Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Genet Med ; 25(11): 100928, 2023 11.
Article in English | MEDLINE | ID: mdl-37427568

ABSTRACT

PURPOSE: HOXD13 is an important regulator of limb development. Pathogenic variants in HOXD13 cause synpolydactyly type 1 (SPD1). How different types and positions of HOXD13 variants contribute to genotype-phenotype correlations, penetrance, and expressivity of SPD1 remains elusive. Here, we present a novel cohort and a literature review to elucidate HOXD13 phenotype-genotype correlations. METHODS: Patients with limb anomalies suggestive of SPD1 were selected for analysis of HOXD13 by Sanger sequencing, repeat length analysis, and next-generation sequencing. Literature was reviewed for HOXD13 heterozygotes. Variants were annotated for phenotypic data. Severity was calculated, and cluster and decision-tree analyses were performed. RESULTS: We identified 98 affected members of 38 families featuring 11 different (likely) causative variants and 4 variants of uncertain significance. The most frequent (25/38) were alanine repeat expansions. Phenotypes ranged from unaffected heterozygotes to severe osseous synpolydactyly, with intra- and inter-familial heterogeneity and asymmetry. A literature review provided 160 evaluable affected members of 49 families with SPD1. Computer-aided analysis only corroborated a positive correlation between alanine repeat length and phenotype severity. CONCLUSION: Our findings support that HOXD13-protein condensation in addition to haploinsufficiency is the molecular pathomechanism of SPD1. Our data may, also, facilitate the interpretation of synpolydactyly radiographs by future automated tools.


Subject(s)
Homeodomain Proteins , Syndactyly , Humans , Homeodomain Proteins/genetics , Transcription Factors/genetics , Syndactyly/genetics , Genotype , Phenotype , Pedigree , Alanine/genetics , Mutation
2.
Am J Med Genet A ; 191(4): 1128-1132, 2023 04.
Article in English | MEDLINE | ID: mdl-36708132

ABSTRACT

The clinical diagnosis criteria for CHARGE syndrome have been revised several times in the last 25 years. Variable expressivity and reduced penetrance are known, particularly in mild and familial cases. Therefore, it has been proposed to include the detection of a pathogenic CHD7 variant as a major diagnostic criterion. However, intronic variants not located at the canonical splice site are still underrepresented in mutation databases, often because functional analysis is not performed in the diagnostic setting. Here, we report a two-generation family that did not meet the criteria for CHARGE syndrome, until the molecular findings were taken into account. By exome sequencing, we detected an intronic variant in a male individual, who presented with unilateral external ear malformation, bilateral semicircular canal aplasia, polydactyly, vertebral body fusion and a heart defect. The variant was inherited by his mother, who also had bilateral semicircular canal aplasia additionally to unilateral sensorineural hearing impairment, unilateral mandibular palpebral synkinesia, orofacial cleft, and dysphagia. Using RNA studies, we were able to demonstrate that aberrant splicing occurs at an upstream cryptic splice acceptor site, resulting in a frameshift and premature stop of translation. Our data show causality of the noncanonical intronic CHD7 variant and end the diagnostic odyssey of this unsolved phenotype of the family.


Subject(s)
CHARGE Syndrome , Cleft Lip , Cleft Palate , Male , Humans , CHARGE Syndrome/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Mutation , Frameshift Mutation , RNA Splice Sites , DNA Helicases/genetics , DNA-Binding Proteins/genetics
3.
Clin Genet ; 103(5): 585-589, 2023 05.
Article in English | MEDLINE | ID: mdl-36562171

ABSTRACT

Known hereditary human diseases featuring impaired copper trafficking across cellular membranes involve ATP7A (Menkes disease, occipital horn disease, X-linked spinal muscular atrophy type 3) and ATP7B (Wilson disease). Herein, we report a newborn infant of consanguineous parents with a homozygous pathogenic variant in a highly conserved sequence of SLC31A1, coding for the copper influx transporter 1, CTR1. This missense variant, c.236T > C, was detected by whole exome sequencing. The infant was born with pulmonary hypoplasia and suffered from severe respiratory distress immediately after birth, necessitating aggressive mechanical ventilation. At 2 weeks of age, multifocal brain hemorrhages were diagnosed by cerebral ultrasound and magnetic resonance imaging, together with increased tortuosity of cerebral arteries. Ensuing seizures were only partly controlled by antiepileptic drugs, and the infant became progressively comatose. Laboratory investigations revealed very low serum concentrations of copper and ceruloplasmin. No hair shaft abnormalities were detected by dermatoscopy or light microscopic analyses of embedded hair shafts obtained at 4 weeks of life. The infant died after redirection of care and elective cessation of invasive mechanical ventilation at 1 month of age. This case adds SLC31A1 to the genes implicated in severe hereditary disorders of copper transport in humans.


Subject(s)
Copper Transporter 1 , Hepatolenticular Degeneration , Menkes Kinky Hair Syndrome , Humans , Infant , Infant, Newborn , Ceruloplasmin/genetics , Copper , Copper Transporter 1/genetics , Copper-Transporting ATPases/genetics , Hepatolenticular Degeneration/genetics , Menkes Kinky Hair Syndrome/genetics , Mutation, Missense
4.
J Bone Miner Res ; 37(8): 1580-1591, 2022 08.
Article in English | MEDLINE | ID: mdl-35689455

ABSTRACT

Hereditary hypophosphatemic rickets with hypercalciuria (HHRH) represents an FGF23-independent disease caused by biallelic variants in the solute carrier family 34-member 3 (SLC34A3) gene. HHRH is characterized by chronic hypophosphatemia and an increased risk for nephrocalcinosis and rickets/osteomalacia, muscular weakness, and secondary limb deformity. Biochemical changes, but no relevant skeletal changes, have been reported for heterozygous SLC34A3 carriers. Therefore, we assessed the characteristics of individuals with biallelic and monoallelic SLC34A3 variants. In 8 index patients and 5 family members, genetic analysis was performed using a custom gene panel. The skeletal assessment comprised biochemical parameters, areal bone mineral density (aBMD), and bone microarchitecture. Pathogenic SLC34A3 variants were revealed in 7 of 13 individuals (2 homozygous, 5 heterozygous), whereas 3 of 13 carried monoallelic variants of unknown significance. Whereas both homozygous individuals had nephrocalcinosis, only one displayed a skeletal phenotype consistent with HHRH. Reduced to low-normal phosphate levels, decreased tubular reabsorption of phosphate (TRP), and high-normal to elevated values of 1,25-OH2 -D3 accompanied by normal cFGF23 levels were revealed independently of mutational status. Interestingly, individuals with nephrocalcinosis showed significantly increased calcium excretion and 1,25-OH2 -D3 levels but normal phosphate reabsorption. Furthermore, aBMD Z-score <-2.0 was revealed in 4 of 8 heterozygous carriers, and HR-pQCT analysis showed a moderate decrease in structural parameters. Our findings highlight the clinical relevance also of monoallelic SLC34A3 variants, including their potential skeletal impairment. Calcium excretion and 1,25-OH2 -D3 levels, but not TRP, were associated with nephrocalcinosis. Future studies should investigate the effects of distinct SLC34A3 variants and optimize treatment and monitoring regimens to prevent nephrocalcinosis and skeletal deterioration. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Familial Hypophosphatemic Rickets , Nephrocalcinosis , Calcium/therapeutic use , Familial Hypophosphatemic Rickets/complications , Familial Hypophosphatemic Rickets/diagnostic imaging , Familial Hypophosphatemic Rickets/genetics , Humans , Hypercalciuria/complications , Hypercalciuria/drug therapy , Hypercalciuria/genetics , Nephrocalcinosis/genetics , Phosphates , Sodium-Phosphate Cotransporter Proteins, Type IIc/genetics
5.
Bone ; 113: 29-40, 2018 08.
Article in English | MEDLINE | ID: mdl-29653293

ABSTRACT

Osteoblastic differentiation is a multistep process characterized by osteogenic induction of mesenchymal stem cells, which then differentiate into proliferative pre-osteoblasts that produce copious amounts of extracellular matrix, followed by stiffening of the extracellular matrix, and matrix mineralization by hydroxylapatite deposition. Although these processes have been well characterized biologically, a detailed transcriptional analysis of murine primary calvaria osteoblast differentiation based on RNA sequencing (RNA-seq) analyses has not previously been reported. Here, we used RNA-seq to obtain expression values of 29,148 genes at four time points as murine primary calvaria osteoblasts differentiate in vitro until onset of mineralization was clearly detectable by microscopic inspection. Expression of marker genes confirmed osteogenic differentiation. We explored differential expression of 1386 protein-coding genes using unsupervised clustering and GO analyses. 100 differentially expressed lncRNAs were investigated by co-expression with protein-coding genes that are localized within the same topologically associated domain. Additionally, we monitored expression of 237 genes that are silent or active at distinct time points and compared differential exon usage. Our data represent an in-depth profiling of murine primary calvaria osteoblast differentiation by RNA-seq and contribute to our understanding of genetic regulation of this key process in osteoblast biology.


Subject(s)
Cell Differentiation/genetics , Osteoblasts/physiology , Osteogenesis/genetics , Transcriptome/genetics , Alternative Splicing , Animals , Cells, Cultured , Gene Expression Profiling , Mice , Mice, Inbred C57BL , RNA/analysis , Skull/physiology
6.
J Bone Miner Res ; 30(5): 796-808, 2015 May.
Article in English | MEDLINE | ID: mdl-25407900

ABSTRACT

MicroRNAs play important roles during cell reprogramming and differentiation. In this study, we identified the miR-497∼195 cluster, a member of the miR-15 family, as strongly upregulated with age of postnatal bone development in vivo and late differentiation stages of primary osteoblasts cultured in vitro. Early expression of miR-195-5p inhibits differentiation and mineralization. Microarray analyses along with quantitative PCR demonstrate that miR-195-5p alters the gene regulatory network of osteoblast differentiation and impairs the induction of bone morphogenetic protein (BMP) responsive genes. Applying reporter gene and Western blot assays, we show that miR-195-5p interferes with the BMP/Smad-pathway in a dose-dependent manner. Systematically comparing the changes in mRNA levels in response to miR-195-5p overexpression with the changes observed in the natural course of osteoblast differentiation, we demonstrate that microRNAs of the miR-15 family affect several target genes involved in BMP signaling. Predicted targets including Furin, a protease that cleaves pro-forms, genes encoding receptors such as Acvr2a, Bmp1a, Dies1, and Tgfbr3, molecules within the cascade like Smad5, transcriptional regulators like Ski and Zfp423 as well as Mapk3 and Smurf1 were validated by quantitative PCR. Taken together, our data strongly suggest that miR-497∼195 cluster microRNAs act as intracellular antagonists of BMP signaling in bone cells.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation/genetics , MicroRNAs/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Signal Transduction/genetics , Aging/genetics , Animals , Animals, Newborn , Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Computational Biology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Expression Regulation/drug effects , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Recombinant Proteins/pharmacology , Reproducibility of Results , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology
8.
Am J Hum Genet ; 92(4): 565-74, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23499309

ABSTRACT

We report that hypofunctional alleles of WNT1 cause autosomal-recessive osteogenesis imperfecta, a congenital disorder characterized by reduced bone mass and recurrent fractures. In consanguineous families, we identified five homozygous mutations in WNT1: one frameshift mutation, two missense mutations, one splice-site mutation, and one nonsense mutation. In addition, in a family affected by dominantly inherited early-onset osteoporosis, a heterozygous WNT1 missense mutation was identified in affected individuals. Initial functional analysis revealed that altered WNT1 proteins fail to activate canonical LRP5-mediated WNT-regulated ß-catenin signaling. Furthermore, osteoblasts cultured in vitro showed enhanced Wnt1 expression with advancing differentiation, indicating a role of WNT1 in osteoblast function and bone development. Our finding that homozygous and heterozygous variants in WNT1 predispose to low-bone-mass phenotypes might advance the development of more effective therapeutic strategies for congenital forms of bone fragility, as well as for common forms of age-related osteoporosis.


Subject(s)
Bone Density/genetics , Bone and Bones/pathology , Mutation/genetics , Osteogenesis Imperfecta/genetics , Osteoporosis/genetics , Wnt1 Protein/genetics , Animals , Base Sequence , Cells, Cultured , Child , Child, Preschool , Female , Heterozygote , Humans , Infant, Newborn , LDL-Receptor Related Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Osteoblasts/metabolism , Osteoblasts/pathology , Osteogenesis Imperfecta/pathology , Osteoporosis/pathology , Pedigree , Phenotype , Pregnancy
9.
J Vasc Surg ; 57(6): 1628-36, 1636.e1-3, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23294503

ABSTRACT

OBJECTIVE: Fibrillin-1 hypomorphic mice (mgR/mgR) are accepted as a model of Marfan syndrome. Phenotypic investigations of this mouse have not previously included quantification of phenotypic features and detailed examinations of the histopathology other than in the ascending aorta. METHODS: We developed a quantitative polymerase chain reaction assay to genotype the mice. Necropsy was performed on 50 male mice after natural death. We then sacrificed 10 mgR/mgR and 10 wild-type mice at 14-19 weeks to perform in vivo computed tomographic scans (n = 3) and microscopic examinations (n = 7). Four aortic segments (ascending, descending, pararenal, and infrarenal aorta) were excised. Each segment was divided into four subsegments and analyzed with Van Gieson staining. The number of elastin breaks and internal aortic diameter were determined twice in randomized, blinded fashion. RESULTS: Computed tomographic scans of mgR/mgR mice revealed aneurysm formation in the ascending aorta and kyphoscoliosis. Elastolysis was present in all four aortic segments of mgR/mgR but was rarely observed in wild-type mice (P < .001). The diameter of the ascending aorta was larger in mgR/mgR than in wild-type mice (P = .01), but para- and infrarenal aortic diameter were even smaller in mgR/mgR mice (P < .001 and P = .01, respectively). Exploratory gene expression analysis showed a number of differentially expressed genes with overrepresentation of immune-related functions. Quantitative polymerase chain reaction analysis confirmed upregulation of selected genes in both the ascending aorta and the abdominal aorta. CONCLUSIONS: Our findings suggest that mgR/mgR mice could be a useful model to study aortic abnormalities in segments other than the ascending aorta in order to understand the molecular mechanisms of aortic disease in Marfan syndrome.


Subject(s)
Aorta/abnormalities , Aorta/metabolism , Disease Models, Animal , Marfan Syndrome , Microfilament Proteins/biosynthesis , Animals , Aorta/pathology , Fibrillin-1 , Fibrillins , Male , Mice , Microfilament Proteins/genetics
10.
Int J Biochem Cell Biol ; 45(3): 696-705, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23262291

ABSTRACT

Osteoblastic differentiation is controlled by complex interplay of several signaling pathways and associated key transcription factors, as well as by microRNAs (miRNAs). In our current study, we found miR-181a to be highly upregulated during BMP induced osteoblastic differentiation of C2C12 and MC3T3 cells. Overexpression of miR-181a led to upregulation of key markers of osteoblastic differentiation as well as enhanced ALP levels and Alizarin red staining, indicating the importance of this miRNA for osteoblastic differentiation. Further, we show that miR-181 isoforms (181a, 181b, 181c) are expressed during different stages of mouse calvarial and tibial development, implying their role in both endochondral and intramembranous ossification. We found several direct and indirect targets of miR-181a to be downregulated by global mRNA expression profiling. Our results demonstrate that miR-181a promotes osteoblastic differentiation via repression of TGF-ß signaling molecules by targeting the negative regulator of osteoblastic differentiation Tgfbi (Tgf-beta induced) and TßR-I/Alk5 (TGF-ß type I receptor). Furthermore, our findings suggest that Rgs4 and Gata6 are direct targets of miR-181a. Taken together, we provide evidence for a crucial functional link between a specific miRNA, miR-181a and osteoblastic differentiation.


Subject(s)
Cell Differentiation , MicroRNAs/genetics , Osteoblasts , Transforming Growth Factor beta/genetics , Animals , Bone Morphogenetic Proteins/metabolism , GATA6 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Mice , MicroRNAs/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Protein Serine-Threonine Kinases/metabolism , RGS Proteins/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Up-Regulation
11.
Hum Mol Genet ; 22(3): 433-43, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23100322

ABSTRACT

Marfan syndrome (MFS) is an inherited disorder of connective tissue caused by mutations in the gene for fibrillin-1 (FBN1). The complex pathogenesis of MFS involves changes in transforming growth factor beta (TGF-ß) signaling and increased matrix metalloproteinase (MMP) expression. Fibrillin-1 and elastin have repeated Gly-x-x- Pro-Gly (GxxPG) motifs that can induce a number of effects including macrophage chemotaxis and increased MMP activity by induction of signaling through the elastin-binding protein (EBP). In this work, we test the hypothesis that antagonism of GxxPG fragments can suppress disease progression in the Marfan aorta. Fibrillin-1 underexpressing mgR/mgR Marfan mice were treated with weekly intraperitoneal (i.p.) injections of an antibody directed against GxxPG fragments. The treatment was started at 3 weeks of age and continued for 8 weeks. The treatment significantly reduced MMP-2, MMP-9 and pSmad2 activity, as well as fragmentation and macrophage infiltration in the aorta of the mgR/mgR mice. Additionally, airspace enlargement and increased pSmad2 activity in the lungs of mgR/mgR animals were prevented by the treatment. Our findings demonstrate the important role of secondary cellular events caused by GxxPG-containing fragments and matrix-induced inflammatory activity in the pathogenesis of thoracic aortic aneurysm (TAA) in mgR/mgR mice. Moreover, the results of the current study suggest that antagonism of the effects of GxxPG fragments may be a fruitful therapeutic strategy in MFS.


Subject(s)
Antibodies, Monoclonal/pharmacology , Aortic Diseases/genetics , Marfan Syndrome/genetics , Peptides/antagonists & inhibitors , Amino Acid Motifs , Animals , Aortic Aneurysm, Thoracic/etiology , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/pathology , Aortic Diseases/complications , Aortic Diseases/drug therapy , Blotting, Western , Disease Models, Animal , Elastin/genetics , Elastin/metabolism , Enzyme-Linked Immunosorbent Assay , Fibrillin-1 , Fibrillins , Immunohistochemistry , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , Macrophages , Marfan Syndrome/complications , Marfan Syndrome/drug therapy , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mutation , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Up-Regulation
12.
Aorta (Stamford) ; 1(1): 5-12, 2013 Jun.
Article in English | MEDLINE | ID: mdl-26798667

ABSTRACT

BACKGROUND: Marfan syndrome (MFS), an inherited disorder of connective tissue characterized by abnormalities in the skeletal, ocular, and cardiovascular systems, is caused by mutations in the gene for fibrillin-1 (FBN1). The high mortality in untreated patients is primarily due to aneurysm and dissection of the ascending aorta. The complex pathogenesis of MFS involves changes in transforming growth factor ß (TGF-ß) signaling, increased matrix metalloproteinase (MMP) expression, and fragmentation of the extracellular matrix. A number of studies have demonstrated increased counts of macrophages and T cells in the ascending aorta of persons or mouse models of MFS, but the efficacy of anti-inflammatory therapy in mouse models of MFS has not yet been assessed. METHODS: FBN1 underexpressing mgR/mgR Marfan mice were treated with oral indomethacin. Treatment was begun at the age of three weeks and continued for 8 weeks, following which the aorta of wild type as well as treated and untreated mgR/mgR mice was compared. RESULTS: Indomethacin treatment led to a statistically significant reduction of aortic elastin degeneration and macrophage infiltration, as well as a lessening of MMP-2, MMP-9, and MMP-12 upregulation. Additionally, indomethacin decreased both cyclooxygenases 2 (COX-2) expression and activity in the aorta of mgR/mgR mice. COX-2-mediated inflammatory infiltrate contributes to the progression of aortic aneurysm in mgR/mgR mice, providing evidence that COX-2 is a relevant therapeutic target in MFS-associated aortic aneurysmal disease. CONCLUSIONS: COX-2 mediated inflammatory infiltration plays an important role in the pathogenesis of aortic aneurysm disease in MFS.

13.
J Med Genet ; 49(7): 437-41, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22717651

ABSTRACT

BACKGROUND: Cleidocranial dysplasia (CCD) is an autosomal dominant skeletal disorder characterised by hypoplastic or absent clavicles, increased head circumference, large fontanels, dental anomalies and short stature. Although CCD is usually caused by mutations leading to haploinsufficiency of RUNX2, the underlying genetic cause remains unresolved in about 25% of cases. METHODS: Array comparative genomic hybridisation was performed to detect copy number variations (CNVs). Identified CNVs were characterised by quantitative PCR and sequencing analyses. The effect of candidate genes on mineralisation was evaluated using viral overexpression in chicken cells. RESULTS: In 2 out of 16 cases, the authors identified microduplications upstream of MSX2 on chromosome 5q35.2. One of the unrelated affected individuals presented with a phenocopy of CCD. In addition to a classical CCD phenotype, the other subject had a complex synpolydactyly of the hands and postaxial polydactyly of the feet which have so far never been reported in association with CCD or CNVs on 5q35.2. The duplications overlap in an ∼219 kb region that contains several highly conserved non-coding elements which are likely to be involved in MSX2 gene regulation. Functional analyses demonstrated that the inhibitory effect of Msx2 overexpression on mineralisation cannot be ameliorated by forced Runx2 expression. CONCLUSIONS: These results indicate that CNVs in non-coding regions can cause developmental defects, and that the resulting phenotype can be distinct from those caused by point mutations within the corresponding gene. Taken together, these findings reveal an additional mechanism for the pathogenesis of CCD, particularly with regard to the regulation of MSX2.


Subject(s)
Chromosome Duplication , Cleidocranial Dysplasia/genetics , Homeodomain Proteins/genetics , Phenotype , Animals , Cells, Cultured , Chickens , Child , Child, Preschool , Chromosomes, Human, Pair 5/genetics , Cleidocranial Dysplasia/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , DNA Copy Number Variations , Female , Gene Expression Regulation , Haploinsufficiency , Heterozygote , Homeodomain Proteins/metabolism , Humans , Male , Oligonucleotide Array Sequence Analysis , Point Mutation , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
14.
BMC Genomics ; 12: 158, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21435219

ABSTRACT

BACKGROUND: The sheep is an important model organism for many types of medically relevant research, but molecular genetic experiments in the sheep have been limited by the lack of knowledge about ovine gene sequences. RESULTS: Prior to our study, mRNA sequences for only 1,556 partial or complete ovine genes were publicly available. Therefore, we developed a composite de novo transcriptome assembly method for next-generation sequence data to combine known ovine mRNA and EST sequences, mRNA sequences from mouse and cow, and sequences assembled de novo from short read RNA-Seq data into a composite reference transcriptome, and identified transcripts from over 12 thousand previously undescribed ovine genes. Gene expression analysis based on these data revealed substantially different expression profiles in standard versus delayed bone healing in an ovine tibial osteotomy model. Hundreds of transcripts were differentially expressed between standard and delayed healing and between the time points of the standard and delayed healing groups. We used the sheep sequences to design quantitative RT-PCR assays with which we validated the differential expression of 26 genes that had been identified by RNA-seq analysis. A number of clusters of characteristic expression profiles could be identified, some of which showed striking differences between the standard and delayed healing groups. Gene Ontology (GO) analysis showed that the differentially expressed genes were enriched in terms including extracellular matrix, cartilage development, contractile fiber, and chemokine activity. CONCLUSIONS: Our results provide a first atlas of gene expression profiles and differentially expressed genes in standard and delayed bone healing in a large-animal model and provide a number of clues as to the shifts in gene expression that underlie delayed bone healing. In the course of our study, we identified transcripts of 13,987 ovine genes, including 12,431 genes for which no sequence information was previously available. This information will provide a basis for future molecular research involving the sheep as a model organism.


Subject(s)
Fracture Healing/genetics , Gene Expression Profiling/methods , Sheep, Domestic/genetics , Animals , Bone and Bones/metabolism , Cattle , Cluster Analysis , Computational Biology/methods , Disease Models, Animal , Female , Mice , Sequence Analysis, DNA/methods
15.
PLoS One ; 6(1): e16250, 2011 Jan 31.
Article in English | MEDLINE | ID: mdl-21305018

ABSTRACT

Elastin production is characteristically turned off during the maturation of elastin-rich organs such as the aorta. MicroRNAs (miRNAs) are small regulatory RNAs that down-regulate target mRNAs by binding to miRNA regulatory elements (MREs) typically located in the 3' UTR. Here we show a striking up-regulation of miR-29 and miR-15 family miRNAs during murine aortic development with commensurate down-regulation of targets including elastin and other extracellular matrix (ECM) genes. There were a total of 14 MREs for miR-29 in the coding sequences (CDS) and 3' UTR of elastin, which was highly significant, and up to 22 miR-29 MREs were found in the CDS of multiple ECM genes including several collagens. This overrepresentation was conserved throughout mammalian evolution. Luciferase reporter assays showed synergistic effects of miR-29 and miR-15 family miRNAs on 3' UTR and coding-sequence elastin constructs. Our results demonstrate that multiple miR-29 and miR-15 family MREs are characteristic for some ECM genes and suggest that miR-29 and miR-15 family miRNAs are involved in the down-regulation of elastin in the adult aorta.


Subject(s)
3' Untranslated Regions , Aorta/growth & development , Down-Regulation/genetics , Elastin/biosynthesis , MicroRNAs/physiology , Open Reading Frames , Regulatory Elements, Transcriptional/genetics , Animals , Binding Sites , Elastin/genetics , Extracellular Matrix/genetics , Gene Expression Regulation, Developmental , Mice , MicroRNAs/genetics , MicroRNAs/metabolism
16.
Nat Genet ; 42(10): 827-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20802478

ABSTRACT

Hyperphosphatasia mental retardation (HPMR) syndrome is an autosomal recessive form of mental retardation with distinct facial features and elevated serum alkaline phosphatase. We performed whole-exome sequencing in three siblings of a nonconsanguineous union with HPMR and performed computational inference of regions identical by descent in all siblings to establish PIGV, encoding a member of the GPI-anchor biosynthesis pathway, as the gene mutated in HPMR. We identified homozygous or compound heterozygous mutations in PIGV in three additional families.


Subject(s)
Exons/genetics , Genetic Predisposition to Disease , Hyperphosphatemia/genetics , Intellectual Disability/genetics , Mannosyltransferases/genetics , Mutation/genetics , Adolescent , Animals , CHO Cells , Child, Preschool , Cricetinae , Cricetulus , Databases, Genetic , Family Health , Female , Glycosylphosphatidylinositols/metabolism , Humans , Infant , Male , Open Reading Frames/genetics , Syndrome , Transfection
17.
J Bone Miner Res ; 24(7): 1247-62, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19257815

ABSTRACT

Whereas mechanical stimulation is essential for bone homeostasis, straining of larger magnitude promotes bone regeneration by directing cell differentiation and proliferation and influencing the gene expression patterns of osteoblasts, which play a vital role in fracture healing by producing and mineralizing osteoid matrix. To elucidate the molecular mechanisms underlying the response of osteoblasts to mechanical strains comparable to those occurring during bone regeneration, MC3T3 S4 (MC4) osteoblast-like cells were stretched in vitro. Analysis based on microarray expression profiling during the first 8 h after straining showed 674 differentially expressed genes. The response to mechanical strain can be divided in an immediate-early response (IER) and later responses. Examination of the approximately 40 genes differentially expressed within the first 60 min, including 11 involved in regulating gene transcription, showed both promiscuous IER genes such as Fos that are upregulated by multiple extracellular stimuli, as well as a number of genes previously shown in neurons to be induced preferentially by depolarization (IPD-IER). Selected differentially expressed genes were validated after mechanical straining and KCl-induced depolarization. The effects of inhibitors for protein kinase A, mitogen-activated protein kinase, and calcineurin pathways were assessed in separate experiments by quantitative RT-PCR and shown to have differential effects on the response of MC4 cells and primary calvaria osteoblasts to both mechanical straining and KCl-induced depolarization. Therefore, our results showed the existence of two distinct pathways that mediate the IER of osteoblasts to large-magnitude mechanical straining and suggest that the IER to depolarizing stimuli is conserved in cell types as different as osteoblasts and neurons.


Subject(s)
Bone Regeneration/physiology , Cell Differentiation/physiology , Cell Polarity/physiology , Gene Expression Regulation/physiology , Osteoblasts/metabolism , Stress, Physiological/physiology , Animals , Bone Regeneration/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Polarity/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Mice , Osteoblasts/cytology , Potassium Chloride/pharmacology , Stress, Physiological/drug effects , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...