Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Genesis ; 58(5): e23357, 2020 05.
Article in English | MEDLINE | ID: mdl-32078250

ABSTRACT

Cystinuria Type A is a relatively common genetic kidney disease occurring in 1 in 7,000 people worldwide that results from mutation of the cystine transporter rBAT encoded by Slc3a1. We used CRISPR/Cas9 technology to engineer cystinuria Type A mice via genome editing of the C57BL/6NHsd background. These mice are an improvement on currently available models as they are on a coisogenic genetic background and have a single defined mutation. In order to use albinism to track Cas9 activity, we co-injected gRNAs targeting Slc3a1 and tyrosinase (Tyr) with Cas9 expressing plasmid DNA into mouse embryos. Two different Slc3a1 mutational alleles were derived, with homozygous mice of both demonstrating elevated urinary cystine levels, cystine crystals, and bladder stones. We used whole genome sequencing to evaluate for potential off-target editing. No off-target indels were observed for the top 10 predicted off-targets for Slc3a1 or Tyr. Therefore, we used CRISPR/Cas9 to generate coisogenic albino cystinuria Type A mice that could be used for in vivo imaging, further study, or developing new treatments of cystinuria.


Subject(s)
Amino Acid Transport Systems, Basic/genetics , Amino Acid Transport Systems, Neutral/genetics , Cystinuria/genetics , Mutation , Animals , CRISPR-Cas Systems , Cysteine/urine , Cystinuria/pathology , Disease Models, Animal , Mice , Mice, Inbred C57BL
2.
Am J Hum Genet ; 101(2): 167-176, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28777929

ABSTRACT

With CRISPR/Cas9 and other genome-editing technologies, successful somatic and germline genome editing are becoming feasible. To respond, an American Society of Human Genetics (ASHG) workgroup developed this position statement, which was approved by the ASHG Board in March 2017. The workgroup included representatives from the UK Association of Genetic Nurses and Counsellors, Canadian Association of Genetic Counsellors, International Genetic Epidemiology Society, and US National Society of Genetic Counselors. These groups, as well as the American Society for Reproductive Medicine, Asia Pacific Society of Human Genetics, British Society for Genetic Medicine, Human Genetics Society of Australasia, Professional Society of Genetic Counselors in Asia, and Southern African Society for Human Genetics, endorsed the final statement. The statement includes the following positions. (1) At this time, given the nature and number of unanswered scientific, ethical, and policy questions, it is inappropriate to perform germline gene editing that culminates in human pregnancy. (2) Currently, there is no reason to prohibit in vitro germline genome editing on human embryos and gametes, with appropriate oversight and consent from donors, to facilitate research on the possible future clinical applications of gene editing. There should be no prohibition on making public funds available to support this research. (3) Future clinical application of human germline genome editing should not proceed unless, at a minimum, there is (a) a compelling medical rationale, (b) an evidence base that supports its clinical use, (c) an ethical justification, and (d) a transparent public process to solicit and incorporate stakeholder input.


Subject(s)
Gene Editing , Genome, Human/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing/ethics , Gene Editing/legislation & jurisprudence , Gene Editing/methods , Humans , Social Change
3.
J Neurosci ; 37(8): 2216-2233, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28130356

ABSTRACT

Characterizing the functional impact of novel mutations linked to autism spectrum disorder (ASD) provides a deeper mechanistic understanding of the underlying pathophysiological mechanisms. Here we show that a de novo Glu183 to Val (E183V) mutation in the CaMKIIα catalytic domain, identified in a proband diagnosed with ASD, decreases both CaMKIIα substrate phosphorylation and regulatory autophosphorylation, and that the mutated kinase acts in a dominant-negative manner to reduce CaMKIIα-WT autophosphorylation. The E183V mutation also reduces CaMKIIα binding to established ASD-linked proteins, such as Shank3 and subunits of l-type calcium channels and NMDA receptors, and increases CaMKIIα turnover in intact cells. In cultured neurons, the E183V mutation reduces CaMKIIα targeting to dendritic spines. Moreover, neuronal expression of CaMKIIα-E183V increases dendritic arborization and decreases both dendritic spine density and excitatory synaptic transmission. Mice with a knock-in CaMKIIα-E183V mutation have lower total forebrain CaMKIIα levels, with reduced targeting to synaptic subcellular fractions. The CaMKIIα-E183V mice also display aberrant behavioral phenotypes, including hyperactivity, social interaction deficits, and increased repetitive behaviors. Together, these data suggest that CaMKIIα plays a previously unappreciated role in ASD-related synaptic and behavioral phenotypes.SIGNIFICANCE STATEMENT Many autism spectrum disorder (ASD)-linked mutations disrupt the function of synaptic proteins, but no single gene accounts for >1% of total ASD cases. The molecular networks and mechanisms that couple the primary deficits caused by these individual mutations to core behavioral symptoms of ASD remain poorly understood. Here, we provide the first characterization of a mutation in the gene encoding CaMKIIα linked to a specific neuropsychiatric disorder. Our findings demonstrate that this ASD-linked de novo CAMK2A mutation disrupts multiple CaMKII functions, induces synaptic deficits, and causes ASD-related behavioral alterations, providing novel insights into the synaptic mechanisms contributing to ASD.


Subject(s)
Autism Spectrum Disorder , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Dendrites/metabolism , Mutation/genetics , Synaptic Transmission/genetics , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/physiopathology , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Cells, Cultured , Cycloheximide/pharmacology , Disease Models, Animal , Embryo, Mammalian , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Exploratory Behavior/physiology , Female , Gene Expression Regulation/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism
4.
Stem Cells ; 34(9): 2342-53, 2016 09.
Article in English | MEDLINE | ID: mdl-27250101

ABSTRACT

Fracture nonunion is a major complication of bone fracture regeneration and repair. The molecular mechanisms that result in fracture nonunion appearance are not fully determined. We hypothesized that fracture nonunion results from the failure of hypoxia and hematoma, the primary signals in response to bone injury, to trigger Bmp2 expression by mesenchymal progenitor cells (MSCs). Using a model of nonstabilized fracture healing in transgenic 5'Bmp2BAC mice we determined that Bmp2 expression appears in close association with hypoxic tissue and hematoma during the early phases of fracture healing. In addition, BMP2 expression is induced when human periosteum explants are exposed to hypoxia ex vivo. Transient interference of hypoxia signaling in vivo with PX-12, a thioredoxin inhibitor, results in reduced Bmp2 expression, impaired fracture callus formation and atrophic-like nonunion by a HIF-1α independent mechanism. In isolated human periosteum-derived MSCs, BMP2 expression could be induced with the addition of platelets concentrate lysate but not with hypoxia treatment, confirming HIF-1α-independent BMP2 expression. Interestingly, in isolated human periosteum-derived mesenchymal progenitor cells, inhibition of BMP2 expression by PX-12 is accomplished only under hypoxic conditions seemingly through dis-regulation of reactive oxygen species (ROS) levels. In conclusion, we provide evidence of a molecular mechanism of hypoxia-dependent BMP2 expression in MSCs where interference with ROS homeostasis specifies fracture nonunion-like appearance in vivo through inhibition of Bmp2 expression. Stem Cells 2016;34:2342-2353.


Subject(s)
Fractures, Ununited/metabolism , Fractures, Ununited/pathology , Homeostasis , Mesenchymal Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Cell Hypoxia/drug effects , Cell Separation , Disulfides/pharmacology , Fracture Healing/drug effects , Homeostasis/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Imidazoles/pharmacology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice, Inbred C57BL , Osteogenesis/drug effects , Oxidative Stress/drug effects , Periosteum/pathology
5.
Development ; 140(22): 4574-82, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24154528

ABSTRACT

Development of the musculoskeletal system requires precise integration of muscles, tendons and bones. The molecular mechanisms involved in the differentiation of each of these tissues have been the focus of significant research; however, much less is known about how these tissues are integrated into a functional unit appropriate for each body position and role. Previous reports have demonstrated crucial roles for Hox genes in patterning the axial and limb skeleton. Loss of Hox11 paralogous gene function results in dramatic malformation of limb zeugopod skeletal elements, the radius/ulna and tibia/fibula, as well as transformation of the sacral region to a lumbar phenotype. Utilizing a Hoxa11eGFP knock-in allele, we show that Hox11 genes are expressed in the connective tissue fibroblasts of the outer perichondrium, tendons and muscle connective tissue of the zeugopod region throughout all stages of development. Hox11 genes are not expressed in differentiated cartilage or bone, or in vascular or muscle cells in these regions. Loss of Hox11 genes disrupts regional muscle and tendon patterning of the limb in addition to affecting skeletal patterning. The tendon and muscle defects in Hox11 mutants are independent of skeletal patterning events as disruption of tendon and muscle patterning is observed in Hox11 compound mutants that do not have a skeletal phenotype. Thus, Hox genes are not simply regulators of skeletal morphology as previously thought, but are key factors that regulate regional patterning and integration of the musculoskeletal system.


Subject(s)
Body Patterning/genetics , Bone and Bones/embryology , Homeodomain Proteins/genetics , Muscles/embryology , Tendons/embryology , Animals , Bone and Bones/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Connective Tissue/embryology , Connective Tissue/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Female , Forelimb/embryology , Forelimb/metabolism , Forelimb/ultrastructure , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Male , Mice , Mice, Mutant Strains , Muscles/metabolism , Mutation/genetics , Osteoblasts/cytology , Osteoblasts/metabolism , Tendons/metabolism
6.
PLoS Genet ; 9(11): e1003929, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24244193

ABSTRACT

Mitochondrial DNA (mtDNA) variation can affect phenotypic variation; therefore, knowing its distribution within and among individuals is of importance to understanding many human diseases. Intra-individual mtDNA variation (heteroplasmy) has been generally assumed to be random. We used massively parallel sequencing to assess heteroplasmy across ten tissues and demonstrate that in unrelated individuals there are tissue-specific, recurrent mutations. Certain tissues, notably kidney, liver and skeletal muscle, displayed the identical recurrent mutations that were undetectable in other tissues in the same individuals. Using RFLP analyses we validated one of the tissue-specific mutations in the two sequenced individuals and replicated the patterns in two additional individuals. These recurrent mutations all occur within or in very close proximity to sites that regulate mtDNA replication, strongly implying that these variations alter the replication dynamics of the mutated mtDNA genome. These recurrent variants are all independent of each other and do not occur in the mtDNA coding regions. The most parsimonious explanation of the data is that these frequently repeated mutations experience tissue-specific positive selection, probably through replication advantage.


Subject(s)
DNA Replication/genetics , DNA, Mitochondrial/genetics , Genome, Mitochondrial , Mutation/genetics , Base Sequence , Humans , Mitochondria/genetics , Muscle, Skeletal/metabolism , Organ Specificity , Polymorphism, Restriction Fragment Length/genetics
7.
Arterioscler Thromb Vasc Biol ; 34(11): 2387-93, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25189570

ABSTRACT

Calcific aortic valve disease (CAVD) is increasingly prevalent worldwide with significant morbidity and mortality. Therapeutic options beyond surgical valve replacement are currently limited. In 2011, the National Heart Lung and Blood Institute assembled a working group on aortic stenosis. This group identified CAVD as an actively regulated disease process in need of further study. As a result, the Alliance of Investigators on CAVD was formed to coordinate and promote CAVD research, with the goals of identifying individuals at risk, developing new therapeutic approaches, and improving diagnostic methods. The group is composed of cardiologists, geneticists, imaging specialists, and basic science researchers. This report reviews the current status of CAVD research and treatment strategies with identification of areas in need of additional investigation for optimal management of this patient population.


Subject(s)
Aortic Valve Stenosis/therapy , Aortic Valve/pathology , Biomedical Research/trends , Calcinosis/therapy , Heart Defects, Congenital/therapy , Heart Valve Diseases/therapy , Aortic Valve/physiopathology , Aortic Valve Stenosis/diagnosis , Aortic Valve Stenosis/physiopathology , Bicuspid Aortic Valve Disease , Calcinosis/diagnosis , Calcinosis/physiopathology , Cardiac Surgical Procedures , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/physiopathology , Heart Valve Diseases/diagnosis , Heart Valve Diseases/physiopathology , Heart Valve Prosthesis Implantation , Hemodynamics/physiology , Humans , Signal Transduction/physiology
8.
Hum Mol Genet ; 20(17): 3517-24, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21653641

ABSTRACT

Multiple sclerosis (MS) is a neurodegenerative, autoimmune disease of the central nervous system, and numerous studies have shown that MS has a strong genetic component. Independent studies to identify MS-associated genes have often indicated multiple signals in physically close genomic regions, although by their proximity it is not always clear if these data indicate redundant or truly independent genetic signals. Recently, three MS study samples were genotyped in parallel using an Illumina Custom BeadChip. These revealed multiple significantly associated single-nucleotide polymorphisms within a 600 kb stretch on chromosome 16p13. Here we present a detailed analysis of variants in this region that clarifies the independent nature of these signals. The linkage disequilibrium patterns in the region and logistic regression analysis of the associations suggest that this region likely harbors three independent MS disease loci. Further, we examined cis-expression QTLs, histone modifications and CCCTC-binding factor (CTCF) binding data in the region. We also tested for correlated expression of the genes from the region using whole-genome expression array data from lymphoblastoid cell lines. Three of the genes show expression correlations across loci. Furthermore, in the GM12878 lymphoblastoid cell line, these three genes are in a continuous region devoid of H3K27 methylation, suggesting an open chromatin configuration. This region likely only contributes minimal risk to MS; however, investigation of this region will undoubtedly provide insight into the functional mechanisms of these genes. These data highlight the importance of taking a closer look at the expression and function of chromosome 16p13 in the pathogenesis of MS.


Subject(s)
Chromosomes, Human, Pair 16/genetics , Lectins, C-Type/genetics , Monosaccharide Transport Proteins/genetics , Multiple Sclerosis/genetics , Suppressor of Cytokine Signaling Proteins/genetics , CCCTC-Binding Factor , Female , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Genotype , Humans , Linkage Disequilibrium/genetics , Logistic Models , Male , Quantitative Trait Loci/genetics , Repressor Proteins/genetics , Suppressor of Cytokine Signaling 1 Protein
9.
J Cell Biol ; 177(6): 1105-17, 2007 Jun 18.
Article in English | MEDLINE | ID: mdl-17576802

ABSTRACT

Despite its clinical significance, joint morphogenesis is still an obscure process. In this study, we determine the role of transforming growth factor beta (TGF-beta) signaling in mice lacking the TGF-beta type II receptor gene (Tgfbr2) in their limbs (Tgfbr2(PRX-1KO)). In Tgfbr2(PRX-1KO) mice, the loss of TGF-beta responsiveness resulted in the absence of interphalangeal joints. The Tgfbr2(Prx1KO) joint phenotype is similar to that in patients with symphalangism (SYM1-OMIM185800). By generating a Tgfbr2-green fluorescent protein-beta-GEO-bacterial artificial chromosome beta-galactosidase reporter transgenic mouse and by in situ hybridization and immunofluorescence, we determined that Tgfbr2 is highly and specifically expressed in developing joints. We demonstrated that in Tgfbr2(PRX-1KO) mice, the failure of joint interzone development resulted from an aberrant persistence of differentiated chondrocytes and failure of Jagged-1 expression. We found that TGF-beta receptor II signaling regulates Noggin, Wnt9a, and growth and differentiation factor-5 joint morphogenic gene expressions. In Tgfbr2(PRX-1KO) growth plates adjacent to interphalangeal joints, Indian hedgehog expression is increased, whereas Collagen 10 expression decreased. We propose a model for joint development in which TGF-beta signaling represents a means of entry to initiate the process.


Subject(s)
Joints/growth & development , Morphogenesis , Signal Transduction , Transforming Growth Factor beta/physiology , Animals , Embryo, Mammalian , Extremities , Joints/chemistry , Joints/embryology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/analysis , Receptors, Transforming Growth Factor beta/deficiency
10.
J Immunol ; 185(3): 1492-501, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20574006

ABSTRACT

Genes, such as IFNG, which are expressed in multiple cell lineages of the immune system, may employ a common set of regulatory elements to direct transcription in multiple cell types or individual regulatory elements to direct expression in individual cell lineages. By employing a bacterial artificial chromosome transgenic system, we demonstrate that IFNG employs unique regulatory elements to achieve lineage-specific transcriptional control. Specifically, a one 1-kb element 30 kb upstream of IFNG activates transcription in T cells and NKT cells but not in NK cells. This distal regulatory element is a Runx3 binding site in Th1 cells and is needed for RNA polymerase II recruitment to IFNG, but it is not absolutely required for histone acetylation of the IFNG locus. These results support a model whereby IFNG uses cis-regulatory elements with cell type-restricted function.


Subject(s)
Cell Lineage/genetics , Cell Lineage/immunology , Gene Expression Regulation/immunology , Genetic Loci/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Animals , Cells, Cultured , Conserved Sequence/genetics , Conserved Sequence/immunology , Core Binding Factor Alpha 3 Subunit/metabolism , Humans , Killer Cells, Natural/enzymology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Natural Killer T-Cells/enzymology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Protein Transport/genetics , Protein Transport/immunology , RNA Polymerase II/metabolism , Regulatory Elements, Transcriptional/immunology , Th1 Cells/enzymology , Th1 Cells/immunology , Th1 Cells/metabolism , Transcription Initiation Site
11.
Front Mol Biosci ; 9: 989851, 2022.
Article in English | MEDLINE | ID: mdl-36148008

ABSTRACT

Although mutations in ADAMTS10 have long been known to cause autosomal recessive Weill-Marchesani Syndrome which is characterized by short stature and ocular abnormalities, more recent work has shown that certain mutations in ADAMTS10 cause glaucoma in dogs. In humans, glaucoma is the leading cause of irreversible vision loss that affects tens of millions of people world-wide. Vision loss in glaucoma is a result of neurodegeneration of retinal ganglion cells that form the inner-most layer of the retina and whose axons form the optic nerve which relays visual information to the brain. ADAMTS10 contributes to the formation of microfibrils which sequester latent transforming growth factor ß (TGFß). Among its many biological functions, TGFß promotes the development of retinal ganglion cells and is also known to play other roles in glaucoma pathogenesis. The aim of this study was to test the hypothesis that ADAMTS10 plays a role in retinal ganglion cell development through regulation of TGFß signaling. To this end, Adamts10 expression was targeted for reduction in zebrafish embryos carrying either a fluorescent reporter that labels retinal ganglion cells, or a fluorescent reporter of pSmad3-mediated TGFß family signaling. Loss of adamts10 function in zebrafish embryos reduced retinal ganglion cell reporter fluorescence and prevented formation of an ordered retinal ganglion cell layer. Targeting adamts10 expression also drastically reduced constitutive TGFß signaling in the eye. Direct inhibition of the TGFß receptor reduced retinal ganglion cell reporter fluorescence similar to the effect of targeting adamts10 expression. These findings unveil a previously unknown role for Adamts10 in retinal ganglion cell development and suggest that the developmental role of Adamts10 is mediated by active TGFß family signaling. In addition, our results show for the first time that Adamts10 is necessary for pSmad3-mediated constitutive TGFß family signaling.

12.
Genes (Basel) ; 13(7)2022 07 04.
Article in English | MEDLINE | ID: mdl-35885978

ABSTRACT

The human capacity to speak is fundamental to our advanced intellectual, technological and social development. Yet so very little is known regarding the evolutionary genetics of speech or its relationship with the broader aspects of evolutionary development in primates. In this study, we describe a large family with evolutionary retrograde development of the larynx and wrist. The family presented with severe speech impairment and incremental retrograde elongations of the pisiform in the wrist that limited wrist rotation from 180° to 90° as in primitive primates. To our surprise, we found that a previously unknown primate-specific gene TOSPEAK had been disrupted in the family. TOSPEAK emerged de novo in an ancestor of extant primates across a 540 kb region of the genome with a pre-existing highly conserved long-range laryngeal enhancer for a neighbouring bone morphogenetic protein gene GDF6. We used transgenic mouse modelling to identify two additional GDF6 long-range enhancers within TOSPEAK that regulate GDF6 expression in the wrist. Disruption of TOSPEAK in the affected family blocked the transcription of TOSPEAK across the 3 GDF6 enhancers in association with a reduction in GDF6 expression and retrograde development of the larynx and wrist. Furthermore, we describe how TOSPEAK developed a human-specific promoter through the expansion of a penta-nucleotide direct repeat that first emerged de novo in the promoter of TOSPEAK in gibbon. This repeat subsequently expanded incrementally in higher hominids to form an overlapping series of Sp1/KLF transcription factor consensus binding sites in human that correlated with incremental increases in the promoter strength of TOSPEAK with human having the strongest promoter. Our research indicates a dual evolutionary role for the incremental increases in TOSPEAK transcriptional interference of GDF6 enhancers in the incremental evolutionary development of the wrist and larynx in hominids and the human capacity to speak and their retrogression with the reduction of TOSPEAK transcription in the affected family.


Subject(s)
Growth Differentiation Factor 6 , Speech , Animals , Biological Evolution , Growth Differentiation Factor 6/genetics , Growth Differentiation Factor 6/metabolism , Humans , Mice , Primates/genetics , Regulatory Sequences, Nucleic Acid
13.
J Cell Biochem ; 112(2): 666-74, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268088

ABSTRACT

BMP2 is a morphogen that controls mesenchymal cell differentiation and behavior. For example, BMP2 concentration controls the differentiation of mesenchymal precursors into myocytes, adipocytes, chondrocytes, and osteoblasts. Sequences within the 3'untranslated region (UTR) of the Bmp2 mRNA mediate a post-transcriptional block of protein synthesis. Interaction of cell and developmental stage-specific trans-regulatory factors with the 3'UTR is a nimble and versatile mechanism for modulating this potent morphogen in different cell types. We show here, that an ultra-conserved sequence in the 3'UTR functions independently of promoter, coding region, and 3'UTR context in primary and immortalized tissue culture cells and in transgenic mice. Our findings indicate that the ultra-conserved sequence is an autonomously functioning post-transcriptional element that may be used to modulate the level of BMP2 and other proteins while retaining tissue specific regulatory elements.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Mesenchymal Stem Cells/metabolism , Regulatory Sequences, Nucleic Acid/genetics , 3' Untranslated Regions/genetics , Animals , Aorta/cytology , Bone Morphogenetic Protein 2/genetics , Cell Line , Cells, Cultured , Immunohistochemistry , Mice , Mice, Transgenic , Polymerase Chain Reaction , Regulatory Sequences, Nucleic Acid/physiology
14.
Dev Dyn ; 239(4): 1047-60, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20201106

ABSTRACT

Skeletal formation is an essential and intricately regulated part of vertebrate development. Humans and mice deficient in growth and differentiation factor 6 (Gdf6) have numerous skeletal abnormalities, including joint fusions and cartilage reductions. The expression of Gdf6 is dynamic and in part regulated by distant evolutionarily conserved cis-regulatory elements. radar/gdf6a is a zebrafish ortholog of Gdf6 and has an essential role in embryonic patterning. Here, we show that radar is transcribed in the cells surrounding and between the developing cartilages of the ventral pharyngeal arches, similar to mouse Gdf6. A 312 bp evolutionarily conserved region (ECR5), 122 kilobases downstream, drives expression in a pharyngeal arch-specific manner similar to endogenous radar/gdf6a. Deletion analysis identified a 78 bp region within ECR5 that is essential for transgene activity. This work illustrates that radar is regulated in the pharyngeal arches by a distant conserved element and suggests radar has similar functions in skeletal development in fish and mammals.


Subject(s)
Branchial Region/metabolism , Growth Differentiation Factor 6/genetics , Regulatory Sequences, Nucleic Acid/physiology , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Base Sequence , Branchial Region/embryology , Cloning, Molecular , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Humans , Mice , Molecular Sequence Data , Organ Specificity/genetics , Regulatory Sequences, Nucleic Acid/genetics , Sequence Homology , Takifugu/embryology , Takifugu/genetics
15.
Invest Ophthalmol Vis Sci ; 62(10): 26, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34424262

ABSTRACT

Purpose: Previously, we identified a G661R mutation of ADAMTS10 (a disintegrin-like and metalloprotease with thrombospondin type 1 motif 10) as being disease causative in a colony of Beagles with inherited primary open-angle glaucoma (POAG). Mutations in ADAMTS10 are known to cause Weill-Marchesani syndrome (WMS), which is also caused by mutations in the fibrillin-1 gene (FBN1), suggesting functional linkage between ADAMTS10 and fibrillin-1, the principal component of microfibrils. Here, we established a mouse line with the G661R mutation of Adamts10 (Adamts10G661R/G661R) to determine if they develop features of WMS and alterations of ocular fibrillin microfibrils. Methods: Intraocular pressure (IOP) was measured using a TonoLab rebound tonometer. Central cornea thickness (CCT), anterior chamber depth (ACD) and axial length (AL) of the eye were examined by spectral-domain optical coherence tomography. Sagittal eye sections from mice at postnatal day 10 (P10) and at 3 and 24 months of age were stained with antibodies against fibrillin-1, fibrillin-2, and ADAMTS10. Results: IOP was not elevated in Adamts10G661R/G661R mice. Adamts10G661R/G661R mice had smaller bodies, thicker CCT, and shallower ACD compared to wild-type mice but normal AL. Adamts10G661R/G661R mice displayed persistent fibrillin-2 and enhanced fibrillin-1 immunofluorescence in the lens zonules and in the hyaloid vasculature and its remnants in the vitreous. Conclusions: Adamts10G661R/G661R mice recapitulate the short stature and ocular phenotypes of WMS. The altered fibrillin-1 and fibrillin-2 immunoactivity in Adamts10G661R/G661R mice suggests that the G661R mutation of Adamts10 perturbs regulation of the fibrillin isotype composition of microfibrils in the mouse eye.


Subject(s)
ADAMTS Proteins/genetics , Anterior Chamber/metabolism , DNA/genetics , Fibrillins/metabolism , Glaucoma, Open-Angle/genetics , Microfibrils/metabolism , Mutation , ADAMTS Proteins/metabolism , Animals , DNA Mutational Analysis , Disease Models, Animal , Female , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/physiopathology , Male , Mice , Signal Transduction
16.
Dev Biol ; 327(2): 590-602, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19159624

ABSTRACT

Bone morphogenetic protein 4 (Bmp4) is a multi-functional, developmentally regulated gene that is essential for mouse development, as most Bmp4-null mouse embryos die at the onset of gastrulation and fail to develop mesoderm. Little is known about the transcriptional regulation of Bmp4. To identify potential long-range cis-regulatory elements that direct its complex spatiotemporal expression patterns, we surveyed the mouse Bmp4 locus using two overlapping bacterial artificial chromosome (BAC) reporter transgenes. Our findings indicate that tissue-specific cis-regulatory elements reside greater than 28 kb 5' or 3' to the mouse Bmp4 transcription unit. In addition, comparative analyses identified three noncoding evolutionarily conserved regions (ECRs), spaced around the gene and conserved from mammals to fish, that are maintained in a syntenic group across vertebrates. Deletion of one of these conserved sequences (ECR2) from a BAC transgene revealed a tissue-specific requirement for ECR2 in driving Bmp4 expression in extraembryonic and embryonic mesoderm. Furthermore, a 467 bp mouse sequence containing ECR2 reproducibly directed lacZ minigene expression in mesoderm. Taken together, this shows that an ancient, mesoderm-specific cis-regulatory element resides nearly 50 kb 5' to mouse Bmp4.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Enhancer Elements, Genetic , Evolution, Molecular , Gene Expression Regulation, Developmental , Mesoderm/physiology , Promoter Regions, Genetic , Animals , Base Sequence , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Genes, Reporter , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Sequence Alignment , Transgenes
17.
Biochem Biophys Res Commun ; 392(2): 124-8, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20060812

ABSTRACT

The level of bone morphogenetic protein 2 (BMP2) profoundly influences essential cell behaviors such as proliferation, differentiation, apoptosis, and migration. The spatial and temporal pattern of BMP2 synthesis, particular in diverse embryonic cells, is highly varied and dynamic. We have identified GC-rich sequences within the BMP2 promoter region that strongly repress gene expression. These elements block the activity of a highly conserved, osteoblast enhancer in response to FGF2 treatment. Both positive and negative gene regulatory elements control BMP2 synthesis. Detecting and mapping the repressive motifs is essential because they impede the identification of developmentally regulated enhancers necessary for normal BMP2 patterns and concentration.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Gene Expression Regulation, Developmental , Regulatory Elements, Transcriptional , Animals , Base Sequence , Cell Line , Conserved Sequence , Enhancer Elements, Genetic , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , GC Rich Sequence , Genes, Reporter , HeLa Cells , Humans , Mice , Molecular Sequence Data , Osteoblasts/drug effects , Osteoblasts/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism
18.
Am J Physiol Regul Integr Comp Physiol ; 298(3): R833-42, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20042692

ABSTRACT

The bone morphogenetic protein (BMP) type 2 receptor ligand, Bmp2, is upregulated in the peripheral pulmonary vasculature during hypoxia-induced pulmonary hypertension (PH). This contrasts with the expression of Bmp4, which is expressed in respiratory epithelia throughout the lung. Unlike heterozygous null Bmp4 mice (Bmp4(LacZ/+)), which are protected from the development of hypoxic PH, mice that are heterozygous null for Bmp2 (Bmp2(+/-)) develop more severe hypoxic PH than their wild-type littermates. This is associated with reduced endothelial nitric oxide synthase (eNOS) expression and activity in the pulmonary vasculature of hypoxic Bmp2(+/-) but not Bmp4(LacZ/+) mutant mice. Furthermore, exogenous BMP2 upregulates eNOS expression and activity in intrapulmonary artery and pulmonary endothelial cell preparations, indicating that eNOS is a target of Bmp2 signaling in the pulmonary vasculature. Together, these data demonstrate that Bmp2 and Bmp4 exert opposing roles in hypoxic PH and suggest that the protective effects of Bmp2 are mediated by increasing eNOS expression and activity in the hypoxic pulmonary vasculature.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Signal Transduction/physiology , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 4/genetics , Cell Division/physiology , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Lac Operon , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase Type III/metabolism , Pulmonary Artery/physiology , Pulmonary Circulation/physiology
19.
Stem Cells ; 27(8): 1887-98, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19544445

ABSTRACT

Mesenchymal stem cells (MSC) have a therapeutic potential in patients with fractures to reduce the time of healing and treat nonunions. The use of MSC to treat fractures is attractive for several reasons. First, MSCs would be implementing conventional reparative process that seems to be defective or protracted. Secondly, the effects of MSCs treatment would be needed only for relatively brief duration of reparation. However, an integrated approach to define the multiple regenerative contributions of MSC to the fracture repair process is necessary before clinical trials are initiated. In this study, using a stabilized tibia fracture mouse model, we determined the dynamic migration of transplanted MSC to the fracture site, their contributions to the repair process initiation, and their role in modulating the injury-related inflammatory responses. Using MSC expressing luciferase, we determined by bioluminescence imaging that the MSC migration at the fracture site is time- and dose-dependent and, it is exclusively CXCR4-dependent. MSC improved the fracture healing affecting the callus biomechanical properties and such improvement correlated with an increase in cartilage and bone content, and changes in callus morphology as determined by micro-computed tomography and histological studies. Transplanting CMV-Cre-R26R-Lac Z-MSC, we found that MSCs engrafted within the callus endosteal niche. Using MSCs from BMP-2-Lac Z mice genetically modified using a bacterial artificial chromosome system to be beta-gal reporters for bone morphogenic protein 2 (BMP-2) expression, we found that MSCs contributed to the callus initiation by expressing BMP-2. The knowledge of the multiple MSC regenerative abilities in fracture healing will allow design of novel MSC-based therapies to treat fractures.


Subject(s)
Bone Regeneration/physiology , Fracture Healing/physiology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Tibial Fractures/pathology , Tibial Fractures/therapy , Animals , Animals, Genetically Modified , Bone Morphogenetic Protein 2/biosynthesis , Bony Callus/cytology , Bony Callus/physiology , Female , Humans , Luminescent Proteins , Mesenchymal Stem Cells/cytology , Mice/genetics , Receptors, CXCR4/metabolism
20.
Mol Cell Biol ; 27(8): 2934-51, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17283059

ABSTRACT

Bone morphogenetic protein 2 (encoded by Bmp2) has been implicated as an important signaling ligand for osteoblast differentiation and bone formation and as a genetic risk factor for osteoporosis. To initially survey a large genomic region flanking the mouse Bmp2 gene for cis-regulatory function, two bacterial artificial chromosome (BAC) clones that extend far upstream and downstream of the gene were engineered to contain a lacZ reporter cassette and tested in transgenic mice. Each BAC clone directs a distinct subset of normal Bmp2 expression patterns, suggesting a modular arrangement of distant Bmp2 regulatory elements. Strikingly, regulatory sequences required for Bmp2 expression in differentiating osteoblasts, as well as tooth buds, hair placodes, kidney, and other tissues, are located more than 53 kilobases 3' to the promoter. By testing BACs with engineered deletions across this distant 3' region, we parsed these regulatory elements into separate locations and more closely refined the location of the osteoblast progenitor element. Finally, a conserved osteoblast progenitor enhancer was identified within a 656-bp sequence located 156.3 kilobases 3' from the promoter. The identification of this enhancer should permit further investigation of upstream regulatory mechanisms that control Bmp2 transcription during osteoblast differentiation and are relevant to further studies of Bmp2 as a candidate risk factor gene for osteoporosis.


Subject(s)
Base Pairing/genetics , Bone Morphogenetic Proteins/genetics , Enhancer Elements, Genetic/genetics , Osteoblasts/metabolism , Promoter Regions, Genetic/genetics , Stem Cells/metabolism , Transcription, Genetic , Transforming Growth Factor beta/genetics , 3' Flanking Region/genetics , Animals , Biomarkers , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/metabolism , Bone and Bones/embryology , Bone and Bones/metabolism , Cell Differentiation , Embryo, Mammalian/metabolism , Gene Expression Profiling , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Osteoblasts/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion , Stem Cells/cytology , Tooth/cytology , Transforming Growth Factor beta/metabolism , Transgenes , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL