcemia/mortality , Hypoparathyroidism/diagnostic imaging , Hypoparathyroidism/mortality , Infant , Infant, Newborn , Male , Mutation, Missense , Parathyroid Hormone/deficiency , Radiography
17.
Am J Med Genet A ; 167A(12): 2902-12, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26286438

ABSTRACT

We report two unrelated patients with a multisystem disease involving liver, eye, immune system, connective tissue, and bone, caused by biallelic mutations in the neuroblastoma amplified sequence (NBAS) gene. Both presented as infants with recurrent episodes triggered by fever with vomiting, dehydration, and elevated transaminases. They had frequent infections, hypogammaglobulinemia, reduced natural killer cells, and the Pelger-Huët anomaly of their granulocytes. Their facial features were similar with a pointed chin and proptosis; loose skin and reduced subcutaneous fat gave them a progeroid appearance. Skeletal features included short stature, slender bones, epiphyseal dysplasia with multiple phalangeal pseudo-epiphyses, and small C1-C2 vertebrae causing cervical instability and myelopathy. Retinal dystrophy and optic atrophy were present in one patient. NBAS is a component of the synthaxin-18 complex and is involved in nonsense-mediated mRNA decay control. Putative loss-of-function mutations in NBAS are already known to cause disease in humans. A specific founder mutation has been associated with short stature, optic nerve atrophy and Pelger-Huët anomaly of granulocytes (SOPH) in the Siberian Yakut population. A more recent report associates NBAS mutations with recurrent acute liver failure in infancy in a group of patients of European descent. Our observations indicate that the phenotypic spectrum of NBAS deficiency is wider than previously known and includes skeletal, hepatic, metabolic, and immunologic aspects. Early recognition of the skeletal phenotype is important for preventive management of cervical instability.


Subject(s)
Abnormalities, Multiple/genetics , Mutation , Neoplasm Proteins/genetics , Abnormalities, Multiple/etiology , Child , Child, Preschool , Female , Humans , Immune System/physiopathology , Infant , Liver Diseases/genetics , Male , Optic Atrophy/genetics , Pelger-Huet Anomaly/etiology , Pregnancy , Retina/pathology , Skin/pathology
18.
Am J Med Genet A ; 167(7): 1501-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25808063

ABSTRACT

Cerebral, ocular, dental, auricular, skeletal anomalies (CODAS) syndrome (MIM 600373) was first described and named by Shehib et al, in 1991 in a single patient. The anomalies referred to in the acronym are as follows: cerebral-developmental delay, ocular-cataracts, dental-aberrant cusp morphology and delayed eruption, auricular-malformations of the external ear, and skeletal-spondyloepiphyseal dysplasia. This distinctive constellation of anatomical findings should allow easy recognition but despite this only four apparently sporadic patients have been reported in the last 20 years indicating that the full phenotype is indeed very rare with perhaps milder or a typical presentations that are allelic but without sufficient phenotypic resemblance to permit clinical diagnosis. We performed exome sequencing in three patients (an isolated case and a brother and sister sib pair) with classical features of CODAS. Sanger sequencing was used to confirm results as well as for mutation discovery in a further four unrelated patients ascertained via their skeletal features. Compound heterozygous or homozygous mutations in LONP1 were found in all (8 separate mutations; 6 missense, 1 nonsense, 1 small in-frame deletion) thus establishing the genetic basis of CODAS and the pattern of inheritance (autosomal recessive). LONP1 encodes an enzyme of bacterial ancestry that participates in protein turnover within the mitochondrial matrix. The mutations cluster at the ATP-binding and proteolytic domains of the enzyme. Biallelic inheritance and clustering of mutations confirm dysfunction of LONP1 activity as the molecular basis of CODAS but the pathogenesis remains to be explored.


Subject(s)
ATP-Dependent Proteases/genetics , Craniofacial Abnormalities/genetics , Exome/genetics , Eye Abnormalities/genetics , Growth Disorders/genetics , Hip Dislocation, Congenital/genetics , Mitochondrial Proteins/genetics , Models, Genetic , Mutation/genetics , Osteochondrodysplasias/genetics , Tooth Abnormalities/genetics , Base Sequence , Genes, Recessive/genetics , Humans , Molecular Sequence Data , Sequence Analysis, DNA , Switzerland
19.
Am J Med Genet A ; 164A(10): 2667-71, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25099136

ABSTRACT

Camurati-Engelmann disease is characterized by hyperostosis of the long bones and the skull, muscle atrophy, severe limb pain, and progressive joint contractures in some patients. It is caused by heterozygous mutations in the transforming growth factor ß1 (TGFß1) believed to result in improper folding of the latency-associated peptide domain of TGFß1 and thus in increased or deregulated bioactivity. Losartan, an angiotensin II type 1 receptor antagonist, has been found to downregulate the expression of TGFß type 1 and 2 receptors. Clinical trials with losartan have shown a benefit in Marfan syndrome, while trials are underway for Duchenne muscular dystrophy and other myopathies associated with TGFß1 signaling. We hypothesized that due to its anti-TGFß1 activity, losartan might be beneficial in Camurati-Engelmann disease. This report concerns a boy who presented at age 13 years with severe limb pain and difficulty in walking. Clinical and radiographic evaluation results were compatible with Camurati-Engelmann disease and the diagnosis was confirmed by mutation analysis (c.652C > T [p.Arg218Cys]). The boy underwent an experimental treatment with losartan at a dosage of 50 mg/day, orally. During the treatment period of 18 months, the intensity and frequency of limb pain decreased significantly (as shown by a pain diary), and muscle strength improved, allowing the boy to resume walking and climbing stairs. No obvious side effects were observed. We cautiously conclude that TGFß1 inhibition with losartan deserves further evaluation in the clinical management of Camurati-Engelmann disease.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Camurati-Engelmann Syndrome/diagnosis , Camurati-Engelmann Syndrome/drug therapy , Losartan/therapeutic use , Adolescent , Camurati-Engelmann Syndrome/genetics , Humans , Male , Mutation/genetics
20.
PLoS Genet ; 7(4): e1002050, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21533187

ABSTRACT

Metachondromatosis (MC) is a rare, autosomal dominant, incompletely penetrant combined exostosis and enchondromatosis tumor syndrome. MC is clinically distinct from other multiple exostosis or multiple enchondromatosis syndromes and is unlinked to EXT1 and EXT2, the genes responsible for autosomal dominant multiple osteochondromas (MO). To identify a gene for MC, we performed linkage analysis with high-density SNP arrays in a single family, used a targeted array to capture exons and promoter sequences from the linked interval in 16 participants from 11 MC families, and sequenced the captured DNA using high-throughput parallel sequencing technologies. DNA capture and parallel sequencing identified heterozygous putative loss-of-function mutations in PTPN11 in 4 of the 11 families. Sanger sequence analysis of PTPN11 coding regions in a total of 17 MC families identified mutations in 10 of them (5 frameshift, 2 nonsense, and 3 splice-site mutations). Copy number analysis of sequencing reads from a second targeted capture that included the entire PTPN11 gene identified an additional family with a 15 kb deletion spanning exon 7 of PTPN11. Microdissected MC lesions from two patients with PTPN11 mutations demonstrated loss-of-heterozygosity for the wild-type allele. We next sequenced PTPN11 in DNA samples from 54 patients with the multiple enchondromatosis disorders Ollier disease or Maffucci syndrome, but found no coding sequence PTPN11 mutations. We conclude that heterozygous loss-of-function mutations in PTPN11 are a frequent cause of MC, that lesions in patients with MC appear to arise following a "second hit," that MC may be locus heterogeneous since 1 familial and 5 sporadically occurring cases lacked obvious disease-causing PTPN11 mutations, and that PTPN11 mutations are not a common cause of Ollier disease or Maffucci syndrome.


Subject(s)
Enchondromatosis/genetics , Exostoses, Multiple Hereditary/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Chromosomes, Human/genetics , DNA Copy Number Variations , Enchondromatosis/pathology , Exons , Gene Deletion , Genetic Linkage , High-Throughput Nucleotide Sequencing , Humans , Loss of Heterozygosity , Mutation , Pedigree , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL