Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
BMJ Open Qual ; 12(2)2023 06.
Article in English | MEDLINE | ID: mdl-37263736

ABSTRACT

BACKGROUND: Early detection of patients with clinical deterioration admitted to the hospital is critical. The early warning system (EWS) is developed to identify early clinical deterioration. Using individual patient's vital sign records, this bedside score can identify early clinical deterioration, triggering a communication algorithm between nurses and physicians, thereby facilitating early patient intervention. Although various models have been developed and implemented in emergency rooms and paediatric units, data remain sparse on the utility of the EWS in patients admitted to general internal medicine wards and the processes and challenges encountered during the implementation. LOCAL PROBLEM: There is a lack of standardised tools to recognise early deterioration of patient condition. METHODS: This was a quality improvement project piloted in the clinical teaching unit of a tertiary care hospital. Data were collected 24 weeks pre-EWS and 55 weeks post-EWS implementation. A series of Plan, Do, Study, Act cycles were conducted to identify the root cause, develop a driver diagram to understand the drivers of unexpected deaths, run a sham test trial run of the EWS, educate and obtained feedback of clinical care teams involved, assess adherence to the EWS during the pilot project (6 weeks pre-EWS and 6 weeks post-EWS implementation), evaluate outcomes by extending the duration to 24 weeks pre-EWS and 55 weeks post-EWS implementation, and retrospectively review the uptake of the EWS. INTERVENTIONS: Implementation of a standardised protocol to detect deterioration in patient condition. RESULTS: During the pre-EWS implementation phase (24 weeks), there were 4.4 events per week (1.2 septic workups, 1.9 observation unit transfers, 0.7 critical care transfers, 0.13 cardiac arrests and 0.46 per week unexpected deaths). In the post-EWS implementation phase (55 weeks), there were 4.2 events per week (1.0 septic workup, 1.9 observation unit transfers, 0.82 critical care transfers, 0.25 cardiac arrests and 0.25 unexpected deaths). CONCLUSION: The EWS can improve patient care; however, more engagement of stakeholders and electronic vital sign documentation may improve the uptake of the system.


Subject(s)
Clinical Deterioration , Heart Arrest , Child , Humans , Pilot Projects , Retrospective Studies , Hospitalization , Critical Care
2.
Bone ; 155: 116263, 2022 02.
Article in English | MEDLINE | ID: mdl-34826632

ABSTRACT

Many key signaling molecules used to build tissues during embryonic development are re-activated at injury sites to stimulate tissue regeneration and repair. Bone morphogenetic proteins provide a classic example, but the mechanisms that lead to reactivation of BMPs following injury are still unknown. Previous studies have mapped a large "injury response element" (IRE) in the mouse Bmp5 gene that drives gene expression following bone fractures and other types of injury. Here we show that the large mouse IRE region is also activated in both zebrafish tail resection and mechanosensory hair cell injury models. Using the ability to test multiple constructs and image temporal and spatial dynamics following injury responses, we have narrowed the original size of the mouse IRE region by over 100 fold and identified a small 142 bp minimal enhancer that is rapidly induced in both mesenchymal and epithelial tissues after injury. These studies identify a small sequence that responds to evolutionarily conserved local signals in wounded tissues and suggest candidate pathways that contribute to BMP reactivation after injury.


Subject(s)
Bone Morphogenetic Proteins , Zebrafish , Animals , Bone Morphogenetic Proteins/metabolism , Embryonic Development , Mice , Regulatory Sequences, Nucleic Acid , Signal Transduction , Zebrafish/genetics
3.
Elife ; 72018 11 30.
Article in English | MEDLINE | ID: mdl-30499775

ABSTRACT

Vertebrate pelvic reduction is a classic example of repeated evolution. Recurrent loss of pelvic appendages in sticklebacks has previously been linked to natural mutations in a pelvic enhancer that maps upstream of Pitx1. The sequence of this upstream PelA enhancer is not conserved to mammals, so we have surveyed a large region surrounding the mouse Pitx1 gene for other possible hind limb control sequences. Here we identify a new pelvic enhancer, PelB, that maps downstream rather than upstream of Pitx1. PelB drives expression in the posterior portion of the developing hind limb, and deleting the sequence from mice alters the size of several hind limb structures. PelB sequences are broadly conserved from fish to mammals. A wild stickleback population lacking the pelvis has an insertion/deletion mutation that disrupts the structure and function of PelB, suggesting that changes in this ancient enhancer contribute to evolutionary modification of pelvic appendages in nature.


Subject(s)
Biological Evolution , Enhancer Elements, Genetic , Paired Box Transcription Factors/genetics , Pelvis/growth & development , Vertebrates/growth & development , Vertebrates/genetics , Animals , Base Sequence , Chromosomes, Artificial, Bacterial/metabolism , Conserved Sequence , Fishes/embryology , Gene Expression Regulation, Developmental , Genetic Loci , Genome , Hindlimb/growth & development , Lizards/embryology , Mice , Paired Box Transcription Factors/metabolism , Sequence Deletion
4.
Cell ; 164(1-2): 45-56, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26774823

ABSTRACT

Changes in bone size and shape are defining features of many vertebrates. Here we use genetic crosses and comparative genomics to identify specific regulatory DNA alterations controlling skeletal evolution. Armor bone-size differences in sticklebacks map to a major effect locus overlapping BMP family member GDF6. Freshwater fish express more GDF6 due in part to a transposon insertion, and transgenic overexpression of GDF6 phenocopies evolutionary changes in armor-plate size. The human GDF6 locus also has undergone distinctive regulatory evolution, including complete loss of an enhancer that is otherwise highly conserved between chimps and other mammals. Functional tests show that the ancestral enhancer drives expression in hindlimbs but not forelimbs, in locations that have been specifically modified during the human transition to bipedalism. Both gain and loss of regulatory elements can localize BMP changes to specific anatomical locations, providing a flexible regulatory basis for evolving species-specific changes in skeletal form.


Subject(s)
Biological Evolution , Evolution, Molecular , Growth Differentiation Factor 6/genetics , Skeleton/physiology , Vertebrates/genetics , Adaptation, Physiological , Animals , Enhancer Elements, Genetic , Fish Proteins/genetics , Fish Proteins/metabolism , Fresh Water , Growth Differentiation Factor 6/metabolism , Humans , Quantitative Trait Loci , Seawater , Skeleton/anatomy & histology , Smegmamorpha/genetics , Smegmamorpha/physiology , Species Specificity , Vertebrates/classification , Vertebrates/growth & development , Vertebrates/metabolism
5.
Bone ; 77: 31-41, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25886903

ABSTRACT

Bone morphogenetic proteins (BMPs) are key signaling molecules required for normal development of bones and other tissues. Previous studies have shown that null mutations in the mouse Bmp5 gene alter the size, shape and number of multiple bone and cartilage structures during development. Bmp5 mutations also delay healing of rib fractures in adult mutants, suggesting that the same signals used to pattern embryonic bone and cartilage are also reused during skeletal regeneration and repair. Despite intense interest in BMPs as agents for stimulating bone formation in clinical applications, little is known about the regulatory elements that control developmental or injury-induced BMP expression. To compare the DNA sequences that activate gene expression during embryonic bone formation and following acute injuries in adult animals, we assayed regions surrounding the Bmp5 gene for their ability to stimulate lacZ reporter gene expression in transgenic mice. Multiple genomic fragments, distributed across the Bmp5 locus, collectively coordinate expression in discrete anatomic domains during normal development, including in embryonic ribs. In contrast, a distinct regulatory region activated expression following rib fracture in adult animals. The same injury control region triggered gene expression in mesenchymal cells following tibia fracture, in migrating keratinocytes following dorsal skin wounding, and in regenerating epithelial cells following lung injury. The Bmp5 gene thus contains an "injury response" control region that is distinct from embryonic enhancers, and that is activated by multiple types of injury in adult animals.


Subject(s)
Bone Morphogenetic Protein 5/genetics , Fractures, Bone/genetics , Gene Expression/genetics , Regulatory Sequences, Nucleic Acid , Soft Tissue Injuries/genetics , Animals , Humans , Male , Mice, Transgenic
6.
Cell Rep ; 8(6): 1659-1667, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25220463

ABSTRACT

Phosphate concentration is tightly regulated at the cellular and organismal levels. The first metazoan phosphate exporter, XPR1, was recently identified, but its in vivo function remains unknown. In a genetic screen, we identified a mutation in a zebrafish ortholog of human XPR1, xpr1b. xpr1b mutants lack microglia, the specialized macrophages that reside in the brain, and also displayed an osteopetrotic phenotype characteristic of defects in osteoclast function. Transgenic expression studies indicated that xpr1b acts autonomously in developing macrophages. xpr1b mutants display no gross developmental defects that may arise from phosphate imbalance. We constructed a targeted mutation of xpr1a, a duplicate of xpr1b in the zebrafish genome, to determine whether Xpr1a and Xpr1b have redundant functions. Single mutants for xpr1a were viable, and double mutants for xpr1b;xpr1a were similar to xpr1b single mutants. Our genetic analysis reveals a specific role for the phosphate exporter Xpr1 in the differentiation of tissue macrophages.


Subject(s)
Cell Differentiation , Macrophages/cytology , Receptors, G-Protein-Coupled/metabolism , Receptors, Virus/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified/metabolism , Bone Development , Bone Remodeling , Brain/metabolism , Embryo, Nonmammalian/metabolism , Humans , Macrophages/metabolism , Microglia/cytology , Microglia/metabolism , Mutation , Phenotype , Phosphates/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Virus/genetics , Xenotropic and Polytropic Retrovirus Receptor , Zebrafish/genetics , Zebrafish Proteins/genetics
7.
Nat Genet ; 46(7): 748-52, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24880339

ABSTRACT

Hair color differences are among the most obvious examples of phenotypic variation in humans. Although genome-wide association studies (GWAS) have implicated multiple loci in human pigment variation, the causative base-pair changes are still largely unknown. Here we dissect a regulatory region of the KITLG gene (encoding KIT ligand) that is significantly associated with common blond hair color in northern Europeans. Functional tests demonstrate that the region contains a regulatory enhancer that drives expression in developing hair follicles. This enhancer contains a common SNP (rs12821256) that alters a binding site for the lymphoid enhancer-binding factor 1 (LEF1) transcription factor, reducing LEF1 responsiveness and enhancer activity in cultured human keratinocytes. Mice carrying ancestral or derived variants of the human KITLG enhancer exhibit significant differences in hair pigmentation, confirming that altered regulation of an essential growth factor contributes to the classic blond hair phenotype found in northern Europeans.


Subject(s)
Enhancer Elements, Genetic/genetics , Genome-Wide Association Study , Hair Color/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Polymorphism, Single Nucleotide/genetics , Stem Cell Factor/genetics , White People/genetics , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin Pigmentation/genetics
8.
Nature ; 471(7337): 216-9, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21390129

ABSTRACT

Humans differ from other animals in many aspects of anatomy, physiology, and behaviour; however, the genotypic basis of most human-specific traits remains unknown. Recent whole-genome comparisons have made it possible to identify genes with elevated rates of amino acid change or divergent expression in humans, and non-coding sequences with accelerated base pair changes. Regulatory alterations may be particularly likely to produce phenotypic effects while preserving viability, and are known to underlie interesting evolutionary differences in other species. Here we identify molecular events particularly likely to produce significant regulatory changes in humans: complete deletion of sequences otherwise highly conserved between chimpanzees and other mammals. We confirm 510 such deletions in humans, which fall almost exclusively in non-coding regions and are enriched near genes involved in steroid hormone signalling and neural function. One deletion removes a sensory vibrissae and penile spine enhancer from the human androgen receptor (AR) gene, a molecular change correlated with anatomical loss of androgen-dependent sensory vibrissae and penile spines in the human lineage. Another deletion removes a forebrain subventricular zone enhancer near the tumour suppressor gene growth arrest and DNA-damage-inducible, gamma (GADD45G), a loss correlated with expansion of specific brain regions in humans. Deletions of tissue-specific enhancers may thus accompany both loss and gain traits in the human lineage, and provide specific examples of the kinds of regulatory alterations and inactivation events long proposed to have an important role in human evolutionary divergence.


Subject(s)
Biological Evolution , DNA/genetics , Genome, Human/genetics , Human Characteristics , Regulatory Sequences, Nucleic Acid/genetics , Sequence Deletion/genetics , Animals , Brain/anatomy & histology , Brain/metabolism , Chromosomes, Mammalian/genetics , Conserved Sequence/genetics , DNA, Intergenic/genetics , Enhancer Elements, Genetic/genetics , Evolution, Molecular , Genes, Tumor Suppressor , Humans , Male , Mice , Organ Specificity , Pan troglodytes/genetics , Penis/anatomy & histology , Penis/metabolism , Species Specificity , Transgenes/genetics
9.
PLoS Genet ; 4(12): e1000308, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19096511

ABSTRACT

Cartilage and bone are formed into a remarkable range of shapes and sizes that underlie many anatomical adaptations to different lifestyles in vertebrates. Although the morphological blueprints for individual cartilage and bony structures must somehow be encoded in the genome, we currently know little about the detailed genomic mechanisms that direct precise growth patterns for particular bones. We have carried out large-scale enhancer surveys to identify the regulatory architecture controlling developmental expression of the mouse Bmp5 gene, which encodes a secreted signaling molecule required for normal morphology of specific skeletal features. Although Bmp5 is expressed in many skeletal precursors, different enhancers control expression in individual bones. Remarkably, we show here that different enhancers also exist for highly restricted spatial subdomains along the surface of individual skeletal structures, including ribs and nasal cartilages. Transgenic, null, and regulatory mutations confirm that these anatomy-specific sequences are sufficient to trigger local changes in skeletal morphology and are required for establishing normal growth rates on separate bone surfaces. Our findings suggest that individual bones are composite structures whose detailed growth patterns are built from many smaller lineage and gene expression domains. Individual enhancers in BMP genes provide a genomic mechanism for controlling precise growth domains in particular cartilages and bones, making it possible to separately regulate skeletal anatomy at highly specific locations in the body.


Subject(s)
Bone Development , Bone Morphogenetic Protein 5/genetics , Nasal Cartilages/growth & development , Regulatory Sequences, Nucleic Acid , Ribs/growth & development , Animals , Bone Morphogenetic Protein 5/chemistry , Bone Morphogenetic Protein 5/metabolism , Enhancer Elements, Genetic , Gene Expression , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nasal Cartilages/embryology , Nasal Cartilages/metabolism , Protein Structure, Tertiary , Ribs/anatomy & histology , Ribs/embryology , Ribs/metabolism , Signal Transduction
10.
Development ; 135(15): 2543-53, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18579682

ABSTRACT

The Tbx4 transcription factor is crucial for normal hindlimb and vascular development, yet little is known about how its highly conserved expression patterns are generated. We have used comparative genomics and functional scanning in transgenic mice to identify a dispersed group of enhancers controlling Tbx4 expression in different tissues. Two independent enhancers control hindlimb expression, one located upstream and one downstream of the Tbx4 coding exons. These two enhancers, hindlimb enhancer A and hindlimb enhancer B (HLEA and HLEB), differ in their primary sequence, in their precise patterns of activity within the hindlimb, and in their degree of sequence conservation across animals. HLEB is highly conserved from fish to mammals. Although Tbx4 expression and hindlimb development occur at different axial levels in fish and mammals, HLEB cloned from either fish or mouse is capable of driving expression at the appropriate position of hindlimb development in mouse embryos. HLEA is highly conserved only in mammals. Deletion of HLEA from the endogenous mouse locus reduces expression of Tbx4 in the hindlimb during embryogenesis, bypasses the embryonic lethality of Tbx4-null mutations, and produces viable, fertile mice with characteristic changes in the size of bones in the hindlimb but not the forelimb. We speculate that dual hindlimb enhancers provide a flexible genomic mechanism for altering the strength and location of Tbx4 expression during normal development, making it possible to separately modify the size of forelimb and hindlimb bones during vertebrate evolution.


Subject(s)
Bone and Bones/metabolism , Enhancer Elements, Genetic/genetics , Hindlimb/embryology , Hindlimb/metabolism , T-Box Domain Proteins/metabolism , Animals , Binding Sites , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Genome/genetics , Humans , Mice , Mice, Transgenic , Mutation/genetics , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , T-Box Domain Proteins/genetics
11.
Am J Med Genet A ; 143A(7): 699-706, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17345627

ABSTRACT

Bone morphogenetic proteins (BMPs) are a highly conserved class of signaling molecules that induce ectopic cartilage and bone formation in vivo. Dysregulated expression of bone morphogenetic protein 4 (BMP4) is found in the cells of patients who have fibrodysplasia ossificans progressiva (FOP), a genetic disorder of axial and appendicular skeletal malformation and progressive heterotopic ossification. Loss of function mutations in the bone morphogenetic protein 5 (bmp5) gene leading to under-expression of BMP5 cause the murine short ear syndrome, characterized by small malformed ears and a broad range of axial skeletal malformations. We found features reminiscent of both the short ear mouse and FOP in a child with malformed external ears, multiple malformations of the axial skeleton, and progressive heterotopic ossification in the neck and back. We examined BMP mRNA expression in transformed lymphocytes by semi-quantitative RT-PCR and protein expression by ELISA assays and immunohistochemistry. Elevated levels of BMP4 and BMP5 mRNA and protein were detected in the patient's cells while levels of BMP2 mRNA were unchanged. Our data suggest that dysregulated expression of BMP4 and BMP5 genes is associated with an array of human axial skeletal abnormalities similar to the short ear mouse and FOP.


Subject(s)
Bone Morphogenetic Proteins/genetics , Bone and Bones/abnormalities , Ossification, Heterotopic/genetics , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 5 , Bone Morphogenetic Proteins/biosynthesis , Humans , Infant , Ossification, Heterotopic/pathology , Syndrome
12.
J Bone Miner Res ; 21(8): 1238-47, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16869722

ABSTRACT

UNLABELLED: To reveal the ANK complete loss of function phenotype in mice, we generated conditional and null alleles. Mice homozygous for the null allele exhibited widespread joint mineralization, similar in severity to animals harboring the original ank allele. A delayed yet similar phenotype was observed in mice with joint-specific loss of ANK function. INTRODUCTION: The ANK pyrophosphate regulator was originally identified and proposed to play a key role in articular cartilage maintenance based on a single spontaneous mouse mutation (ank) that causes severe generalized arthritis. A number of human mutations have subsequently been reported in the human ortholog (ANKH), some of which produce skull and long bone defects with no apparent defects in joints or articular cartilage. None of the currently known mouse or human mutations clearly eliminate the function of the endogenous gene. MATERIALS AND METHODS: Two new Ank alleles were generated using homologous recombination in mouse embryonic stem (ES) cells. Joint range of motion assays and muCT studies were used to quantitatively assess phenotypic severity in wildtype, heterozygous, and homozygous mice carrying either the null (Anknull) or original (Ankank) allele. A Gdf5-Cre expressing line was crossed to mice harboring the conditional (Ankfloxp) allele to eliminate ANK function specifically in the joints. Histological stains and beta-galactosidase (LACZ) activity were used to determine the correlation between local loss of ANK function and defective joint phenotypes. RESULTS: Anknull/Anknull mice develop severe ectopic postnatal crystal deposition in almost every joint of the body, leading to eventual joint fusion and loss of mobility. The severity of phenotype in these mice is indistinguishable from that of Ankank/Ankank mice. In addition, despite the widespread expression of Ank in many tissues, the specific deletion of Ank in joints also produces joint mineralization and ankylosis. CONCLUSIONS: These studies show that ANK function is required locally in joints to inhibit mineral formation and that the Ank gene plays a key role in postnatal maintenance of joint mobility and function.


Subject(s)
Ankylosis/genetics , Ankylosis/metabolism , Joints/metabolism , Membrane Proteins/physiology , Minerals/metabolism , Alleles , Animals , Ankylosis/pathology , Arthrography , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Gene Targeting , Joints/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Phenotype , Phosphate Transport Proteins , Tomography, X-Ray Computed
13.
Dev Biol ; 284(2): 301-10, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15979607

ABSTRACT

Asymmetric cell division occurs when a mother cell divides to generate two distinct daughter cells, a process that promotes the generation of cellular diversity in metazoans. During Caenorhabditis elegans development, the asymmetric divisions of neural progenitors generate neurons, neural support cells and apoptotic cells. C. elegans HAM-1 is an asymmetrically distributed cortical protein that regulates several of these asymmetric neuroblast divisions. Here, we show that HAM-1 is a novel protein and define residues important for HAM-1 function and distribution to the cell cortex. Our phenotypic analysis of ham-1 mutant embryos suggests that HAM-1 controls only neuroblast divisions that produce apoptotic cells. Moreover, ham-1 mutant embryos contain many unusually large cell-death corpses. An investigation of this corpse phenotype revealed that it results from a reversal of neuroblast polarity. A misplacement of the neuroblast cleavage plane generates daughter cells of abnormal size, with the apoptotic daughters larger than normal. Thus, HAM-1 regulates the position of the cleavage plane, apoptosis and mitotic potential in C. elegans asymmetric cell divisions.


Subject(s)
Apoptosis , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/embryology , Helminth Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Alleles , Amino Acid Sequence , Amino Acid Substitution , Animals , Arginine/metabolism , Aspartic Acid/metabolism , Base Sequence , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/isolation & purification , Cell Division , Cell Size , Consensus Sequence , Conserved Sequence , Green Fluorescent Proteins/metabolism , Helminth Proteins/chemistry , Helminth Proteins/genetics , Helminth Proteins/isolation & purification , Immunohistochemistry , Models, Biological , Molecular Sequence Data , Mutation, Missense , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/isolation & purification , Neurons/cytology , Sequence Deletion , Sequence Homology, Amino Acid , Transgenes
14.
Genome Res ; 13(9): 2069-81, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12915490

ABSTRACT

Regulatory sequences in higher genomes can map large distances from gene coding regions, and cannot yet be identified by simple inspection of primary DNA sequence information. Here we describe an efficient method of surveying large genomic regions for gene regulatory information, and subdividing complex sets of distant regulatory elements into smaller intervals for detailed study. The mouse Gdf6 gene is expressed in a number of distinct embryonic locations that are involved in the patterning of skeletal and soft tissues. To identify sequences responsible for Gdf6 regulation, we first isolated a series of overlapping bacterial artificial chromosomes (BACs) that extend varying distances upstream and downstream of the gene. A LacZ reporter cassette was integrated into the Gdf6 transcription unit of each BAC using homologous recombination in bacteria. Each modified BAC was injected into fertilized mouse eggs, and founder transgenic embryos were analyzed for LacZ expression mid-gestation. The overlapping segments defined by the BAC clones revealed five separate regulatory regions that drive LacZ expression in 11 distinct anatomical locations. To further localize sequences that control expression in developing skeletal joints, we created a series of BAC constructs with precise deletions across a putative joint-control region. This approach further narrowed the critical control region to an area containing several stretches of sequence that are highly conserved between mice and humans. A distant 2.9-kilobase fragment containing the highly conserved regions is able to direct very specific expression of a minimal promoter/LacZ reporter in proximal limb joints. These results demonstrate that even distant, complex regulatory sequences can be identified using a combination of BAC scanning, BAC deletion, and comparative sequencing approaches.


Subject(s)
Bone Morphogenetic Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Chromosome Deletion , Chromosomes, Artificial, Bacterial/genetics , Embryo, Mammalian , Genetic Engineering/methods , Genetic Markers/genetics , Growth Differentiation Factor 6 , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic/genetics , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Transforming Growth Factor beta/genetics , Transgenes/genetics , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...