Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Osteoarthritis Cartilage ; 27(7): 979-993, 2019 07.
Article in English | MEDLINE | ID: mdl-31028883

ABSTRACT

OBJECTIVE: Falls are common after total hip arthroplasty (THA) and total knee arthroplasty (TKA). While previous studies have investigated various risk factors for falls in patients following THA and TKA, no systematic reviews have summarized these risk factors. Therefore, the current systematic review aimed to summarize evidence regarding risk factors for falls in patients after THA and/or TKA. METHODS: MEDLINE, EMBASE, CINAHL, SPORTDiscus, and Physiotherapy Evidence Database (from inception to June 30, 2018) were searched. The methodological quality and quality of evidence of the included studies were assessed by two independent reviewers. Relevant data regarding participants' characteristics, study design, follow-up time points, and identified risk factors were extracted. Meta-analyses and narrative syntheses were performed. RESULTS: Twelve studies with a total of 1,292,689 participants were included. Twenty-nine identified risk factors for post-THA/TKA falls were classified into either inpatient or post-discharge risk factors. Key risk factors for both post-THA and/or post-TKA inpatient falls that showed moderate level of evidence included: postoperative complications or comorbidities and revision THA/TKA. Likewise, risk factors for post-discharge falls after THA and/or TKA that demonstrated moderate level of evidence included: medications, psychiatric diseases, living alone, prior history of TKA, falls history and female gender. The quality of the included studies varied and sample sizes were not justified. CONCLUSIONS: This review summarized both non-modifiable and modifiable risk factors for post-THA/TKA falls. Our findings highlight the importance of developing strategies to lower the falls risk among patients following THA/TKA.


Subject(s)
Accidental Falls/statistics & numerical data , Arthroplasty, Replacement, Hip/methods , Arthroplasty, Replacement, Knee/methods , Osteoarthritis, Hip/surgery , Osteoarthritis, Knee/surgery , Accidental Falls/prevention & control , Age Factors , Aged , Arthroplasty, Replacement, Hip/adverse effects , Arthroplasty, Replacement, Knee/adverse effects , Female , Humans , Male , Middle Aged , Osteoarthritis, Hip/diagnostic imaging , Osteoarthritis, Knee/diagnostic imaging , Prevalence , Prognosis , Risk Assessment , Severity of Illness Index , Sex Factors
2.
Ultrason Sonochem ; 26: 163-168, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25682465

ABSTRACT

The stochastic behavior of cavitation can lead to major problems of initiation and maintenance of cavitation during sonication, responsible of poor reproducibility of US-induced bioeffects in the context of sonoporation for instance. To overcome these disadvantages, the injection of ultrasound contrast agents as cavitation nuclei ensures fast initiation and lower acoustic intensities required for cavitation activity. More recently, regulated-cavitation devices based on the real-time modulation of the applied acoustic intensity have shown their potential to maintain a stable cavitation state during an ultrasonic shot, in continuous or pulsed wave conditions. In this paper is investigated the interest, in terms of cavitation activity, of using such regulated-cavitation device or injecting ultrasound contrast agents in the sonicated medium. When using fixed applied acoustic intensity, results showed that introducing ultrasound contrast agents increases reproducibility of cavitation activity (coefficient of variation 62% and 22% without and with UCA, respectively). Moreover, the use of the regulated-cavitation device ensures a given cavitation activity (coefficient of variation less 0.4% in presence of UCAs or not). This highlights the interest of controlling cavitation over time to free cavitation-based application from the use of UCAs. Interestingly, during a one minute sonication, while ultrasound contrast agents progressively disappear, the regulated-cavitation device counterbalance their destruction to sustain a stable inertial cavitation activity.

3.
Gene Ther ; 17(10): 1225-33, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20520649

ABSTRACT

Different members of the galectin family may have inhibitory or stimulatory roles in controlling immune responses and regulating inflammatory reactions in autoimmune diseases such as rheumatoid arthritis (RA). A hypothetical model of a cross talk between galectin-1 and galectin-3 has been established in the circumstance of rheumatoid joints. As galectin-3 is a positive regulator and galectin-1 is a negative regulator of inflammation and autoimmune responses, in this study we evaluated the effects of local knockdown of galectin-3 or overexpression of galectin-1 on ameliorating collagen-induced arthritis (CIA) in rats. Lentiviral vectors encoding galectin-3 small hairpin RNA (shRNA) and galectin-1, as well as two control vectors expressing luciferase shRNA and green fluorescent protein, were individually injected intra-articularly into the ankle joints of rats with CIA, and their treatment responses were monitored by measuring the clinical, radiological and histological changes. Our results show that both knockdown of galectin-3 and overexpression of galectin-1 induced higher percentages of antigen-induced T-cell death in the lymph node cells from arthritic rats. Furthermore, these treatments significantly reduced articular index scores, radiographic scores and histological scores, accompanied with decreased T-cell infiltrates and reduced microvessel density in the ankle joints. Our findings implicate galectin-3 and galectin-1 as potential therapeutic targets for the treatment of RA.


Subject(s)
Arthritis, Experimental/therapy , Galectin 1/genetics , Galectin 3/genetics , Genetic Vectors/administration & dosage , Lentivirus/genetics , RNA, Small Interfering/genetics , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/therapy , Cells, Cultured , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Injections, Intra-Articular , Rats , Transfection
4.
Proc Natl Acad Sci U S A ; 105(24): 8298-303, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18541910

ABSTRACT

Bves is an integral membrane protein with no determined function and no homology to proteins outside of the Popdc family. It is widely expressed throughout development in myriad organisms. Here, we demonstrate an interaction between Bves and guanine nucleotide exchange factor T (GEFT), a GEF for Rho-family GTPases. This interaction represents the first identification of any protein that has a direct physical interaction with any member of the Popdc family. Bves and GEFT are shown to colocalize in adult skeletal muscle. We also demonstrate that exogenous expression of Bves reduces Rac1 and Cdc42 activity levels while not affecting levels of active RhoA. Consistent with a repression of Rac1 and Cdc42 activity, we show changes in speed of cell locomotion and cell roundness also result from exogenous expression of Bves. Modulation of Rho-family GTPase signaling by Bves would be highly consistent with previously described phenotypes occurring upon disruption of Bves function in a wide variety of model systems. Therefore, we propose Bves as a novel regulator of the Rac1 and Cdc42 signaling cascades.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Movement , Cell Shape , Guanine Nucleotide Exchange Factors/metabolism , Muscle Cells/metabolism , Muscle Proteins/metabolism , Neuropeptides/metabolism , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Movement/genetics , Cell Shape/genetics , Cytoplasm/metabolism , DNA Mutational Analysis , Guanine Nucleotide Exchange Factors/genetics , Mice , Muscle Cells/cytology , Muscle Proteins/genetics , NIH 3T3 Cells , Neuropeptides/genetics , Protein Interaction Domains and Motifs/genetics , Rho Guanine Nucleotide Exchange Factors , Sequence Deletion , Two-Hybrid System Techniques , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
5.
Am J Physiol Heart Circ Physiol ; 294(6): H2701-11, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18424640

ABSTRACT

We recovered a novel mouse mutant exhibiting neonatal lethality associated with severe fetal cardiac hypertrophy and with some adult mice dying suddenly with left ventricular hypertrophic cardiomyopathy. Using Doppler echocardiography, we screened surviving adult mice in this mutant line for cardiac hypertrophy. Cardiac dimensions were obtained either from two-dimensional images collected using a novel ECG-gated ultra-high-frequency ultrasound system or by traditional M-mode imaging on a clinical ultrasound system. These analyses identified, among the littermates, two populations of mice: those with apparent cardiac hypertrophy with hypercontractile function characterized by ejection fraction of 75-80%, and normal littermates with ejection fraction of 53-55%. Analysis of the ECG-gated two-dimensional cines indicated that the hypertrophy was of the nonobstructive type. Further analysis of heart-to-body weight ratio confirmed the ultrasound diagnosis of left ventricular hypertrophic cardiomyopathy. Histopathology showed increased ventricular wall thickness, enlarged myocyte size, and mild myofiber disarray. Ultrastructural analysis by electron microscopy revealed mitochondria hyperproliferation and dilated sarcoplasmic reticulum. Genome scanning using microsatellite DNA markers mapped the mutation to the X chromosome. DNA sequencing showed no mutations in the coding regions of several candidate genes on the X chromosome, including several known to be associated with left ventricular hypertrophic cardiomyopathy. These findings suggest that this mouse line may harbor a mutation in a novel gene causing X-linked cardiomyopathy.


Subject(s)
Genetic Diseases, X-Linked/genetics , Hypertrophy, Left Ventricular/genetics , Mutation , X Chromosome , Aging , Animals , Chromosome Mapping , Disease Models, Animal , Echocardiography, Doppler, Color , Genetic Diseases, X-Linked/pathology , Genetic Diseases, X-Linked/physiopathology , Heart Ventricles/ultrastructure , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Microsatellite Repeats , Microscopy, Electron , Mitochondria, Heart/ultrastructure , Myocardial Contraction/genetics , Myocytes, Cardiac/ultrastructure , Sarcoplasmic Reticulum/ultrastructure , Stroke Volume/genetics , Ventricular Function, Left/genetics
6.
Ultrasound Med Biol ; 34(5): 741-52, 2008 May.
Article in English | MEDLINE | ID: mdl-18328616

ABSTRACT

To establish a developmental profile of fetal mouse cardiovascular parameters, we analyzed a large body of ultrasound measurements obtained by in utero echocardiography of C57BL/6J fetal mice from embryonic day 12.5 to 19.5 (term). Measurements were obtained using two-dimensional (2D), spectral Doppler and M-mode imaging with standard clinical cardiac ultrasound imaging planes. As these studies were conducted as part of a large scale mouse mutagenesis screen, stringent filtering criteria were used to eliminate potentially abnormal fetuses. Our analysis showed heart rate increased from 190 to 245 beats per minute as the mouse fetus grew from 8 mm at embryonic day 12.5 to 18.7 mm at term. This was accompanied by increases in peak outflow velocity, E-wave, E/A ratio and ventricular dimensions. In contrast, the A-wave, myocardial performance index and isovolemic contraction time decreased gradually. Systolic function remained remarkably stable at 80% ejection fraction. Analysis of intra- and interobserver variabilities showed these parameters were reproducible, with most comparing favorably to clinical ultrasound measurements in human fetuses. A comprehensive database was generated comprising 23 echocardiographic parameters delineating fetal mouse cardiovascular function from embryonic day 12.5 to term. This database can serve as a standard for evaluating cardiovascular pathophysiology in genetically altered and mutant mouse models.


Subject(s)
Fetal Heart/diagnostic imaging , Ultrasonography, Prenatal/methods , Animals , Echocardiography, Doppler , Female , Fetal Heart/embryology , Gestational Age , Heart Rate, Fetal , Mice , Mice, Inbred C57BL , Models, Animal , Pregnancy , Regional Blood Flow , Signal Processing, Computer-Assisted , Stroke Volume , Systole
7.
Cancer Gene Ther ; 14(11): 904-17, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17704754

ABSTRACT

DNA vaccine and dendritic cells (DCs)-based vaccine have emerged as promising strategies for cancer immunotherapy. Fms-like tyrosine kinase 3-ligand (Flt3L) and granulocyte-macrophage-colony-stimulating factor (GM-CSF) have been exploited for the expansion of DC. It was reported previously that combination of plasmid encoding GM-CSF with HER2/neu DNA vaccine induced predominantly CD4(+) T-cell-mediated antitumor immune response. In this study, we investigated the modulation of immune responses by murine Flt3L and GM-CSF, which acted as genetic adjuvants in the forms of bicistronic (pFLAG) and monocistronic (pFL and pGM) plasmids for HER2/neu DNA vaccine (pN-neu). Coexpression of Flt3L and GM-CSF significantly enhanced maturation and antigen-presentation abilities of splenic DC. Increased numbers of infiltrating DC at the immunization site, higher interferon-gamma production, and enhanced cytolytic activities by splenocytes were prominent in mice vaccinated with pN-neu in conjunction with pFLAG. Importantly, a potent CD8(+) T-cell-mediated antitumor immunity against bladder tumors naturally overexpressing HER2/neu was induced in the vaccinated mice. Collectively, our results indicate that murine Flt3L and GM-CSF genes coexpressed by a bicistronic plasmid modulate the class of immune responses and may be superior to those codelivered by two separate monocistronic plasmids as the genetic adjuvants for HER2/neu DNA vaccine.


Subject(s)
Cancer Vaccines/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Membrane Proteins/genetics , Receptor, ErbB-2/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/therapy , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Dendritic Cells/immunology , Genetic Therapy , Genetic Vectors/genetics , Mice , Mice, Inbred Strains , Plasmids/genetics , Spleen/immunology , Tumor Burden , Urinary Bladder Neoplasms/pathology , Vaccination , Vaccines, DNA/genetics , Vaccines, DNA/therapeutic use
8.
Physiol Genomics ; 24(1): 23-36, 2005 Dec 14.
Article in English | MEDLINE | ID: mdl-16174781

ABSTRACT

As part of a large-scale noninvasive fetal ultrasound screen to recover ethylnitrosourea (ENU)-induced mutations causing congenital heart defects in mice, we established a high-throughput ultrasound scanning strategy for interrogating fetal mice in utero utilizing three orthogonal imaging planes defined by the fetus' vertebral column and body axes, structures readily seen by ultrasound. This contrasts with the difficulty of acquiring clinical ultrasound imaging planes which are defined by the fetal heart. By use of the three orthogonal imaging planes for two-dimensional (2D) imaging together with color flow, spectral Doppler, and M-mode imaging, all of the major elements of the heart can be evaluated. In this manner, 10,091 ENU-mutagenized mouse fetuses were ultrasound scanned between embryonic days 12.5 and 19.5, with 324 fetuses found to die prenatally and 425 exhibiting cardiovascular defects. Further analysis by necropsy and histology showed heart defects that included conotruncal anomalies, obstructive lesions, and shunt lesions as well as other complex heart diseases. Ultrasound imaging also identified craniofacial/head defects and body wall closure defects, which necropsy revealed as encephalocele, holoprosencephaly, omphalocele, or gastroschisis. Genome scanning mapped one ENU-induced mutation associated with persistence truncus arteriosus and holoprosencephaly to mouse chromosome 2, while another mutation associated with cardiac defects and omphalocele was mapped to mouse chromosome 17. These studies show the efficacy of this novel ultrasound scanning strategy for noninvasive ultrasound phenotyping to facilitate the recovery of ENU-induced mutations causing congenital heart defects and other extracardiac anomalies.


Subject(s)
Abnormalities, Multiple/diagnostic imaging , Cardiovascular Physiological Phenomena , Ethylnitrosourea/toxicity , Fetus/radiation effects , Heart Defects, Congenital/diagnostic imaging , Mutation , Ultrasonography, Prenatal , Abnormalities, Multiple/embryology , Animals , Female , Heart Defects, Congenital/embryology , Mice , Pregnancy
9.
Calcif Tissue Int ; 74(6): 561-73, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15354864

ABSTRACT

Tbx2 belongs to a family of developmental transcription regulatory factors. We evaluated whether the gap junction protein Connexin43 (Cx43), an important regulator of osteoblast function and bone development, may be a downstream target gene regulated by Tbx2. The Cx43 promoter contains direct repeats of the consensus T-box binding motif, TCACAC, and moreover, Tbx2 and Cx43 show overlapping expression domains in precursors to bone and in osteoblasts. In vitro analysis showed that the Cx43 promoter contains two Tbx2 binding sites, and this binding was dependent on the TCACAC consensus sequence. Transient transfection analysis with a Cx43 promoter-driven lacZ reporter construct revealed negative regulation mediated by these two Tbx2 binding sites in osteoblast-like cells. Thus, downregulation of Tbx2 led to de-repression of wild-type Cx43 promoter activity, whereas a promoter construct with mutated binding sites showed no de-repression. In stably transfected osteosarcoma cells in which expression of the endogenous Tbx2 gene was downregulated with a Tbx2 antisense construct, a marked de-repression of the endogenous Cx43 gene was observed. This was accompanied by a marked increase in the abundance of Cx43 gap junctions and increased functional gap junction-mediated cell-cell communication. Analysis of lacZ expression in transgenic mice containing the mutated Cx43 promoter-driven lacZ construct further suggested de-repression of the Cx43 promoter in limb buds, a region destined to give rise to long bones of the limbs. Taken together, these findings indicate that the promoter of Cx43 is repressible by Tbx2, both in cultured osteoblast-like cells in vitro and likely in the developing embryo.


Subject(s)
Cell Cycle Proteins/genetics , Connexin 43/biosynthesis , Down-Regulation , Osteoblasts/metabolism , T-Box Domain Proteins/genetics , Animals , Base Sequence , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Connexin 43/genetics , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , NIH 3T3 Cells , Osteosarcoma/metabolism , Rats , Sequence Alignment , T-Box Domain Proteins/metabolism , Transcription, Genetic , Transfection
10.
Cell Commun Adhes ; 10(1): 37-50, 2003.
Article in English | MEDLINE | ID: mdl-12881039

ABSTRACT

We examined BMP regulation of the gap junction gene Gjal (Cx43alpha1) using a series of lacZ reporter constructs containing up to 6.7 kbs of mouse Cx43alpha1 promoter sequence. Using transient transfection assays, we showed that BMP2, BMP4, and GDF5, but not BMP6 or BMP7, can modulate Cx43alpha1 promoter activity in the osteosarcoma cell line ROS17/2.8. Positive regulatory elements were found at the proximal and distal ends of the 6.7 kb promoter fragment, while negative regulatory elements were found in the intervening region. Comparison of Cx43alpha1 promoter sequences from the human vs. mouse showed five regions with significant sequence conservation, two of which contained Smad binding elements in conjunction with a BMP response element. Analysis of a transgenic mouse line containing a Cx43alpha1 promoter driven lacZ reporter construct revealed lacZ expression in the developing joints, an expression pattern similar to that previously reported for Gdf5. LacZ expression was also observed in axial regions of the skeletal anlage, which in situ hybridization analysis confirmed as sites of Gdf5 transcript expression. When the Cx43alpha1 promoter driven lacZ transgene was bred into the brachypodism mouse Gdf5(bpJ)(bp) harboring a Gdf5 loss of function mutation, lacZ expression was extinguished. This was observed in homozygous and heterozygous bp animals, suggesting that Cx43alpha1 promoter regulation by GDF5 is subject to haploinsufficiency. Overall, these observations are consistent with recent studies by others indicating a role for Cx43alpha1 in osteogenesis and osteoblastic function during mouse development.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Connexin 43/biosynthesis , Connexin 43/genetics , Gene Expression Regulation, Developmental , Osteoblasts/metabolism , Promoter Regions, Genetic , Transforming Growth Factor beta , Amino Acid Motifs , Animals , Base Sequence , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 6 , Bone Morphogenetic Protein 7 , Conserved Sequence , Genes, Reporter , Growth Differentiation Factor 5 , Heterozygote , Homozygote , Humans , In Situ Hybridization , Lac Operon , Luciferases/metabolism , Mice , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Mutation , Plasmids/metabolism , Protein Binding , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid , Transcription, Genetic , Transfection , beta-Galactosidase/metabolism
11.
Exp Dermatol ; 12(2): 191-7, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12702148

ABSTRACT

Recently, we identified several missense mutations of the connexin gene GJB3 encoding connexin 31 (Cx31) in erythrokeratodermia variabilis (EKV), an autosomal dominant skin disorder. These mutations include G12D, which replaces a conserved glycine residue in the amino-terminus of Cx31 and is associated with a severe EKV phenotype. In contrast, the biologic relevance of the GJB3 sequence variant R32W located in the first transmembrane domain of Cx31 is disputed. To examine the effects of these sequence variants on Cx31 biogenesis and gap junction activity we expressed wild type and mutant Cx31-Flag constructs in HeLa cells. Using immunostaining, all expression variants were detected in the cytoplasm and in a punctate pattern at the cell surface, indicating that G12D and R32W did not interfere with either protein synthesis or transport to the cell membrane. Similarly, oligomerization into hemichannels appeared not impaired when expressing either Cx31 mutant as assessed by size exclusion chromatography, immunoblotting and immunostaining. However, dye transfer experiments and monitoring of intracellular calcium levels in response to serum stimulation revealed that G12D-Cx31 did not form functional gap junction channels, probably due to incorrect assembly or altered properties of Cx31 channels. In contrast, intercellular coupling between cells expressing R32W-Cx31 was comparable to that of wtCx31, suggesting that R32W is a functionally inconsequential polymorphism of Cx31.


Subject(s)
Connexins/genetics , Connexins/physiology , Base Sequence , Calcium Signaling , Connexins/chemistry , DNA, Complementary/genetics , Gap Junctions/physiology , Genetic Variation , HeLa Cells , Humans , In Vitro Techniques , Mutation, Missense , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/physiopathology , Transfection
12.
J Orthop Surg (Hong Kong) ; 10(2): 185-93, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12493933

ABSTRACT

PURPOSE: Owing to unpredictable femoral neck reduction, reconstruction nails are not suitable for fixation of group 3 ipsilateral femoral neck-shaft fractures. We developed a new one-step fixation technique to overcome this problem. This study aims to assess this new technique at the Orthopaedic Department, Chi-Mei Foundation Medical Center, Tainan. METHODS: Of 31 consecutive patients with femoral fractures treated by reconstruction nails, five patients had group 3 ipsilateral femoral neck-shaft fractures, 4 of whom were treated by a new surgical technique. Two 5.0-mm drills were firstly inserted to tether the trochanter fragment, and distal locking screws were secondly applied to immobilise the shaft fracture. The neck-shaft angle was then restored in a closed fashion and proximal cephalomedullary screws were attached. Patients were followed up by post-operative radiography. RESULTS: All 5 cases of group 3 ipsilateral femoral neck-shaft fracture obtained radiographic union without significant surgical sequelae. Three of the patients had implants removed. No patients presented with osteonecrosis at the 3-year follow-up. CONCLUSION: The new approach to manage ipsilateral femoral neck-shaft fractures by using reconstruction nails obtains relatively good clinical results.


Subject(s)
Bone Nails , Femoral Fractures/surgery , Femoral Neck Fractures/surgery , Fracture Fixation, Internal/instrumentation , Adult , Female , Femoral Fractures/diagnostic imaging , Femoral Neck Fractures/diagnostic imaging , Fluoroscopy , Humans , Male , Middle Aged , Treatment Outcome
13.
Development ; 129(8): 2031-42, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11934868

ABSTRACT

Connexin43 knockout mice die neonatally from conotruncal heart malformation and outflow obstruction. Previous studies have indicated the involvement of neural crest perturbations in these cardiac anomalies. We provide evidence for the involvement of another extracardiac cell population, the proepicardial cells. These cells give rise to the vascular smooth muscle cells of the coronary arteries and cardiac fibroblasts in the heart. We have observed the abnormal presence of fibroblast and vascular smooth muscle cells in the infundibular pouches of the connexin43 knockout mouse heart. In addition, the connexin43 knockout mice exhibit a variety of coronary artery patterning defects previously described for neural crest-ablated chick embryos, such as anomalous origin of the coronary arteries, absent left or right coronary artery, and accessory coronary arteries. However, we show that proepicardial cells also express connexin43 gap junctions abundantly. The proepicardial cells are functionally well coupled, and this coupling is significantly reduced with the loss of connexin43 function. Further analysis revealed an elevation in the speed of cell locomotion and cell proliferation rate in the connexin43-deficient proepicardial cells. A parallel analysis of proepicardial cells in transgenic mice with dominant negative inhibition of connexin43 targeted only to neural crest cells showed none of these coupling, proliferation or migration changes. These mice exhibit outflow obstruction, but no infundibular pouches. Together these findings indicate an important role for connexin43 in coronary artery patterning, a role that probably involves the proepicardial and cardiac neural crest cells. We discuss the potential involvement of connexin43 in human cardiovascular anomalies involving the coronary arteries.


Subject(s)
Connexin 43/physiology , Coronary Vessels/embryology , Gap Junctions/metabolism , Neovascularization, Physiologic/physiology , Animals , Biomarkers , Cell Differentiation , Cell Division , Cell Movement , Connexin 43/genetics , Coronary Circulation , Heart , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/biosynthesis , Muscle Proteins/biosynthesis , Muscle, Smooth, Vascular/metabolism , Myosins/biosynthesis , Pericardium/embryology
14.
J Cell Sci ; 114(Pt 22): 4013-24, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11739633

ABSTRACT

Cells that express multiple connexins have the capacity to form heteromeric (mixed) gap junction hemichannels. We used a dominant negative connexin construct, consisting of bacterial beta-galactosidase fused to the C terminus of connexin43 (Cx43/beta-gal), to examine connexin compatibility in NIH 3T3 cells. Cx43/beta-gal is retained in a perinuclear compartment and inhibits Cx43 transport to the cell surface. The intracellular connexin pool induced by Cx43/beta-gal colocalized with a medial Golgi apparatus marker and was readily disassembled by treatment with brefeldin A. This was unexpected, since previous studies indicated that Cx43 assembly into hexameric hemichannels occurs in the trans-Golgi network (TGN) and is sensitive to brefeldin A. Further analysis by sucrose gradient fractionation showed that Cx43 and Cx43/beta-gal were assembled into a subhexameric complex. Cx43/beta-gal also specifically interacted with Cx46, but not Cx32, consistent with the ability of Cx43/beta-gal to simultaneously inhibit multiple connexins. We confirmed that interactions between Cx43/beta-gal and Cx46 reflect the ability of Cx43 and Cx46 to form heteromeric complexes, using HeLa and alveolar epithelial cells, which express both connexins. In contrast, ROS osteoblastic cells, which differentially sort Cx43 and Cx46, did not form Cx43/Cx46 heteromers. Thus, cells have the capacity to regulate whether or not compatible connexins intermix.


Subject(s)
Connexins/metabolism , Gap Junctions/metabolism , Recombinant Fusion Proteins/metabolism , trans-Golgi Network/metabolism , 3T3 Cells , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Brefeldin A/pharmacology , Cell Fractionation , Cells, Cultured , Connexins/genetics , Detergents/chemistry , Gap Junctions/chemistry , HeLa Cells , Humans , Immunohistochemistry , Mice , Microscopy, Electron , Octoxynol/chemistry , Protein Synthesis Inhibitors/pharmacology , Protein Transport/physiology , Pulmonary Alveoli/cytology , Rats , Recombinant Fusion Proteins/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
15.
J Cell Biol ; 154(1): 217-30, 2001 Jul 09.
Article in English | MEDLINE | ID: mdl-11449002

ABSTRACT

Connexin 43 (Cx43alpha1) gap junction has been shown to have an essential role in mediating functional coupling of neural crest cells and in modulating neural crest cell migration. Here, we showed that N-cadherin and wnt1 are required for efficient dye coupling but not for the expression of Cx43alpha1 gap junctions in neural crest cells. Cell motility was found to be altered in the N-cadherin-deficient neural crest cells, but the alterations were different from that elicited by Cx43alpha1 deficiency. In contrast, wnt1-deficient neural crest cells showed no discernible change in cell motility. These observations suggest that dye coupling may not be a good measure of gap junction communication relevant to motility. Alternatively, Cx43alpha1 may serve a novel function in motility. We observed that p120 catenin (p120ctn), an Armadillo protein known to modulate cell motility, is colocalized not only with N-cadherin but also with Cx43alpha1. Moreover, the subcellular distribution of p120ctn was altered with N-cadherin or Cx43alpha1 deficiency. Based on these findings, we propose a model in which Cx43alpha1 and N-cadherin may modulate neural crest cell motility by engaging in a dynamic cross-talk with the cell's locomotory apparatus through p120ctn signaling.


Subject(s)
Cadherins/metabolism , Connexin 43/metabolism , Gap Junctions , Neural Crest/cytology , Zebrafish Proteins , Animals , Bromodeoxyuridine/metabolism , Catenins , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/physiology , Cell Communication , Cell Division , Cell Movement , Cells, Cultured , Genotype , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Video , Phosphoproteins/metabolism , Phosphoproteins/physiology , Protein Binding , Proto-Oncogene Proteins/metabolism , Time Factors , Wnt Proteins , Wnt1 Protein , Delta Catenin
16.
Circ Res ; 88(11): 1196-202, 2001 Jun 08.
Article in English | MEDLINE | ID: mdl-11397787

ABSTRACT

Connexin43 (Cx43) is the principal connexin isoform in the mouse ventricle, where it is thought to provide electrical coupling between cells. Knocking out this gene results in anatomic malformations that nevertheless allow for survival through early neonatal life. We examined electrical wave propagation in the left (LV) and right (RV) ventricles of isolated Cx43 null mutated (Cx43(-/-)), heterozygous (Cx43(+/)(-)), and wild-type (WT) embryos using high-resolution mapping of voltage-sensitive dye fluorescence. Consistent with the compensating presence of the other connexins, no reduction in propagation velocity was seen in Cx43(-/-) ventricles at postcoital day (dpc) 12.5 compared with WT or Cx43(+/)(-) ventricles. A gross reduction in conduction velocity was seen in the RV at 15.5 dpc (in cm/second, mean [1 SE confidence interval], WT 9.9 [8.7 to 11.2], Cx43(+/)(-) 9.9 [9.0 to 10.9], and Cx43(-/-) 2.2 [1.8 to 2.7; P<0.005]) and in both ventricles at 17.5 dpc (in RV, WT 8.4 [7.6 to 9.3], Cx43(+/)(-) 8.7 [8.1 to 9.3], and Cx43(-/-) 1.1 [0.1 to 1.3; P<0.005]; in LV, WT 10.1 [9.4 to 10.7], Cx43(+/)(-) 8.3 [7.8 to 8.9], and Cx43(-/-) 1.7 [1.3 to 2.1; P<0.005]) corresponding with the downregulation of Cx40. Cx40 and Cx45 mRNAs were detectable in ventricular homogenates even at 17.5 dpc, probably accounting for the residual conduction function. Neonatal knockout hearts were arrhythmic in vivo as well as ex vivo. This study demonstrates the contribution of Cx43 to the electrical function of the developing mouse heart and the essential role of this gene in maintaining heart rhythm in postnatal life.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Connexin 43/deficiency , Heart Ventricles/physiopathology , Ventricular Dysfunction/physiopathology , Animals , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/embryology , Body Surface Potential Mapping , Cardiac Pacing, Artificial , Connexin 43/genetics , Connexin 43/metabolism , Connexins/genetics , Connexins/metabolism , Disease Models, Animal , Electrocardiography/methods , Electrophysiologic Techniques, Cardiac , Fluorescent Dyes , Heart Conduction System/physiopathology , Heart Rate , Heart Ventricles/chemistry , Heart Ventricles/embryology , Heterozygote , Homozygote , In Vitro Techniques , Mice , Mice, Inbred Strains , Mice, Knockout , Optics and Photonics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Ventricular Dysfunction/embryology , Ventricular Dysfunction/genetics , Video Recording , Gap Junction alpha-5 Protein
17.
Cell Commun Adhes ; 8(4-6): 249-52, 2001.
Article in English | MEDLINE | ID: mdl-12064597

ABSTRACT

A connexin construct consisting of bacterial beta-galactosidase fused to the C-terminus of connexin43 (Cx43/beta-gal) was used to examine Cx43 assembly in NIH 3T3 cells. Cx43/beta-gal is retained in a perinuclear compartment and inhibits Cx43 transport to the cell surface. The intracellular connexin pool trapped by Cx43/beta-gal was retained in a compartment that co-localized with a medial Golgi apparatus marker by immunofluorescence microscopy and that was readily disassembled by treatment with brefeldin A. Further analysis by sucrose gradient fractionation showed that Cx43 and Cx43/beta-gal were assembled into a sub-hexameric complex, and that Cx43/beta-gal expression also inhibited Cx43 assembly into hemichannels. While this is consistent with Cx43 hemichannel assembly in the trans Golgi network (TGN), these data also suggest that the dominant negative effect of Cx43/beta-gal on Cx43 trafficking may reflect a putative sub-hexameric assembly intermediate formed in the Golgi apparatus.


Subject(s)
Connexin 43/metabolism , Golgi Apparatus/metabolism , Protein Transport/physiology , beta-Galactosidase/metabolism , 3T3 Cells , Animals , Connexin 43/genetics , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , beta-Galactosidase/genetics , trans-Golgi Network/metabolism
18.
Cell Commun Adhes ; 8(4-6): 289-92, 2001.
Article in English | MEDLINE | ID: mdl-12064604

ABSTRACT

The Cx43alpha1 gap junctions play an important role in cardiovascular development. Studies using transgenic mouse models have indicated that this involves an essential role for Cx43alpha1 in modulating neural crest cell motility. We previously showed that a 6.8 kb mouse genomic sequence containing the promoter and upstream regulatory sequences of the Cx43alpha1 gene can drive lacZ reporter gene expression in all neural crest cell lineages in the mouse embryo. To obtain further insights into the sequence motifs and regulatory pathways involved in targeting Cx43alpha1 gene expression in neural crest cells, we assayed the activity of the mouse Cx43alpha1 promoter in evolutionarily distantly related zebrafish embryos. For these studies, the 6.8kb Cx43alpha1 genomic sequence and various deletion derivatives were used to generate GFP or lacZ expression vectors. The transcriptional activities of these constructs were analyzed in vivo after microinjection into one- or two- cell stage zebrafish embryos. These studies indicated that the mouse Cx43alpha1 promoter can drive lacZ expression in neural crest cells in the zebrafish embryos. Analysis by whole mount in situ hybridization showed that the endogenous zebrafish Cx43alpha1 gene is expressed maternally and zygotically, and expression is observed in regions where neural crest cells are found. To further elucidate the developmental regulation of Cx43alpha1 gene expression, we screened a zebrafish BAC library and identified a clone containing the entire zebrafish Cx43alpha1 gene and flanking upstream and downstream sequences. The upstrean Cx43alpha1 promoter sequences from zebrafish, mouse, and human were analyzed for evolutionarily conserved DNA motifs. Overall these studies suggest that the sequence motifs and transcriptional regulation involved in the targeting Cx43alpha1 expression to neural crest cells are evolutionarily conserved in zebrafish and mouse embryos.


Subject(s)
Connexin 43/genetics , Connexins/genetics , Gap Junctions/metabolism , Promoter Regions, Genetic , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Connexin 43/metabolism , Connexins/metabolism , Genes, Reporter , Green Fluorescent Proteins , Heart/embryology , Heart/growth & development , Indicators and Reagents/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Knockout , Neural Crest/cytology , Neural Crest/physiology , Sequence Analysis, DNA , Zebrafish Proteins/metabolism
19.
Cell Commun Adhes ; 8(4-6): 321-4, 2001.
Article in English | MEDLINE | ID: mdl-12064611

ABSTRACT

Our previous studies showed an essential role for connexin 43 or alpha1 connexin (Cx43alpha1) gap junctions in the modulation of neural crest cell motility. Cx43alpha1 gap junctions and N-cadherin containing adherens junctions are expressed in migrating cardiac neural crest cells. Analysis of the N-cadherin knockout (KO) mouse model revealed that N-cadherin is essential for gap junction mediated dye coupling but not for expression of Cx43alpha1 gap junctions in neural crest cells. Time lapse videomicroscopy and motion analysis showed that the motility of N-cadherin KO neural crest cells were altered, but the motility changes differed compared to Cx43alpha1 KO neural crest cells. These observations suggest that the role of N-cadherin in cell motility is not simply mediated via the modulation of Cx43alpha1 mediated cell-cell communication. This was confirmed by a parallel analysis of wnt-1 deficient neural crest cells, which also showed a reduction in dye coupling, and yet no change in cell motility. Analysis of p120 catenin (p120ctn), an Amardillo family protein known to play a role in cell motility, showed that it is colocalized with N-cadherin and Cx43alpha1 in migrating neural crest cells. This subcellular distribution was altered in the N-cadherin and Cx43alpha1 KO neural crest cells. Given these results, we propose that N-cadherin and Cx43alpha1 may modulate neural crest cell motility by engaging in a dynamic cross-talk with the cell's locomotory apparatus through p120ctn signaling.


Subject(s)
Cadherins/metabolism , Cell Movement/physiology , Connexin 43/metabolism , Neural Crest/physiology , Zebrafish Proteins , Animals , Catenins , Cell Adhesion Molecules/metabolism , Cell Communication/physiology , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Gap Junctions/metabolism , Heart/embryology , Mice , Microscopy, Video , Neural Crest/cytology , Phosphoproteins/metabolism , Proto-Oncogene Proteins/metabolism , Time Factors , Wnt Proteins , Wnt1 Protein , Delta Catenin
20.
Anat Rec ; 260(3): 279-93, 2000 11 01.
Article in English | MEDLINE | ID: mdl-11066038

ABSTRACT

The trisomy 16 (Ts16) mouse is generally considered a model for human Down's syndrome (trisomy 21). However, many of the cardiac defects in the Ts16 mouse do not reflect the heart malformations seen in patients suffering from this chromosomal disorder. In this study we describe the conotruncal malformations in mice with trisomy 16. The development of the outflow tract was immunohistochemically studied in serially sectioned hearts from 34 normal and 26 Ts16 mouse embryos ranging from 8.5 to 14.5 embryonic days. Conotruncal malformations observed in the Ts 16 embryos included double outlet right ventricle, persistent truncus arteriosus, Tetralogy of Fallot, and right-sided aortic arch. This spectrum of malformations is remarkably similar to that seen in humans suffering from DiGeorge syndrome (DGS). As perturbation of neural crest development has been proposed in the pathogenesis of DGS we specifically focussed on the fate of neural crest derived cells during outflow tract development of the Ts16 mouse using an antibody that enabled us to trace these cells during development. Severe perturbation of the neural crest-derived cell population was observed in each trisomic specimen. The abnormalities pertained to: 1) the size of the columns of neural crest-derived cells (or prongs); 2) the spatial orientation of these prongs within the mesenchymal tissues of the outflow tract; and 3) the location in which the neural crest cells interact with the myocardium. The latter abnormality appeared to be responsible for ectopic myocardialization found in trisomic embryos. Our observations strongly suggest that abnormal neural crest cell behavior is involved in the pathogenesis of the conotruncal malformations in the Ts16 mouse.


Subject(s)
Heart Defects, Congenital/embryology , Neural Crest/abnormalities , Trisomy , Animals , Connexin 43/analysis , DiGeorge Syndrome/embryology , DiGeorge Syndrome/etiology , DiGeorge Syndrome/pathology , Disease Models, Animal , Down Syndrome/etiology , Down Syndrome/pathology , Female , Fluorescent Antibody Technique, Indirect , Heart Defects, Congenital/etiology , Heart Defects, Congenital/pathology , Heart Ventricles/abnormalities , Karyotyping , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neural Crest/chemistry , Neural Crest/pathology , Pregnancy , Yolk Sac/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...