Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Osteoarthritis Cartilage ; 29(10): 1474-1484, 2021 10.
Article in English | MEDLINE | ID: mdl-34166809

ABSTRACT

OBJECTIVE: Growth of mandibular condylar cartilage (MCC) is associated with cell proliferation within the polymorphic cell layer and subsequent differentiation into chondrocytes that reside along the condylar surface and along the cartilage/subchondral bone interface. We examined whether cells in the polymorphic layer would proliferate and repopulate toxin-induced cell-depleted areas in MCCs of adult mice. METHOD: We induced diphtheria toxin (DTA) expression (ROSA26l-s-lDTA) to cell-autonomously kill large fractions of MCC chondrocytes throughout the cartilage or along the articular cartilage surface with Aggrecan-CreERt2 (AcanCreERt2) or Lubricin-CreERt2 (Prg4CreERt2) Cre-recombinase-inducible mice, respectively. We examined MCCs from these mice shortly after cell killing or several months later with histology and confocal microscopy for evidence of chondrocyte proliferation and repopulation. RESULTS: AcanCreERt2-induced DTA expression killed an average of 53% MCC chondrocytes in adult mice after 1 week (39-66%, 95% confidence interval (CI)). Twelve weeks later, surviving chondrocytes had proliferated but not migrated to cell depleted areas. Prg4CreERt2-induced DTA expression killed an average of 24% surface chondrocytes in mice after 5 weeks (14-34% CI). After thirteen weeks there was 34% fewer surface chondrocytes (4-63% CI) in Prg4CreERt2 DTA-induced mice compared to controls. CONCLUSION: In adult mice, after diphtheria toxin-mediated chondrocyte killing, cell depleted areas within MCC cartilage are not repopulated by new cells.


Subject(s)
Cartilage, Articular/pathology , Chondrocytes/pathology , Diphtheria Toxin/toxicity , Mandibular Condyle/pathology , Animals , Apoptosis , Cell Proliferation , Mice, Inbred C57BL , Microscopy, Confocal
2.
Clin Genet ; 93(5): 1075-1080, 2018 05.
Article in English | MEDLINE | ID: mdl-29231959

ABSTRACT

Congenital lipomatous overgrowth with vascular, epidermal, and skeletal (CLOVES) anomalies and Klippel-Trenaunay (KTS) syndromes are caused by somatic gain-of-function mutations in PIK3CA, encoding a catalytic subunit of phosphoinositide 3-kinase. Affected tissue is needed to find mutations, as mutant alleles are not detectable in blood. Because some patients with CLOVES develop Wilms tumor, we tested urine as a source of DNA for mutation detection. We extracted DNA from the urine of 17 and 24 individuals with CLOVES and KTS, respectively, and screened 5 common PIK3CA mutation hotspots using droplet digital polymerase chain reaction. Six of 17 CLOVES participants (35%) had mutant PIK3CA alleles in urine. Among 8 individuals in whom a mutation had been previously identified in affected tissue, 4 had the same mutant allele in the urine. One study participant with CLOVES had been treated for Wilms tumor. We detected the same PIK3CA mutation in her affected tissue, urine, and tumor, indicating Wilms tumors probably arise from PIK3CA mutant cells in patients with CLOVES. No urine sample from a participant with KTS had detectable PIK3CA mutations. We suggest that urine, which has the advantage of being collected non-invasively, is useful when searching for mutations in individuals with CLOVES syndrome.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Klippel-Trenaunay-Weber Syndrome/genetics , Lipoma/genetics , Musculoskeletal Abnormalities/genetics , Nevus/genetics , Vascular Malformations/genetics , Wilms Tumor/genetics , Adolescent , Adult , Alleles , Child , Child, Preschool , DNA/genetics , DNA/urine , Female , Genetic Predisposition to Disease , Humans , Infant , Klippel-Trenaunay-Weber Syndrome/pathology , Klippel-Trenaunay-Weber Syndrome/urine , Lipoma/pathology , Lipoma/urine , Male , Middle Aged , Musculoskeletal Abnormalities/pathology , Musculoskeletal Abnormalities/urine , Mutation , Nevus/pathology , Nevus/urine , Phenotype , Vascular Malformations/pathology , Vascular Malformations/urine , Wilms Tumor/pathology , Wilms Tumor/urine
3.
Bone ; 105: 253-261, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28942122

ABSTRACT

Dominant negative mutations in CLCN7, which encodes a homodimeric chloride channel needed for matrix acidification by osteoclasts, cause Albers-Schönberg disease (also known as autosomal dominant osteopetrosis type 2). More than 25 different CLCN7 mutations have been identified in patients affected with Albers-Schönberg disease, but only one mutation (Clcn7G213R) has been introduced in mice to create an animal model of this disease. Here we describe a mouse with a different osteopetrosis-causing mutation (Clcn7F318L). Compared to Clcn7+/+ mice, 12-week-old Clcn7F318L/+ mice have significantly increased trabecular bone volume, consistent with Clcn7F318L acting as a dominant negative mutation. Clcn7F318L/F318L and Clcn7F318L/G213R mice die by 1month of age and resemble Clcn7 knockout mice, which indicate that p.F318L mutant protein is non-functional and p.F318L and p.G213R mutant proteins do not complement one another. Since it has been reported that treatment with interferon gamma (IFN-G) improves bone properties in Clcn7G213R/+ mice, we treated Clcn7F318L/+ mice with IFN-G and observed a decrease in osteoclast number and mineral apposition rate, but no overall improvement in bone properties. Our results suggest that the benefits of IFN-G therapy in patients with Albers-Schönberg disease may be mutation-specific.


Subject(s)
Alleles , Chloride Channels/genetics , Osteopetrosis/pathology , Animals , Bone and Bones/pathology , Cancellous Bone/pathology , Cell Count , Chloride Channels/metabolism , Disease Models, Animal , Gene Knock-In Techniques , Genes, Dominant , Heterozygote , Homozygote , Interferon-gamma/therapeutic use , Loss of Function Mutation/genetics , Mice , Organ Size , Osteoclasts/metabolism , Osteoclasts/pathology , Phenotype
4.
Sci Rep ; 7: 43848, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28266618

ABSTRACT

Mice are commonly used to study the temporomandibular joint (TMJ) and to model human TMJ disease. However, evaluating TMJ pathology in mice using standard histologic methods is time consuming, labor intensive, and dependent upon investigators' expertise at consistently orienting and sectioning across tiny specimens. We describe a method that uses confocal microscopy to rapidly and reliably assess indicators of mandibular condyle cartilage pathology in mice. We demonstrate the utility of this method for detecting abnormalities in chondrocyte distribution in mice lacking lubricin (Prg4), the major boundary lubricant of articular cartilage. We further show that the method can provide information about recombination sites and efficiency in mandibular cartilage for Cre-driver strains. Because specimen preparation and data acquisition with confocal microscopy are simple and fast, the method can serve as a primary screening tool for TMJ pathology, before proceeding to complicated, time consuming, secondary analyses.


Subject(s)
Cartilage, Articular/pathology , Chondrocytes/pathology , Mandibular Condyle/pathology , Microscopy, Confocal/methods , Animals , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Mandibular Condyle/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proteoglycans/genetics , Proteoglycans/metabolism , Reproducibility of Results , Temporomandibular Joint/metabolism , Temporomandibular Joint/pathology , Temporomandibular Joint Disorders/diagnosis , Temporomandibular Joint Disorders/genetics , Temporomandibular Joint Disorders/metabolism
6.
Proc Natl Acad Sci U S A ; 104(15): 6194-9, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17404241

ABSTRACT

Synovial fluid is a semidilute hyaluronate (HA) polymer solution, the rheology of which depends on HA-protein interactions, and lubricin is a HA-binding protein found in synovial fluid and at cartilage surfaces, where it contributes to boundary lubrication under load. Individuals with genetic deficiency of lubricin develop precocious joint failure. The role of lubricin in synovial fluid rheology is not known. We used a multiple-particle-tracking microrheology technique to study the molecular interactions between lubricin and HA in synovial fluid. Particles (200 nm mean diameter) embedded in normal and lubricin-deficient synovial fluid samples were tracked separately by using multiple-particle-tracking microrheology. The time-dependent ensemble-averaged mean-squared displacements of all of the particles were measured over a range of physiologically relevant frequencies. The mean-squared displacement correlation with time lag had slopes with values of unity for simple HA solutions and for synovial fluid from an individual who genetically lacked lubricin, in contrast to slopes with values less than unity (alpha approximately 0.6) for normal synovial fluid. These data correlated with bulk rheology studies of the same samples. We found that the subdiffusive and elastic behavior of synovial fluid, at physiological shear rates, was absent in fluid from a patient who lacks lubricin. We conclude that lubricin provides synovial fluid with an ability to dissipate strain energy induced by mammalian locomotion, which is a chondroprotective feature that is distinct from boundary lubrication.


Subject(s)
Glycoproteins/chemistry , Hyaluronic Acid/chemistry , Rheology/methods , Synovial Fluid/chemistry , Animals , Biomechanical Phenomena , Biophysical Phenomena , Biophysics , Cattle , Glycerol , Glycoproteins/genetics , Humans , Microscopy, Fluorescence , Microspheres , Mutation/genetics
7.
Arthritis Res Ther ; 8(1): R25, 2006.
Article in English | MEDLINE | ID: mdl-16507126

ABSTRACT

In order to determine whether there is a genetic component to hip or knee joint failure due to idiopathic osteoarthritis (OA), we invited patients (probands) undergoing hip or knee arthroplasty for management of idiopathic OA to provide detailed family histories regarding the prevalence of idiopathic OA requiring joint replacement in their siblings. We also invited their spouses to provide detailed family histories about their siblings to serve as a control group. In the probands, we confirmed the diagnosis of idiopathic OA using American College of Rheumatology criteria. The cohorts included the siblings of 635 probands undergoing total hip replacement, the siblings of 486 probands undergoing total knee replacement, and the siblings of 787 spouses. We compared the prevalence of arthroplasty for idiopathic OA among the siblings of the probands with that among the siblings of the spouses, and we used logistic regression to identify independent risk factors for hip and knee arthroplasty in the siblings. Familial aggregation for hip arthroplasty, but not for knee arthroplasty, was observed after controlling for age and sex, suggesting a genetic contribution to end-stage hip OA but not to end-stage knee OA. We conclude that attempts to identify genes that predispose to idiopathic OA resulting in joint failure are more likely to be successful in patients with hip OA than in those with knee OA.


Subject(s)
Arthroplasty, Replacement, Hip , Osteoarthritis, Hip/genetics , Osteoarthritis, Hip/surgery , Aged , Aged, 80 and over , Arthroplasty, Replacement, Hip/statistics & numerical data , Arthroplasty, Replacement, Knee/statistics & numerical data , Body Mass Index , Cohort Studies , Female , Humans , Male , Middle Aged , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/surgery , Risk Factors , Spouses
8.
Osteoarthritis Cartilage ; 13(6): 497-507, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15922184

ABSTRACT

OBJECTIVE: We sought to determine whether sequence variations in cartilage collagen genes are associated with primary, early-onset osteoarthritis (OA). METHODS: The cartilage collagen genes, COL2A1, COL9A1, COL9A2, COL9A3, COL11A1 and COL11A2, were screened for sequence variations in 72 Finnish probands and one US family with primary early-onset hip and/or knee OA. In addition, allelic association studies were performed using six to 12 common polymorphisms from each gene by genotyping 72 OA patients and 103 controls. RESULTS: Altogether 239 sequence variations were found, of which 16 were not present in the controls. Seven of the unique variations, four in COL11A1, two in COL11A2 and one in COL2A1, were studied further, because they resulted in the substitution of conserved amino acids or were predicted to affect mRNA splicing. Co-segregation of a sequence variation and the phenotype was found in all four families available for study. Association analysis failed to identify any common predisposing alleles. CONCLUSIONS: Early-onset OA demonstrates locus and allelic heterogeneity since the identified variations were in three different collagen genes and each of the six probands had a different mutation. It is also possible that some OA cases represent the mild end of the chondrodysplasia phenotypic spectrum. The major susceptibility alleles in this form of OA, however, remain to be identified.


Subject(s)
Collagen/genetics , Mutation/genetics , Osteoarthritis/genetics , Adult , Aged , Cartilage, Articular/physiology , Collagen Type II/genetics , Collagen Type IX/genetics , Collagen Type XI/genetics , Female , Humans , Male , Middle Aged , Pedigree , Polymerase Chain Reaction , Polymorphism, Genetic , Sequence Analysis, RNA
9.
Arthritis Rheum ; 52(6): 1746-55, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15934070

ABSTRACT

OBJECTIVE: To study the relationship between the boundary-lubricating ability of synovial fluid (SF) and articular cartilage damage in a rabbit knee injury model, to correlate collagen markers of such damage with SF boundary-lubricating ability and elastase activity, and to examine the lubricating ability of SF, together with collagen markers of articular cartilage damage, under the inflammatory conditions of knee joint synovitis (KJS) and rheumatoid arthritis (RA). METHODS: SF was aspirated weekly from the affected knee joints of 10 adult rabbits following transection of the anterior and posterior cruciate ligaments. The boundary-lubricating ability of SF was determined in vitro using a previously described friction apparatus. Lubricin concentrations and type II collagen (CII) peptides were quantified by sandwich enzyme-linked immunosorbent assays (ELISAs). Levels of the C-terminal neoepitope 9A4 (derived from collagenase degradation of CI, CII, and CIII) and of epitope 5-D-4 of keratan sulfate (a marker of proteoglycan depletion) were quantified by inhibition ELISAs. Elastase activity was measured spectrophotometrically. The sensitivity of purified human lubricin to digestion by neutrophil elastase (NE) was examined by Western blotting. RESULTS: The lubricating ability of SF from injured rabbit knees was significantly decreased at weeks 2 and 3 compared with week 1 after injury. Lubricin concentrations were significantly higher at week 1 than at weeks 2 and 3. CII peptide concentrations increased significantly at weeks 2 and 3 compared with week 1, while 9A4 neoepitope concentrations increased significantly at week 3 compared with weeks 1 and 2. There were no significant differences in epitope 5-D-4 concentrations among the 3 weeks. Elastase activity in SF increased significantly at weeks 2 and 3 compared with week 1. Elastase activity correlated significantly with diminishing lubrication at weeks 1, 2, and 3. SF from patients with KJS or RA exhibited deficient lubrication and elevated levels of CII peptides compared with SF from normal controls. NE was shown to completely degrade purified human lubricin in vitro. CONCLUSION: Loss of boundary-lubricating ability of SF after injury is associated with damage to the articular cartilage matrix. This can be attributed to inflammatory processes resulting from the injury, particularly in the early phases. This association also exists in patients with acute knee injuries or progressive chronic inflammatory arthritis.


Subject(s)
Arthritis/physiopathology , Cartilage, Articular/physiopathology , Knee Injuries/physiopathology , Animals , Anterior Cruciate Ligament Injuries , Arthritis/etiology , Collagen Type II/analysis , Glycoproteins/analysis , Humans , Knee Injuries/complications , Knee Joint , Models, Animal , Pancreatic Elastase/analysis , Rabbits , Synovial Fluid/chemistry , Synovial Fluid/physiology , Synovitis/etiology , Synovitis/physiopathology
10.
Neurology ; 61(2): 226-30, 2003 Jul 22.
Article in English | MEDLINE | ID: mdl-12874403

ABSTRACT

OBJECTIVE: To define the clinical and biochemical abnormalities of an autosomal dominant form of acute encephalopathy. METHODS: The clinical details of 11 affected family members in comparison with 63 unaffected relatives were analyzed. RESULTS: Affected children become comatose after onset of a febrile illness. Outcomes include full recovery, permanent neurologic impairment, and death. Recurrences produce more severe impairments. Lesions of necrotizing encephalopathy of the thalamus and brainstem are present on autopsy and MRI. Oxidative phosphorylation of intact mitochondria from a muscle biopsy shows loose coupling. Unaffected family members, including obligate carriers, share no clinical characteristics, demonstrating incomplete penetrance. CONCLUSIONS: Characteristic pathology and MRI findings define this disorder of autosomal dominant acute encephalopathy. Leigh syndrome and sporadic acute necrotizing encephalopathy share similarities but are distinct.


Subject(s)
Genes, Dominant , Leukoencephalitis, Acute Hemorrhagic/genetics , Brain/pathology , Brain Damage, Chronic/etiology , Child, Preschool , Diseases in Twins , Electron Transport , Fatal Outcome , Female , Fever/complications , Humans , Infant , Infections/complications , Leukoencephalitis, Acute Hemorrhagic/etiology , Leukoencephalitis, Acute Hemorrhagic/pathology , Magnetic Resonance Imaging , Male , Mitochondria/ultrastructure , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Oxidative Phosphorylation , Pedigree , Phenotype
11.
Proc Natl Acad Sci U S A ; 98(20): 11353-8, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11562478

ABSTRACT

Secreted noggin protein regulates bone morphogenetic protein activity during development. In mice, a complete loss of noggin protein leads to multiple malformations including joint fusion, whereas mice heterozygous for Nog loss-of-function mutations are normal. In humans, heterozygous NOG missense mutations have been found in patients with two autosomal dominant disorders of joint development, multiple synostosis syndrome (SYNS1) and a milder disorder proximal symphalangism (SYM1). This study investigated the effect of one SYNS1 and two SYM1 disease-causing missense mutations on the structure and function of noggin. The SYNS1 mutation abolished, and the SYM1 mutations reduced, the secretion of functional noggin dimers in transiently transfected COS-7 cells. Coexpression of mutant noggin with wild-type noggin, to resemble the heterozygous state, did not interfere with wild-type noggin secretion. These data indicate that the human disease-causing mutations are hypomorphic alleles that reduce secretion of functional dimeric noggin. Therefore, we conclude that noggin has both species-specific and joint-specific dosage-dependent roles during joint formation. Surprisingly, in contrast to the COS-7 cell studies, the SYNS1 mutant was able to form dimers in Xenopus laevis oocytes. This finding indicates that there also exist species-specific differences in the ability to process mutant noggin polypeptides.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Mutation, Missense , Proteins/genetics , Proteins/metabolism , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , COS Cells , Carrier Proteins , Chlorocebus aethiops , Dimerization , Disulfides , Female , Humans , Oocytes/physiology , Protein Biosynthesis , Recombinant Proteins/metabolism , Synostosis/genetics , Transfection , Xenopus laevis
12.
Clin Orthop Relat Res ; (379 Suppl): S40-54, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11039750

ABSTRACT

This current study describes how human genetic approaches are used to identify and understand the roles of genes and their protein products, during skeletal development, growth, and homeostasis. Searches for the genes responsible for brachydactyly, symphalangism, spondyloepiphyseal dysplasia, and camptodactyly-arthropathy syndrome are presented to exemplify these approaches. The important role of the orthopaedic surgeon in this process is discussed.


Subject(s)
Bone Development/genetics , Growth/genetics , Homeostasis/genetics , Animals , Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/therapy , Chromosome Mapping , Genetic Therapy , Humans , Mutation
14.
Am J Hum Genet ; 66(2): 368-77, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10677296

ABSTRACT

Otospondylomegaepiphyseal dysplasia (OSMED) is an autosomal recessive skeletal dysplasia accompanied by severe hearing loss. The phenotype overlaps that of the autosomal dominant disorders-Stickler and Marshall syndromes-but can be distinguished by disproportionately short limbs, severe hearing loss, and lack of ocular involvement. In one family with OSMED, a homozygous Gly-->Arg substitution has been described in COL11A2, which codes for the alpha2 chain of type XI collagen. We report seven further families with OSMED. All affected individuals had a remarkably similar phenotype: profound sensorineural hearing loss, skeletal dysplasia with limb shortening and large epiphyses, cleft palate, an extremely flat face, hypoplasia of the mandible, a short nose with anteverted nares, and a flat nasal bridge. We screened affected individuals for mutations in COL11A2 and found different mutations in each family. Individuals from four families, including three with consanguineous parents, were homozygous for mutations. Individuals from three other families, in whom parents were nonconsanguineous, were compound heterozygous. Of the 10 identified mutations, 9 are predicted to cause premature termination of translation, and 1 is predicted to cause an in-frame deletion. We conclude that the OSMED phenotype is highly homogenous and results from homozygosity or compound heterozygosity for COL11A2 mutations, most of which are predicted to cause complete absence of alpha2(XI) chains.


Subject(s)
Collagen/genetics , Deafness/genetics , Genes, Recessive/genetics , Mutation/genetics , Osteochondrodysplasias/genetics , Adult , Alternative Splicing/genetics , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , Codon, Terminator/genetics , Collagen/deficiency , Consanguinity , Deafness/physiopathology , Diseases in Twins/genetics , Exons/genetics , Female , Humans , Infant , Male , Molecular Sequence Data , Osteochondrodysplasias/diagnostic imaging , Osteochondrodysplasias/physiopathology , Pedigree , RNA, Messenger/analysis , RNA, Messenger/genetics , Radiography , Sequence Deletion/genetics
15.
Nat Genet ; 23(3): 319-22, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10545950

ABSTRACT

Altered growth and function of synoviocytes, the intimal cells which line joint cavities and tendon sheaths, occur in a number of skeletal diseases. Hyperplasia of synoviocytes is found in both rheumatoid arthritis and osteoarthritis, despite differences in the underlying aetiologies of the two disorders. We have studied the autosomal recessive disorder camptodactyly-arthropathy-coxa vara-pericarditis syndrome (CACP; MIM 208250) to identify biological pathways that lead to synoviocyte hyperplasia, the principal pathological feature of this syndrome. Using a positional-candidate approach, we identified mutations in a gene (CACP) encoding a secreted proteoglycan as the cause of CACP. The CACP protein, which has previously been identified as both 'megakaryocyte stimulating factor precursor' and 'superficial zone protein', contains domains that have homology to somatomedin B, heparin-binding proteins, mucins and haemopexins. In addition to expression in joint synovium and cartilage, CACP is expressed in non-skeletal tissues including liver and pericardium. The similarity of CACP sequence to that of other protein families and the expression of CACP in non-skeletal tissues suggest it may have diverse biological activities.


Subject(s)
Joint Diseases/genetics , Pericarditis/genetics , Proteoglycans/genetics , Proteoglycans/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cattle , DNA Mutational Analysis , Female , Genotype , Humans , Hyperplasia/genetics , Hyperplasia/pathology , Joint Diseases/pathology , Male , Molecular Sequence Data , Mutation , Pericarditis/pathology , Phenotype , Proteoglycans/chemistry , RNA, Messenger/analysis , RNA, Messenger/genetics , Sequence Homology, Amino Acid , Syndrome , Synovial Membrane/metabolism , Synovial Membrane/pathology
16.
Am J Hum Genet ; 65(4): 974-83, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10486316

ABSTRACT

Stickler and Marshall syndromes are dominantly inherited chondrodysplasias characterized by midfacial hypoplasia, high myopia, and sensorineural-hearing deficit. Since the characteristics of these syndromes overlap, it has been argued whether they are distinct entities or different manifestations of a single syndrome. Several mutations causing Stickler syndrome have been found in the COL2A1 gene, and one mutation causing Stickler syndrome and one causing Marshall syndrome have been detected in the COL11A1 gene. We characterize here the genomic structure of the COL11A1 gene. Screening of patients with Stickler, Stickler-like, or Marshall syndrome pointed to 23 novel mutations. Genotypic-phenotypic comparison revealed an association between the Marshall syndrome phenotype and splicing mutations of 54-bp exons in the C-terminal region of the COL11A1 gene. Null-allele mutations in the COL2A1 gene led to a typical phenotype of Stickler syndrome. Some patients, however, presented with phenotypes of both Marshall and Stickler syndromes.


Subject(s)
Abnormalities, Multiple/genetics , Collagen/genetics , Exons/genetics , Mutation/genetics , Osteochondrodysplasias/genetics , RNA Splicing/genetics , Abnormalities, Multiple/physiopathology , Adolescent , Adult , Child , Child, Preschool , DNA Mutational Analysis , Female , Genotype , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/physiopathology , Humans , Introns/genetics , Male , Molecular Sequence Data , Myopia/genetics , Myopia/physiopathology , Osteochondrodysplasias/physiopathology , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion/genetics , Syndrome
17.
Nat Genet ; 23(1): 94-8, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10471507

ABSTRACT

Members of the CCN (for CTGF, cyr61/cef10, nov) gene family encode cysteine-rich secreted proteins with roles in cell growth and differentiation. Cell-specific and tissue-specific differences in the expression and function of different CCN family members suggest they have non-redundant roles. Using a positional-candidate approach, we found that mutations in the CCN family member WISP3 are associated with the autosomal recessive skeletal disorder progressive pseudorheumatoid dysplasia (PPD; MIM 208230). PPD is an autosomal recessive disorder that may be initially misdiagnosed as juvenile rheumatoid arthritis. Its population incidence has been estimated at 1 per million in the United Kingdom, but it is likely to be higher in the Middle East and Gulf States. Affected individuals are asymptomatic in early childhood. Signs and symptoms of disease typically develop between three and eight years of age. Clinically and radiographically, patients experience continued cartilage loss and destructive bone changes as they age, in several instances necessitating joint replacement surgery by the third decade of life. Extraskeletal manifestations have not been reported in PPD. Cartilage appears to be the primary affected tissue, and in one patient, a biopsy of the iliac crest revealed abnormal nests of chondrocytes and loss of normal cell columnar organization in growth zones. We have identified nine different WISP3 mutations in unrelated, affected individuals, indicating that the gene is essential for normal post-natal skeletal growth and cartilage homeostasis.


Subject(s)
Growth Substances/genetics , Immediate-Early Proteins , Intercellular Signaling Peptides and Proteins , Mutation , Oncogene Proteins , Osteochondrodysplasias/genetics , Adolescent , Bone and Bones/physiology , CCN Intercellular Signaling Proteins , Cartilage/growth & development , Cartilage/physiology , Chromosomes, Human, Pair 6 , Connective Tissue Growth Factor , Hand/diagnostic imaging , Haplotypes , Humans , Intracellular Signaling Peptides and Proteins , Male , Molecular Sequence Data , Nephroblastoma Overexpressed Protein , Osteochondrodysplasias/diagnostic imaging , Proto-Oncogene Proteins , Radiography
18.
J Biol Chem ; 274(32): 22469-75, 1999 Aug 06.
Article in English | MEDLINE | ID: mdl-10428822

ABSTRACT

We report the complete sequence of the human COL9A3 gene that encodes the alpha3 chain of heterotrimeric type IX collagen, a member of the fibril-associated collagens with interrupted triple helices family of collagenous proteins. Nucleotide sequencing defined over 23,000 base pairs (bp) of the gene and about 3000 bp of the 5'-flanking sequences. The gene contains 32 exons. The domain and exon organization of the gene is almost identical to a related gene, the human COL9A2 gene. However, exon 2 of the COL9A3 gene codes for one -Gly-X-Y- triplet less than exon 2 of the COL9A2 gene. The difference is compensated by an insertion of 9 bp coding for an additional triplet in exon 4 of the COL9A3 gene. As a result, the number of -Gly-X-Y- repeats in the third collagenous domain remains the same in both genes and ensures the formation of an in-register triple helix. In the course of screening this gene for mutations, heterozygosity for separate 9-bp deletions within the COL1 domain were identified in two kindreds. In both instances, the deletions did not co-segregate with any disease phenotype, suggesting that they were neutral variants. In contrast, similar deletions in triple helical domain of type I collagen are lethal. To study whether alpha3(IX) chains with the deletion will participate in the formation of correctly folded heterotrimeric type IX collagen, we expressed mutant alpha3 chains together with normal alpha1 and alpha2 chains in insect cells. We show here that despite the deletion, mutant alpha3 chains were secreted as heterotrimeric, triple helical molecules consisting of three alpha chains in a 1:1:1 ratio. The results suggest that the next noncollagenous domain (NC2) is capable of correcting the alignment of the alpha chains, and this ensures the formation of an in-register triple helix.


Subject(s)
Collagen Type IX , Collagen/genetics , Genetic Variation , Mutation , Sequence Deletion , Amino Acid Sequence , Base Sequence , Collagen/chemistry , Exons , Genetic Testing , Genomic Library , Heterozygote , Humans , Introns , Models, Molecular , Molecular Sequence Data , Osteochondrodysplasias/genetics , Protein Folding , Protein Structure, Secondary , Sequence Analysis, DNA
19.
Am J Med Genet ; 85(4): 413-8, 1999 Aug 06.
Article in English | MEDLINE | ID: mdl-10398270

ABSTRACT

We report on the clinical manifestations in six affected individuals from a four-generation family that segregates brachydactyly type D (BDD). All affected individuals have either bilateral and symmetric or unilateral first distal phalangeal hypoplasia. Metacarpal-phalangeal profiles show that some affected individuals also have a more generalized involvement of the apical skeleton. However, other than first distal phalangeal hypoplasia, there is no consistent pattern of associated skeletal involvement. Linkage analyses were preformed between the BDD phenotype in this family and six loci known to contain genes involved in apical skeletal patterning. No statistically significant linkage was detected.


Subject(s)
Foot Deformities, Congenital/diagnostic imaging , Foot Deformities, Congenital/genetics , Genetic Linkage , Hand Deformities, Congenital/diagnostic imaging , Hand Deformities, Congenital/genetics , Foot Deformities, Congenital/pathology , Humans , Pedigree , Radiography , Thumb/abnormalities , Thumb/pathology
20.
Am J Hum Genet ; 65(1): 125-33, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10364524

ABSTRACT

Venous malformations (VMs) are localized defects of vascular morphogenesis. They can occur in every organ system, most commonly in skin and muscle. They can cause pain and bleeding, and in some critical locations they can be life threatening. Usually venous anomalies occur sporadically, but families with dominant inheritance have been identified. Using linkage analysis, we have established in earlier reports that some families with inherited VMs show linkage to chromosome 9p21; the mutation causes ligand-independent activation of an endothelial cell-specific receptor tyrosine kinase, TIE-2. Here we show that VMs with glomus cells (known as "glomangiomas"), inherited as an autosomal dominant trait in five families, are not linked to 9p21 but, instead, link to a new locus, on 1p21-p22, called "VMGLOM" (LOD score 12.70 at recombination fraction.00). We exclude three known positional candidate genes, DR1 (depressor of transcription 1), TGFBR3 (transforming growth factor-beta receptor, type 3), and TFA (tissue factor). We hypothesize that cutaneous venous anomalies (i.e., glomangiomas) are caused by mutations in a novel gene that may act to regulate angiogenesis, in concert with the TIE-2 signaling pathway.


Subject(s)
Chromosomes, Human, Pair 1 , Glomus Tumor/genetics , Skin Neoplasms/genetics , Genetic Linkage , Genetic Markers , Glomus Tumor/pathology , Humans , Lod Score , Models, Genetic , Pedigree , Skin Neoplasms/pathology , Veins/anatomy & histology , Veins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...