Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Matrix Biol ; 90: 40-60, 2020 08.
Article in English | MEDLINE | ID: mdl-32173581

ABSTRACT

Prolyl 3-hydroxylation is a rare collagen type I post translational modification in fibrillar collagens. The primary 3Hyp substrate sites in type I collagen are targeted by an endoplasmic reticulum (ER) complex composed by cartilage associated protein (CRTAP), prolyl 3-hydroxylase 1 (P3H1) and prolyl cis/trans isomerase B, whose mutations cause recessive forms of osteogenesis imperfecta with impaired levels of α1(I)3Hyp986. The absence of collagen type I 3Hyp in wild type zebrafish provides the unique opportunity to clarify the role of the complex in vertebrate. Zebrafish knock outs for crtap and p3h1 were generated by CRISPR/Cas9. Mutant fish have the typical OI patients' reduced size, body disproportion and altered mineralization. Vertebral body fusions, deformities and fractures are accompanied to reduced size, thickness and bone volume. Intracellularly, collagen type I is overmodified, and partially retained causing enlarged ER cisternae. In the extracellular matrix the abnormal collagen type I assembles in disorganized fibers characterized by altered diameter. The data support the defective chaperone role of the 3-hydroxylation complex as the primary cause of the skeletal phenotype.


Subject(s)
Collagen Type II/metabolism , Collagen Type I/metabolism , Extracellular Matrix Proteins/genetics , Osteogenesis Imperfecta/genetics , Prolyl Hydroxylases/genetics , Animals , CRISPR-Cas Systems , Cyclophilins/genetics , Disease Models, Animal , Gene Knockout Techniques , Hydroxylation , Osteogenesis Imperfecta/metabolism , Phenotype , Prolyl Hydroxylases/chemistry , Zebrafish , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
2.
Sci Rep ; 6: 21540, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26876635

ABSTRACT

Over the last years the zebrafish imposed itself as a powerful model to study skeletal diseases, but a limit to its use is the poor characterization of collagen type I, the most abundant protein in bone and skin. In tetrapods collagen type I is a trimer mainly composed of two α1 chains and one α2 chain, encoded by COL1A1 and COL1A2 genes, respectively. In contrast, in zebrafish three type I collagen genes exist, col1a1a, col1a1b and col1a2 coding for α1(I), α3(I) and α2(I) chains. During embryonic and larval development the three collagen type I genes showed a similar spatio-temporal expression pattern, indicating their co-regulation and interdependence at these stages. In both embryonic and adult tissues, the presence of the three α(I) chains was demonstrated, although in embryos α1(I) was present in two distinct glycosylated states, suggesting a developmental-specific collagen composition. Even though in adult bone, skin and scales equal amounts of α1(I), α3(I) and α2(I) chains are present, the presented data suggest a tissue-specific stoichiometry and/or post-translational modification status for collagen type I. In conclusion, this data will be useful to properly interpret results and insights gained from zebrafish models of skeletal diseases.


Subject(s)
Bone Development/genetics , Collagen Type I/genetics , Collagen/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Collagen/biosynthesis , Collagen Type I/biosynthesis , Gene Expression Regulation, Developmental , Protein Processing, Post-Translational , Skin/growth & development , Skin/metabolism , Zebrafish/growth & development , Zebrafish Proteins/biosynthesis
3.
J Med Genet ; 45(12): 827-31, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18708426

ABSTRACT

BACKGROUND: Mutations in the sulfate transporter gene SLC26A2 (DTDST) cause a continuum of skeletal dysplasia phenotypes that includes achondrogenesis type 1B (ACG1B), atelosteogenesis type 2 (AO2), diastrophic dysplasia (DTD), and recessive multiple epiphyseal dysplasia (rMED). In 1972, de la Chapelle et al reported two siblings with a lethal skeletal dysplasia, which was denoted "neonatal osseous dysplasia" and "de la Chapelle dysplasia" (DLCD). It was suggested that DLCD might be part of the SLC26A2 spectrum of phenotypes, both because of the Finnish origin of the original family and of radiographic similarities to ACG1B and AO2. OBJECTIVE: To test the hypothesis whether SLC26A2 mutations are responsible for DLCD. METHODS: We studied the DNA from the original DLCD family and from seven Finnish DTD patients in whom we had identified only one copy of IVS1+2T>C, the common Finnish mutation. A novel SLC26A2 mutation was found in all subjects, inserted by site-directed mutagenesis in a vector harbouring the SLC26A2 cDNA, and expressed in sulfate transport deficient Chinese hamster ovary (CHO) cells to measure sulfate uptake activity. RESULTS: We identified a hitherto undescribed SLC26A2 mutation, T512K, homozygous in the affected subjects and heterozygous in both parents and in the unaffected sister. T512K was then identified as second pathogenic allele in the seven Finnish DTD subjects. Expression studies confirmed pathogenicity. CONCLUSIONS: DLCD is indeed allelic to the other SLC26A2 disorders. T512K is a second rare "Finnish" mutation that results in DLCD at homozygosity and in DTD when compounded with the milder, common Finnish mutation.


Subject(s)
Anion Transport Proteins/genetics , Mutation , Osteochondrodysplasias/genetics , Animals , Anion Transport Proteins/metabolism , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Female , Finland , Humans , Infant, Newborn , Male , Osteochondrodysplasias/pathology , Pedigree , Population Groups/genetics , Sulfate Transporters , Transfection
4.
Amino Acids ; 35(4): 739-52, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18340504

ABSTRACT

Here we summarized what is known at the present about function, structure and effect of mutations in the human prolidase. Among the peptidases, prolidase is the only metalloenzyme that cleaves the iminodipeptides containing a proline or hydroxyproline residue at the C-terminal end. It is relevant in the latest stage of protein catabolism, particularly of those molecules rich in imino acids such as collagens, thus being involved in matrix remodelling. Beside its intracellular functions, prolidase has an antitoxic effect against some organophosphorus molecules, can be used in dietary industry as bitterness reducing agent and recently has been used as target enzyme for specific melanoma prodrug activation. Recombinant human prolidase was produced in prokaryotic and eukaryotic hosts with biochemical properties similar to the endogenous enzyme and represents a valid tool both to better understand the structure and biological function of the enzyme and to develop an enzyme replacement therapy for the prolidase deficiency (PD). Prolidase deficiency is a rare recessive disorder caused by mutations in the prolidase gene and characterized by severe skin lesions. Single amino acid substitutions, exon splicing, deletions and a duplication were described as causative for the disease and are mainly located at highly conserved amino acids in the sequence of prolidase from different species. The pathophysiology of PD is still poorly understood; we offer here a review of the molecular mechanisms so far hypothesized.


Subject(s)
Dipeptidases/deficiency , Dipeptidases/genetics , Dipeptidases/physiology , Mutation , Proline/chemistry , Amino Acid Sequence , Dipeptidases/chemistry , Enzyme Activation , Enzyme Therapy , Genotype , Humans , Metals/chemistry , Molecular Sequence Data , Phenotype , Prodrugs , Protein Structure, Tertiary , Pyrococcus/metabolism , Sequence Homology, Amino Acid
5.
J Inherit Metab Dis ; 30(5): 814, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17570078

ABSTRACT

This report describes the case of a boy with prolidase deficiency who presented with splenomegaly and leg ulcers. Laboratory examination revealed hypergammaglobulinaemia, hyperimmunoglobulinaemia E, increased erythrocyte sedimentation rate, elevated transaminases, positive antinuclear and anti-double-stranded DNA antibodies, and complement consumption. No haematological, renal or articular problems were detected; neutrophil count was normal. The skin lesions were thought to be of vasculitic origin, and a diagnosis of systemic lupus erythematosus was made although the requirements for diagnosis of systemic lupus erythematosus based on American Rheumatism Association criteria were not satisfied. The child was treated with immunosuppressive drugs with worsening of skin lesions before the diagnosis of prolidase deficiency. Prolidase deficiency and systemic lupus erythematosus share a number of common immunological features and at least three patients with prolidase deficiency and immunological and clinical findings fulfilling the diagnostic criteria for systemic lupus erythematosus of the American Rheumatism Association are reported in the literature. Here we review pathogenetic hypothesis linking the metabolic defect to the disturbance in immune function. In particular we discuss the role of highly increased rates of apoptosis and/or abnormal processing of apoptotic keratinocytes in prolidase deficiency and the role of C1q deficiency, which is associated with the failure of normal clearance of apoptotic cells bearing on their surfaces many of the autoantigens involved in systemic lupus erythematosus.


Subject(s)
Diagnostic Errors , Dipeptidases/deficiency , Lupus Erythematosus, Systemic/diagnosis , Metabolism, Inborn Errors/diagnosis , Child , Humans , Lupus Erythematosus, Systemic/immunology , Male , Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/immunology , Practice Guidelines as Topic
6.
J Med Genet ; 43(12): e58, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17142620

ABSTRACT

Prolidase deficiency (PD) is a rare autosomal recessive connective tissue disorder caused by mutations in the prolidase gene. The PD patients show a wide range of clinical outcomes characterised mainly by intractable skin ulcers, mental retardation and recurrent respiratory infections. Here we describe five different PEPD mutations in six European patients. We identified two new PEPD mutant alleles: a 13 bp duplication in exon 8, which is the first reported duplication in the prolidase gene and a point mutation resulting in a change in amino acid E412, a highly conserved residue among different species. The E412K substitution is responsible for the first reported phenotypic variability within a family with severe and asymptomatic outcomes.


Subject(s)
Dipeptidases/deficiency , Dipeptidases/genetics , Gene Duplication , Mutation/genetics , Adult , Amino Acid Sequence , Child , Child, Preschool , Connective Tissue Diseases/enzymology , Connective Tissue Diseases/genetics , Connective Tissue Diseases/pathology , DNA Mutational Analysis , Denmark , Family Health , Female , Genotype , Humans , Intellectual Disability/pathology , Italy , Male , Molecular Sequence Data , Mutation, Missense/genetics , Pedigree , Phenotype , Sequence Homology, Amino Acid , Skin Ulcer/pathology , Turkey
7.
J Periodontal Res ; 38(6): 568-74, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14632919

ABSTRACT

BACKGROUND: Several clinical trials have shown the effectiveness of Emdogain(R) (EMD) in promoting tissue regeneration, even though the underlining biological mechanism is still poorly known. OBJECTIVES: The aim of the present study was to verify the effect of EMD on the proliferation of human periodontal ligament (PDL) fibroblasts and on their colonization and differentiation following contact with the root surface of extracted teeth in vitro. METHODS AND RESULTS: Fibroblasts from PDL were seeded on Petri dishes and cell growth was evaluated by cell counting in the presence and absence of EMD, after 1, 3 and 8 d of culture. A significant effect of EMD upon cellular proliferation at d 3 and 8 was detected. When PDL cells were grown for 12 d with EMD on etched human root surface, a change in cell morphology was observed. Scanning electron microscopy revealed that cells grown on root EMD-treated surface present a body with a flattened surface closely adherent to the substrate and an outer smooth surface rounded in shape. From the flattened surface some thin and elongated cellular processes connecting with the substrate were also observable. PDL cells grown on EMD-treated surface showed lack of alkaline phosphatase activity, as some authors noticed in cementoblasts in vitro. CONCLUSIONS: In conclusion, our data indicate that EMD enhances human PDL fibroblast proliferation. Furthermore, the cells in the presence of EMD show morphological changes that make them more similar to cementoblasts than to fibroblasts, suggesting a process of cellular differentiation that could play an important role in periodontal tissue repair.


Subject(s)
Dental Enamel Proteins/pharmacology , Fibroblasts/drug effects , Periodontal Ligament/drug effects , Tooth Root/drug effects , Adult , Alkaline Phosphatase/analysis , Analysis of Variance , Cell Adhesion/drug effects , Cell Count , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Size/drug effects , Cells, Cultured , Dental Cementum/cytology , Dental Cementum/drug effects , Female , Fibroblasts/enzymology , Humans , Microscopy, Electron, Scanning , Multivariate Analysis , Periodontal Ligament/cytology , Time Factors
8.
Biomaterials ; 23(14): 2863-9, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12069326

ABSTRACT

To reveal the biocompatibility of TiMo12Zr6Fe2 (TMZF), a new titanium alloy used since 1998 for orthopaedic prosthesis, we compared the behavior of primary human fibroblasts and osteoblasts grown on TMZF discs or on plastic tissue culture dishes, a widely used material specifically treated by the manufacturer to enhance cell growth. Proliferation, differentiation. RNA and collagen type I expression level of human cells were carried out. The analysis were performed over a period of 96 h. Fibroblasts behaved at the same way on the two different supports after 48 h, their number increased after 96 h when cells were grown on the alloy. Osteoblasts adhered and proliferated on the alloy discs as well as on plastic. RNA expression level was not affected. Interestingly, cell number at each time point was higher for fibroblasts than for osteoblasts. The RNA expression level was higher for the osteoblasts. Both cell types cultured on the alloy revealed an increase in the amount of type I collagen and a similar electrophoretic pattern was found for collagen produced by fibroblasts and osteoblasts grown on either supports. These results indicate good biocompatibility of the TMZF alloy, which allowed adhesion and proliferation of both the examined cell types and suggest that TMZF is a promising material for orthopaedic implants.


Subject(s)
Alloys/metabolism , Biocompatible Materials/metabolism , Fibroblasts/physiology , Osteoblasts/physiology , Prostheses and Implants , Cell Adhesion , Cell Division , Cells, Cultured , Collagen Type I/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , In Vitro Techniques , Iron/metabolism , Materials Testing , Molybdenum/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Phenotype , RNA/metabolism , Titanium/metabolism , Zirconium/metabolism
9.
Mol Genet Metab ; 71(1-2): 225-32, 2000.
Article in English | MEDLINE | ID: mdl-11001814

ABSTRACT

Osteogenesis Imperfecta (OI) is a dominant negative disorder of connective tissue. OI patients present with bone fragility and skeletal deformity within a broad phenotypic range. Defects in the COL1A1 or COL1A2 genes, coding, respectively, for the alpha1 and alpha2 chains of type I collagen, are the causative mutations. Over 150 mutations have been characterized. Both quantitative defects, such as null COL1A1 alleles, and qualitative defects, such as glycine substitutions, exon skipping, deletions, and insertions, have been described in type I collagen. Quantitative and structural mutations are associated with the milder and more severe forms of OI, respectively. A more detailed relationship between genotype and phenotype is still incompletely understood; several models have been proposed and are being tested. Transgenic and knock-out murine models for OI have previously been created. We have recently generated a knock-in murine model (the Brittle mouse) carrying a typical glycine substitution in type I collagen. This mouse will permit a better understanding of OI pathophysiology and phenotypic variability. It will also be used for gene therapeutic approaches to OI, especially mutation suppression by hammerhead ribozymes. The present review will provide an update of OI clinical and molecular data and outline gene therapeutic approaches being tested on OI murine models for this disorder.


Subject(s)
Genetic Therapy , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/therapy , Alleles , Animals , Disease Models, Animal , Gene Expression , Humans , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Phenotype
10.
J Biol Chem ; 274(53): 37923-31, 1999 Dec 31.
Article in English | MEDLINE | ID: mdl-10608859

ABSTRACT

We utilized the Cre/lox recombination system to develop the first knock-in murine model for osteogenesis imperfecta (OI). The moderately severe OI phenotype was obtained from an alpha1(I) Gly(349) --> Cys substitution in type I collagen, reproducing the mutation in a type IV OI child. We introduced four single nucleotide (nt) changes into murine col1a1 exon 23: the disease causing G-->T transversion (nt 1546), an adjacent G-->T change (nt 1551) to generate a GUC ribozyme cleavage site, and two transversions (nt 1567 C-->A and nt 1569 C-->G) to cause a Leu --> Met substitution. We also introduced a 3.2-kilobase pair transcription/translation stop cassette in intron 22, flanked by directly repeating lox recombination sites. After homologous recombination in ES cells, two male chimeras were obtained. Chimeras were mated with transgenic females expressing Cre recombinase to remove the stop cassette from a portion of the progeny's cells. To generate mice with full expression of the Gly(349) --> Cys mutation, these offspring were then mated with wild-type females. Skeletal staining and bone histology of the F2 revealed a classical OI phenotype with deformity, fragility, osteoporosis and disorganized trabecular structure. We designate these mice BrtlIV (Brittle IV). BrtlIV mice have phenotypic variability ranging from perinatal lethality to long term survival with reproductive success. The phenotypic variability is not associated with differences in expression levels of the mutant allele in total RNA derived from tissue extracts. Expression of the mutant protein is also equivalent in different phenotypes. Thus, these mice are an excellent model for delineation of the modifying factors postulated to affect human OI phenotypes. In addition, we generated knock-in mice carrying an "intronic" inclusion by mating chimeras with wild-type females. Alternative splicing involving the stop cassette results in retention of non-collagenous sequences. These mice reproduce the lethal phenotype of similar human mutations and are designated BrtlII.


Subject(s)
Integrases/metabolism , Osteogenesis Imperfecta/genetics , Viral Proteins , Alternative Splicing , Amino Acid Substitution , Animals , Base Sequence , Chimera , Collagen/genetics , Collagen/metabolism , Cysteine/genetics , Disease Models, Animal , Female , Glycine/genetics , Heterozygote , Humans , Male , Mice , Mice, Mutant Strains , Molecular Sequence Data , Phenotype , RNA, Messenger/genetics
11.
Hum Mutat ; 11(5): 395-403, 1998.
Article in English | MEDLINE | ID: mdl-9600458

ABSTRACT

In three cases of type IV osteogenesis imperfecta (OI), we identified unique point mutations in type I collagen alpha1(I) cDNA. In two cases, the appearance of dimers indicated the presence of cysteine substitutions in the alpha1(I) protein chain. Cyanogen bromide digestion localized these cross-links to CB8 and 3, respectively. In the third case, the overmodification pattern of the CNBr peptides was compatible with a substitution in the aa 123-402 region of either type I collagen chain. We identified a unique point mutation in each proband, which resulted in substitutions for glycine residues in a 300-aa region of the alpha1(I) helix, specifically, Gly to Ala at codon 220 (GGT-->GCT), Gly to Cys at codon 349 (GGT-->TGT) and Gly to Cys at codon 523 (GGT-->TGT). We compared each proband's fibroblast and osteoblast collagen directly, as well as with fibroblast and osteoblast controls. For all cases, the OI osteoblast collagen was more electrophoretically delayed than OI fibroblast collagen. In the patient with G349C, OI fibroblast and osteoblast collagen synthesized in the presence of alpha,alpha'-dipyridyl co-migrated on gels, demonstrating that the electrophoretic discrepancy resulted from differences in post-translational modification. Melting temperature curves for stability of the collagen helix yielded an identical Tm for control fibroblast and osteoblast collagen (41.2 degrees C). By contrast, for collagen with the gly349-->cys substitution, the Tm of the fibroblast collagen was 1 degree C lower than the Tm of the osteoblast collagen. These data indicate that the metabolism of mutant collagen might be cell-specific and has significant implications for understanding the phenotype/genotype correlations and the pathophysiology of OI.


Subject(s)
Collagen/genetics , Fibroblasts/chemistry , Osteoblasts/chemistry , Osteogenesis Imperfecta/genetics , Point Mutation , Adolescent , Amino Acid Sequence , Cells, Cultured , Child , Child, Preschool , Collagen/chemistry , Cyanogen Bromide , DNA, Complementary/analysis , Electrophoresis, Polyacrylamide Gel , Female , Femur/diagnostic imaging , Genes, Dominant , Glycine/genetics , Humans , Male , Osteogenesis Imperfecta/metabolism , Polymorphism, Single-Stranded Conformational , Radiography , Sequence Analysis, DNA , Temperature
12.
Matrix Biol ; 17(8-9): 575-84, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9923651

ABSTRACT

Type I collagen synthesized by cultured skin fibroblasts was analyzed biochemically and molecularly to characterize the defect in a patient affected by lethal Osteogenesis Imperfecta. The SDS-Urea-PAGE of procollagen and collagen revealed a broad alpha1(I) band, a normal alpha2(I) and another alpha2(I) band migrating equidistant between alpha1 and alpha2. When synthesized in the presence of alphaalpha'-dipyridyl, an inhibitor of prolyl and lysyl hydroxylation, procollagen and collagen of media and cell layers contained both normal and slower alpha2(I), but only normal alpha1(I). The persistence of the two forms of alpha2(I) chains suggested a mutation in a COL1A2 gene. CNBr cleavage of collagen yielded overmodified alpha1(I) CB3 and CB7 peptides and delayed migration of the alpha2(I) CB3-5 peptide. A delayed CB3-5 was also found after alpha,alpha'-dipyridyl treatment. These data localized the mutation between aa 353 and 551 in alpha2(I) (CB3-5). Sequencing the subcloned alleles in this region revealed a G-->A transition at nt 1671 in one allele, changing Gly 421 to Asp in an alpha2(I) chain. The mutation was demonstrated to occur on the paternally derived allele, using a common C-->A polymorphism at alpha2(I) nt 1585 and by the presence of a rare variant, Arg618-->Gln (Phillips et al., 1990), in the paternal genomic DNA and the proband's mutant allele. Procollagen processing was normal. The Tm of the slow alpha2(I) collagen was 2 degrees C lower than the control, indicating decreased triple helix stability. Mutant collagen was incorporated in the extracellular matrix deposited by cultured fibroblasts. The dramatic delay in alpha2(I) electrophoretic mobility must be induced by the Gly-->Asp substitution, since the Arg-->Gln variant causes only mild electrophoretic delay. Substantial delay in gel mobility even in the absence of overmodification suggested the presence of a kink in the mutated alpha2(I) chains. Rotary shadowing electron microscopy of secreted fibroblast procollagen confirmed the presence of a kink in the region of the helix containing the glycine substitution. The kinking of the collagen helix occurs in the absence of dimer formation. Kinking may interfere with normal helix folding, as well as with the interactions of collagen fibrils with the collagenous and non-collagenous extracellular matrix proteins.


Subject(s)
Aspartic Acid/genetics , Collagen/genetics , Glycine/genetics , Osteogenesis Imperfecta/genetics , Amino Acid Substitution , Aspartic Acid/physiology , Fatal Outcome , Glycine/physiology , Humans , Infant , Male
13.
Nucleic Acids Res ; 25(17): 3451-8, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9254703

ABSTRACT

We report here the in vitro use of hammerhead ribozymes as an approach to the gene therapy of osteogenesis imperfecta (OI). Our strategy for the treatment of this dominant genetic disorder is based on selective reduction of the level of the mRNA transcripts from the mutant allele. We studied the in vitro cleavage activity of five different hammerhead ribozymes targeted against synthetic transcripts of two naturally occurring human collagen mutations and against a point mutation introduced into a construct containing a portion of the mouse COL1A1 gene. This is the first demonstration that ribozyme cleavage is absolutely dependent on the presence of the ribozyme cleavage site introduced by the disease-causing mutation. Cleavage specificity and activity were unchanged when the cleavage site was located in transcripts of progressively longer length. Cleavage efficiency depended directly on the ratio of ribozyme/substrate, as well as on the time and temperature of incubation. We investigated the competitive effects of both total RNA and normal synthetic transcripts on ribozyme cleavage activity. The ribozyme was able to localize and cleave its specific target even in the presence of a vast excess of total RNA. However, cleavage efficiency was linearly inhibited by the presence of a non- cleavable competitor substrate which contained a ribozyme binding site identical to the site present in the cleavable target. Although this competition could be eliminated by introducing a mismatch into one ribozyme binding arm, the presence of the mismatch decreased ribozyme cleavage efficiency. The mutation- specificity of ribozyme cleavage demonstrated in this work provides support for in vivo studies aimed at ribozyme development as a treatment for dominant negative genetic disorders.


Subject(s)
Collagen/genetics , Genetic Therapy , Mutation , Osteogenesis Imperfecta/genetics , RNA, Catalytic/metabolism , RNA/metabolism , Animals , Base Sequence , Binding, Competitive , Electrophoresis, Polyacrylamide Gel , Humans , Mice , Mutagenesis, Site-Directed , Osteogenesis Imperfecta/therapy , Point Mutation
14.
Biochem Mol Med ; 62(1): 26-35, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9367795

ABSTRACT

We examined the type I collagen synthesized by cultured dermal fibroblasts from a patient affected with osteogenesis imperfecta (OI) type IV. Both normal and abnormal trimers were produced. The mutant collagen molecules were excessively modified intracellularly, had a melting temperature 4 degrees C lower than the control, were secreted at a reduced rate, and underwent delayed processing to mature alpha chains.Molecular investigations identified a G --> A transition in one COL1A2 allele, resulting in a Gly922 --> Ser substitution in the alpha2(I) chain. The proband's mutation was demonstrated to arise "de novo" by the absence of the mutant allele restriction enzyme pattern from parental genomic DNA.We analyzed the insoluble extracellular matrix deposited by long-term cultured fibroblasts from our patient and from a previously described unrelated individual who carries an identical substitution. In both cases, the mutant chain constituted 10-15% of the total alpha chains deposited.We also present here the first detailed comparison of phenotype between unrelated OI patients with an identical collagen mutation. These two patients are both Caucasian females, ages 8 and 9 years, each diagnosed as type IV OI by the Sillence classification. They have a similar phenotype including moderate skeletal fragility with several femur fractures, dentinogenesis imperfecta, wormian bone, and reduced height and weight. We conclude that this phenotype is related both to the location of this mutation and to the similar extent of matrix incorporation by the mutant chains. Molecular and biochemical studies of unrelated individuals with identical amino acid substitutions in type I collagen resulting in either similar or dissimilar clinical outcomes will make a significant contribution to identifying the factors involved in the modulation of the OI phenotype.


Subject(s)
Collagen/metabolism , Mutation , Osteogenesis Imperfecta/genetics , Cells, Cultured , Child , Female , Humans , Phenotype
15.
J Biol Chem ; 271(45): 28617-23, 1996 Nov 08.
Article in English | MEDLINE | ID: mdl-8910493

ABSTRACT

We have identified a novel multiexon genomic deletion in one COL1A1 collagen allele that results in three alternative forms of mutant mRNA. This mutation occurs in a 9-year-old girl and her father, both affected with severe type III osteogenesis imperfecta (OI). We previously reported detection of a mismatch in their alpha1(I) amino acids 558-861 region by RNA/RNA hybrid analysis (Grange, D. K., Gottesman, G. S., Lewis, M. B., and Marini, J. C. (1990) Nucleic Acids Res. 18, 4227-4236). Single Strand Conformational Polymorphism further localized the mRNA mutation to the amino acids 579-679 coding region. At the gene level, polymerase chain reaction (PCR) amplification of patient leukocyte DNA from the exon 33-38 region yielded the normal 1004-base pair (bp) fragment and an additional 442-bp fragment. Sequencing of the shorter genomic PCR product confirmed the presence of a 562-bp deletion, extending from the last 3 nucleotides (nt) of exon 34 to 156 nt from the 3'-end of intron 36. The genomic deletion was also detected in the clinically normal grandmother, who was confirmed to be a mosaic carrier. PCR amplification and RNase protection experiments were used to investigate the mRNA structure and occurrence of alternative splicing. One form of the mutant cDNA has a deletion with end points that are identical to the genomic deletion. This results in a combination deletion/insertion, with a deletion of amino acids 603-639 followed by an insertion of 156 nt from the 3'-end of intron 36. In addition, we found two alternatively spliced forms. One form uses a cryptic donor site in exon 34 and the exon 37 acceptor. The second form uses the normal exon 32 splice donor and exon 37 acceptor. Use of the cryptic donor results in a coding sequence that is out-of-frame. Both the retained intron form and the use of the exon 32 donor site result in coding sequences that are in-frame. This is the first report of a collagen defect in OI with alternative splicing generating both in-frame and out-of-frame forms of mRNA. Although the in-frame forms constitute more than 60% of the mRNA from the mutant allele, no mutant protein chain was identified. Collagen produced by cultured OI osteoblasts showed a significant increase in the relative amount of type III collagen but no mutant alpha1(I) chain.


Subject(s)
Alleles , Alternative Splicing , Collagen/genetics , Osteogenesis Imperfecta/genetics , RNA, Messenger/chemistry , Adult , Aged , Child , DNA, Complementary/analysis , Female , Humans , Male , Pedigree , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Procollagen/genetics , Sequence Analysis, DNA
16.
J Neurol Sci ; 141(1-2): 33-8, 1996 Sep 15.
Article in English | MEDLINE | ID: mdl-8880689

ABSTRACT

An altered equilibrium of protease/protease-inhibitor factors may be involved in the pathogenesis of aneurysm rupture: alpha 1-antitrypsin (alpha 1-AT) represents the most relevant inhibitor of elastase, a proteolytic enzyme enhancing catabolic processes of collagen metabolism. Cigarette smoking has been shown to significantly reduce the inhibitory effect of alpha 1-AT on proteases. In the present study we test the hypothesis whether the activity of alpha 1-AT is altered in patients with subarachnoid haemorrhage (SAH) and if is there any relationship between alpha 1-AT activity and the high risk of aneurysm rupture in smokers. The patients were subdivided in the following groups: (a) patients with unruptured aneurysm (n = 10); (b) patients presenting with SAH admitted within 48 h after the episode (n = 20); (c) patients presenting with SAH admitted > 48 h after the episode (n = 14); (d) controls (n = 10): patients with neither cerebrovascular nor acute disease. Blood samples were obtained immediately at admission. Measurement of alpha 1-AT level was determined by immunoturbidimetric method. In order to obtain qualitative data about the anti-protease activity of alpha 1-AT (expressed as collagenase inhibitory percentage capacity (CIC) at different doses) we consider the 20 cases admitted for SAH within 48 h. The mean serum level of patients with unruptured aneurysms is significantly lower than that of patients with SAH (p < 0.01), while the mean serum level of alpha 1-AT in controls does not significantly differ from other groups. The mean serum level of alpha 1-AT in patients admitted > 48 h after SAH is significantly higher than that of patients admitted within 48 h after the haemorrhage (p < 0.02). Considering the smoking habit of patients, there is no significant difference in alpha 1-AT levels in each subgroup of patients. A multivariate analysis considering alpha 1-AT CIC, showed that alpha 1-AT CIC in patients with ruptured aneurysms is significantly reduced if compared to controls and unruptured aneurysms (F = 50.759; p < 0.001). Moreover, considering alpha 1-AT CIC and smoking habit in each group the covariance analysis showed that while in controls and unruptured aneurysms there is no difference in alpha 1-AT CIC between smokers and non smokers, in cases of SAH, cigarette smoking significantly influences the alpha 1-AT CIC. The present results suggest that the basic mechanism behind the increased risk of SAH in smokers involves a qualitative deficiency of alpha 1-AT.


Subject(s)
Aneurysm, Ruptured/enzymology , Smoking/adverse effects , Subarachnoid Hemorrhage/etiology , alpha 1-Antitrypsin/metabolism , Adult , Aged , Analysis of Variance , Aneurysm, Ruptured/epidemiology , Aneurysm, Ruptured/pathology , Female , Humans , Male , Middle Aged , Regression Analysis , Risk Factors , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/epidemiology
17.
Am J Med Genet ; 63(1): 161-6, 1996 May 03.
Article in English | MEDLINE | ID: mdl-8723103

ABSTRACT

In osteogenesis imperfecta (OI) the effects of mutations in type I collagen genes generally reflect their nature and localization. Unrelated individuals sharing identical mutations present, in general, similar clinical phenotypes. However, in some such cases the clinical phenotype differs. This variable clinical expression could be the result of abnormalities in other connective tissue proteins. Since decorin is a component of connective tissue, binds to type I collagen fibrils and plays a role in matrix assembly, we studied decorin production in skin fibroblasts from OI patients. Cultured fibroblasts from one patient with extremely severe osteogenesis imperfecta (classified as type II/III) who has an alpha 1(I)gly415ser mutation were found to secrete barely detectable amounts of decorin into culture medium. Western blotting using antibodies raised against decorin confirmed the reduction of the decorin core protein and Northern blot analysis showed decorin mRNA levels below the limit of detection. Cells from a patient, with a less severe phenotype, bearing a mutation in the same position of the triple helix (alpha 1(I)gly415) expressed decorin normally. The different clinical phenotypes could be due to the differing genetic backgrounds of the patients so it is tempting to conclude that in our most severely affected patient the absence of decorin aggravates the clinical phenotype.


Subject(s)
Osteogenesis Imperfecta/genetics , Point Mutation , Proteoglycans/genetics , Blotting, Northern , Blotting, Western , Cells, Cultured , Decorin , Extracellular Matrix Proteins , Female , Fibroblasts/metabolism , Genes, Lethal , Genetic Carrier Screening , Glycine , Humans , Infant, Newborn , Male , Osteogenesis Imperfecta/classification , Osteogenesis Imperfecta/metabolism , Phenotype , Proteoglycans/analysis , Proteoglycans/biosynthesis , RNA, Messenger/biosynthesis , Serine , Skin/metabolism , Skin/pathology
18.
Life Sci ; 59(1): 15-20, 1996.
Article in English | MEDLINE | ID: mdl-8684266

ABSTRACT

An altered equilibrium of protease/protease-inhibitor factors may be involved in the pathogenesis of aneurysm rupture: alpha 1-antitrypsin (alpha 1-AT) represents the most relevant inhibitor of elastase, a proteolytic enzyme enhancing catabolic processes of collagen metabolism. In the present study we test the hypothesis whether the activity of alpha 1-AT is altered in SAH patients; 5 cases with unruptured intracranial aneurysm and 27 patients with diagnosis of aneurysm SAH were included in the study. Blood samples were obtained immediately at admission. As control samples we consider the 5 cases of unruptured aneurysm, 15 cases of unruptured aortic aneurysms and 10 patients with non-vascular CNS diseases. Measurement of alpha 1-AT level was determined by immunoturbidimetric method. Serum levels of alpha 1-AT are significantly lower in patients admitted within 72 hours after SAH, if compared to patients admitted in a delayed phase. The linear relationship between alpha 1-AT and collagenase inhibitory percentage capacity (CIC) was shown to be different in the 4 subgroups considered, and so were the mean % CIC values in the between-groups comparison, except for unruptured aneurysm vs controls. The alpha 1-AT CIC in patients with SAH is shown to be the lowest when compared to controls and unruptured aneurysms (p = 0.0001).


Subject(s)
Collagenases/metabolism , Subarachnoid Hemorrhage/metabolism , alpha 1-Antitrypsin/metabolism , Dose-Response Relationship, Drug , Humans , Intracranial Aneurysm/metabolism , Linear Models , Pancreatic Elastase/metabolism
20.
Hum Mol Genet ; 3(12): 2201-6, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7881420

ABSTRACT

The molecular defects responsible for three cases of severe (type III) osteogenesis imperfecta (OI) were investigated. The mutation sites were localized in pro alpha 1(I) and pro alpha 2(I) mRNA molecules, respectively, by chemical cleavage of mismatch in heteroduplex nucleic acids. Mutation identification was achieved by reverse transcription-polymerase chain reaction-DNA amplification, followed by cloning and sequencing. Two unrelated patients were demonstrated to bear the same G-A transition at nucleotide 2418 of the pro alpha 1(I) coding region, leading to G589S substitution and resulting in very similar clinical manifestations. In the latter patient, a G-T transversion at nucleotide 2166 was found in one pro alpha 2(I) allele, which caused a G586V substitution and again severe OI. Presumably all three mutations occurred de novo in the probands, since they were not found in their parents' DNA. The biochemical findings on type I collagen were very similar in all the probands: the mutations here described had little destabilizing effects on triple helix formation, secretion and stability. The half-life of the collagen incorporated into the insoluble matrix was comparable with that of controls. These mutations are localized in the gap zone of the fibrils where mineral nucleation occurs. This fact suggests that they probably do not exert destabilizing effects on the individual collagen molecules, but rather on the mineralization process, once the defective molecules are incorporated into the fibrils, hence causing severe phenotypes.


Subject(s)
Collagen/genetics , Osteogenesis Imperfecta/genetics , Point Mutation/genetics , Base Sequence , Child , Child, Preschool , Collagen/chemistry , Collagen/metabolism , Female , Glycine/genetics , Humans , Male , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...