Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Biol Pharm Bull ; 43(11): 1715-1728, 2020.
Article in English | MEDLINE | ID: mdl-33132317

ABSTRACT

SV40-encoded microRNA (miRNA), miR-S1, downregulates the large and small T antigens (LTag and STag), which promote viral replication and cellular transformation, thereby presumably impairing LTag and STag functions essential for the viral life cycle. To explore the functional significance of miR-S1-mediated downregulation of LTag and STag as well as the functional roles of miR-S1, we evaluated viral DNA replication and proinflammatory cytokine induction in cells transfected with simian virus 40 (SV40) genome plasmid and its mutated form lacking miR-S1 expression. The SV40 genome encodes two mature miR-S1s, miR-S1-3p and miR-S1-5p, of which miR-S1-3p is the predominantly expressed form. MiR-S1-3p exerted strong repressive effects on a reporter containing full-length sequence complementarity, but only marginal effect on one harboring a sequence complementary to its seed sequence. Consistently, miR-S1-3p downregulated LTag and STag transcripts with complete sequence complementarity through miR-S1-3p-Ago2-mediated mRNA decay. Transfection of SV40 plasmid induced higher DNA replication and lower LTag and STag transcripts in most of the examined cells compared to that miR-S1-deficient SV40 plasmid. However, miR-S1 itself did not affect DNA replication without the downregulation of LTag transcripts. Both LTag and STag induced the expression of tumor necrosis factor α (TNFα) and interleukin (IL)-17F, which was slightly reduced by miR-S1 due to miR-S1-mediated downregulation of LTag and STag. Forced miR-S1 expression did not affect TNFα expression, but increased IL-17F expression. Overall, our findings suggest that miR-S1-3p is a latent modifier of LTag and STag functions, ensuring efficient viral replication and attenuating cytokine expression detrimental to the viral life cycle.


Subject(s)
Antigens, Viral, Tumor/genetics , Gene Expression Regulation, Viral/immunology , MicroRNAs/metabolism , RNA, Viral/metabolism , Simian virus 40/genetics , A549 Cells , DNA Replication/immunology , DNA, Viral/biosynthesis , HEK293 Cells , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Interleukin-17/metabolism , Interleukin-8/metabolism , Polyomavirus Infections/genetics , Polyomavirus Infections/immunology , Polyomavirus Infections/virology , Simian virus 40/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Virus Infections/genetics , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Virus Replication/immunology
2.
Genes Cells ; 21(1): 25-40, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26592235

ABSTRACT

We previously identified a distal regulatory element located approximately 5.5-kb upstream of the signal transducer and activator of transcription 1 (STAT1) gene, thereafter designating it as 5.5-kb upstream regulatory region (5.5URR). In this study, we investigated the functional roles of 5.5URR in the transcriptional regulation of STAT1 gene. A chromosome conformation capture assay indicated physical interaction of 5.5URR with the STAT1 core promoter. In luciferase reporter assays, 5.5URR-combined STAT1 core promoter exhibited significant increase in reporter activity enhanced by forced STAT1 expression or interferon (IFN) treatment, but STAT1 core promoter alone did not. The 5.5URR contained IFN-stimulated response element and GAS sites, which bound STAT1 complexes in electrophoretic mobility shift assays. Consistently, chromatin immunoprecipitation (ChIP) assays of HEK293 cells with Halo-tagged STAT1 expression indicated the association of Halo-tagged STAT1 with 5.5URR. ChIP assays with IFN treatment demonstrated that IFNs promoted the recruitment of Halo-tagged STAT1 to 5.5URR. Forced STAT1 expression or IFN treatment increased the expression of endogenous STAT1 and other IFN signaling pathway components, such as STAT2, IRF9 and IRF1, besides IFN-responsive genes. Collectively, the results suggest that 5.5URR may provide a regulatory platform for positive feedback control of STAT1 expression possibly to amplify or sustain the intracellular IFN signals.


Subject(s)
Feedback, Physiological , Gene Expression Regulation , Promoter Regions, Genetic , STAT1 Transcription Factor/genetics , 3T3 Cells , Animals , Base Sequence , Genes, Reporter , HEK293 Cells , Humans , Interferon Regulatory Factor-1/metabolism , Interferons/metabolism , Mice , Models, Biological , Molecular Sequence Data , Protein Binding/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction/genetics , Transcription, Genetic , Up-Regulation/genetics
3.
Exp Cell Res ; 336(2): 287-97, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26101156

ABSTRACT

Single-nucleotide polymorphisms associated with type 2 diabetes (T2D) have been identified in Jazf1, which is also involved in the oncogenesis of endometrial stromal tumors. To understand how Jazf1 variants confer a risk of tumorigenesis and T2D, we explored the functional roles of JAZF1 and searched for JAZF1 target genes in myogenic C2C12 cells. Consistent with an increase of Jazf1 transcripts during myoblast proliferation and their decrease during myogenic differentiation in regenerating skeletal muscle, JAZF1 overexpression promoted cell proliferation, whereas it retarded myogenic differentiation. Examination of myogenic genes revealed that JAZF1 overexpression transcriptionally repressed MEF2C and MRF4 and their downstream genes. AMP deaminase1 (AMPD1) was identified as a candidate for JAZF1 target by gene array analysis. However, promoter assays of Ampd1 demonstrated that mutation of the putative binding site for the TR4/JAZF1 complex did not alleviate the repressive effects of JAZF1 on promoter activity. Instead, JAZF1-mediated repression of Ampd1 occurred through the MEF2-binding site and E-box within the Ampd1 proximal regulatory elements. Consistently, MEF2C and MRF4 expression enhanced Ampd1 promoter activity. AMPD1 overexpression and JAZF1 downregulation impaired AMPK phosphorylation, while JAZF1 overexpression also reduced it. Collectively, these results suggest that aberrant JAZF1 expression contributes to the oncogenesis and T2D pathogenesis.


Subject(s)
AMP Deaminase/genetics , Carrier Proteins/genetics , Cell Transformation, Neoplastic/genetics , Diabetes Mellitus, Type 2/genetics , Muscle Development/genetics , Nuclear Proteins/genetics , AMP Deaminase/biosynthesis , Animals , Binding Sites/genetics , Carrier Proteins/biosynthesis , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Co-Repressor Proteins , DNA-Binding Proteins , Diabetes Mellitus, Type 2/pathology , Gene Expression Regulation/genetics , MEF2 Transcription Factors/biosynthesis , MEF2 Transcription Factors/genetics , Mice , Muscle Fibers, Skeletal/cytology , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Nuclear Proteins/biosynthesis , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering , Transcription, Genetic/genetics
4.
Sci Rep ; 3: 2183, 2013.
Article in English | MEDLINE | ID: mdl-23851606

ABSTRACT

The molecular mechanism of muscle degeneration in a lethal muscle disorder Duchene muscular dystrophy (DMD) has not been fully elucidated. The dystrophic dog, a model of DMD, shows a high mortality rate with a marked increase in serum creatine kinase (CK) levels in the neonatal period. By measuring serum CK levels in cord and venous blood, we found initial pulmonary respiration resulted in massive diaphragm damage in the neonates and thereby lead to the high serum CK levels. Furthermore, molecular biological techniques revealed that osteopontin was prominently upregulated in the dystrophic diaphragm prior to the respiration, and that immediate-early genes (c-fos and egr-1) and inflammation/immune response genes (IL-6, IL-8, COX-2, and selectin E) were distinctly overexpressed after the damage by the respiration. Hence, we segregated dystrophic phases at the molecular level before and after mechanical damage. These molecules could be biomarkers of muscle damage and potential targets in pharmaceutical therapies.


Subject(s)
Creatine Kinase/blood , Diaphragm/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Respiration , Animals , Animals, Newborn , Diaphragm/immunology , Diaphragm/metabolism , Disease Models, Animal , Dogs , Gene Expression Regulation , Hyalin/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Molecular Sequence Annotation , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/immunology , Osteopontin/genetics , Osteopontin/metabolism , Proteolysis , Respiration/genetics , Signal Transduction , Transcription, Genetic , Transcriptome
5.
Exp Cell Res ; 319(3): 77-88, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23142026

ABSTRACT

MicroRNA-1 (miR-1) has recently been suggested to function as a tumor suppressor. Its functional relevance was assessed by exploring structural and tumorigenic properties of lung cancer A549 cells stably transduced with retrovirus containing pre-miR-1. A549 cells overexpressing miR-1 exhibited a significant morphological change from a mesenchymal to an epithelial phenotype characterized by cell polarization and intercellular junctions. The cells showed increased expression of E-cadherin, which colocalized with cortical actin filaments and vinculin to form typical adherens junction at the apical regions of intercellular borders. Additionally, they exhibited occludin-positive tight junctions at similar apical regions. Moreover, their migratory and invasive activities were inhibited, and their sensitivity to doxorubicin was increased slightly compared to control mock-infected cells. These structural and tumorigenic properties induced by miR-1 were associated with the reduced expression of Slug, which was a transcriptional repressor of E-cadherin or an inducer of epithelial-to-mesenchymal transition. Consistently, Slug was identified as a miR-1 target by bioinformatics and a luciferase reporter assay with plasmids containing luciferase-Slug 3'UTR. Collectively, the data presented here suggest that re-expression of miR-1 may be an effective therapy that prevents cancer malignancy by converting cells from a mesenchymal phenotype to an epithelial phenotype via the downregulation of Slug.


Subject(s)
Adenocarcinoma/pathology , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/pathology , MicroRNAs/physiology , Transcription Factors/genetics , Adenocarcinoma/genetics , Base Sequence , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Humans , Lung Neoplasms/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness , RNA Interference , Snail Family Transcription Factors , Transcription Factors/antagonists & inhibitors , Transfection
6.
FEBS Lett ; 586(19): 3464-70, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22979984

ABSTRACT

Chromatin loops formed between distant regulatory elements and promoters modulate gene expression. We identified a novel distant regulatory element located approximately 120kb downstream of the gls promoter, and examined its regulatory relevance to gls gene expression in C2C12 cells by a chromosome conformation capture assay. The distant element physically interacted with the gls promoter in myoblasts but not in myotubes. Semiquantitative analysis by real-time PCR showed more abundant gls transcripts in myoblasts than in myotubes. These findings suggest that this distant element differentially regulates gls gene expression through dynamic formation and abrogation of a chromatin loop during myogenesis.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Glutaminase/genetics , Muscle Development/genetics , 3T3 Cells , Animals , Base Sequence , Cell Line , Conserved Sequence , Culture Media , DNA/genetics , Mice , Molecular Sequence Data , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Promoter Regions, Genetic , Regulatory Elements, Transcriptional , Sequence Homology, Nucleic Acid
7.
Mol Ther ; 17(1): 73-80, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18941441

ABSTRACT

Recombinant adeno-associated virus (rAAV)-mediated gene transfer is an attractive approach to the treatment of Duchenne muscular dystrophy (DMD). We investigated the muscle transduction profiles and immune responses associated with the administration of rAAV2 and rAAV8 in normal and canine X-linked muscular dystrophy in Japan (CXMD(J)) dogs. rAAV2 or rAAV8 encoding the lacZ gene was injected into the skeletal muscles of normal dogs. Two weeks after the injection, we detected a larger number of beta-galactosidase-positive fibers in rAAV8-transduced canine skeletal muscle than in rAAV2-transduced muscle. Although immunohistochemical analysis using anti-CD4 and anti-CD8 antibodies revealed less T-cell response to rAAV8 than to rAAV2, beta-galactosidase expression in rAAV8-injected muscle lasted for <4 weeks with intramuscular transduction. Canine bone marrow-derived dendritic cells (DCs) were activated by both rAAV2 and rAAV8, implying that innate immunity might be involved in both cases. Intravenous administration of rAAV8-lacZ into the hind limb in normal dogs and rAAV8-microdystrophin into the hind limb in CXMD(J) dogs resulted in improved transgene expression in the skeletal muscles lasting over a period of 8 weeks, but with a declining trend. The limb perfusion transduction protocol with adequate immune modulation would further enhance the rAAV8-mediated transduction strategy and lead to therapeutic benefits in DMD gene therapy.


Subject(s)
Dependovirus/genetics , Dependovirus/immunology , Muscle, Skeletal , Transduction, Genetic/methods , Animals , Blotting, Western , Dogs , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors/genetics , Genetic Vectors/immunology , Male , Muscular Dystrophy, Animal/immunology , Muscular Dystrophy, Animal/therapy , Reverse Transcriptase Polymerase Chain Reaction , beta-Galactosidase/genetics
8.
Cell Struct Funct ; 33(2): 163-9, 2008.
Article in English | MEDLINE | ID: mdl-18827405

ABSTRACT

miR-1, miR-133a, and miR-206 are muscle-specific microRNAs expressed in skeletal muscles and have been shown to contribute to muscle development. To gain insight into the pathophysiological roles of these three microRNAs in dystrophin-deficient muscular dystrophy, their expression in the tibialis anterior (TA) muscles of mdx mice and CXMD(J) dogs were evaluated by semiquantitative RT-PCR and in situ hybridization. Their temporal and spatial expression patterns were also analyzed in C2C12 cells during muscle differentiation and in cardiotoxin (CTX)-injured TA muscles to examine how muscle degeneration and regeneration affect their expression. In dystrophic TA muscles of mdx mice, miR-206 expression was significantly elevated as compared to that in control TA muscles of age-matched B10 mice, whereas there were no differences in miR-1 or miR-133a expression between B10 and mdx TA muscles. On in situ hybridization analysis, intense signals for miR-206 probes were localized in newly formed myotubes with centralized nuclei, or regenerating muscle fibers, but not in intact pre-degenerated fibers or numerous small mononucleated cells, possibly proliferating myoblasts and inflammatory infiltrates. Similar increased expression of miR-206 was also found in C2C12 differentiation and CTX-induced regeneration, in which differentiated myotubes or regenerating fibers showed abundant expression of miR-206. However, CXMD(J) TA muscles contained smaller amounts of miR-206, miR-1, and miR-133a than controls. They exhibited more severe and more progressive degenerative alterations than mdx TA muscles. Taken together, these observations indicated that newly formed myotubes showed markedly increased expression of miR-206, which might reflect active regeneration and efficient maturation of skeletal muscle fibers.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Animals , Cells, Cultured , Cobra Cardiotoxin Proteins/metabolism , Dogs , Dystrophin/deficiency , Dystrophin/genetics , Dystrophin/metabolism , In Situ Hybridization , Mice , Mice, Inbred mdx/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Regeneration/genetics , Regeneration/physiology , Reverse Transcriptase Polymerase Chain Reaction
9.
J Cell Sci ; 121(Pt 12): 2062-74, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18505798

ABSTRACT

In skeletal muscles, the sarcolemma is possibly stabilized and protected against contraction-imposed stress by intermediate filaments (IFs) tethered to costameric sarcolemma. Although there is emerging evidence that plectin links IFs to costameres through dystrophin-glycoprotein complexes (DGC), the molecular organization from plectin to costameres still remains unclear. Here, we show that plectin 1, a plectin isoform expressed in skeletal muscle, can interact with beta-synemin, actin and a DGC component, alpha-dystrobrevin, in vitro. Ultrastructurally, beta-synemin molecules appear to be incorporated into costameric dense plaques, where they seem to serve as actin-associated proteins rather than IF proteins. In fact, they can bind actin and alpha-dystrobrevin in vitro. Moreover, in vivo immunoprecipitation analyses demonstrated that beta-synemin- and plectin-immune complexes from lysates of muscle light microsomes contained alpha-dystrobrevin, dystrophin, nonmuscle actin, metavinculin, plectin and beta-synemin. These findings suggest a model in which plectin 1 interacts with DGC and integrin complexes directly, or indirectly through nonmuscle actin and beta-synemin within costameres. The DGC and integrin complexes would cooperate to stabilize and fortify the sarcolemma by linking the basement membrane to IFs through plectin 1, beta-synemin and actin. Besides, the two complexes, together with plectin and IFs, might have their own functions as platforms for distinct signal transduction.


Subject(s)
Actins/metabolism , Dystrophin-Associated Proteins/metabolism , Intermediate Filament Proteins/metabolism , Plectin/metabolism , Actins/genetics , Animals , Dystrophin/deficiency , Dystrophin/metabolism , Dystrophin-Associated Proteins/genetics , Intermediate Filament Proteins/genetics , Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Mice , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Plectin/chemistry , Plectin/genetics , Protein Binding , Rats , Sarcolemma/metabolism , Sarcolemma/ultrastructure
10.
Circulation ; 117(19): 2437-48, 2008 May 13.
Article in English | MEDLINE | ID: mdl-18458171

ABSTRACT

BACKGROUND: Respiratory support therapy significantly improves life span in patients with Duchenne muscular dystrophy; cardiac-related fatalities, including lethal arrhythmias, then become a crucial issue. It is therefore important to more thoroughly understand cardiac involvement, especially pathology of the conduction system, in the larger Duchenne muscular dystrophy animal models such as dystrophic dogs. METHODS AND RESULTS: When 10 dogs with canine X-linked muscular dystrophy in Japan (CXMD(J)) were examined at the age of 1 to 13 months, dystrophic changes of the ventricular myocardium were not evident; however, Purkinje fibers showed remarkable vacuolar degeneration as early as 4 months of age. The degeneration of CXMD(J) Purkinje fibers was coincident with overexpression of Dp71 at the sarcolemma and translocation of mu-calpain to the cell periphery near the sarcolemma or in the vacuoles. Immunoblotting of the microdissected fraction showed that mu-calpain-sensitive proteins such as desmin and cardiac troponin-I or -T were selectively degraded in the CXMD(J) Purkinje fibers. Utrophin was highly upregulated in the earlier stage of CXMD(J) Purkinje fibers, but the expression was dislocated when vacuolar degeneration was recognized at 4 months of age. Nevertheless, the expression of dystrophin-associated proteins alpha-, beta-, gamma-, and delta-sarcoglycans and beta-dystroglycan was well maintained at the sarcolemma of Purkinje fibers. CONCLUSIONS: Selective vacuolar degeneration of Purkinje fibers was found in the early stages of dystrophin deficiency. Dislocation of utrophin besides upregulation of Dp71 can be involved with this pathology. The degeneration of Purkinje fibers can be associated with the distinct deep Q waves in ECG and fatal arrhythmia seen in dystrophin deficiency.


Subject(s)
Dystrophin-Associated Proteins/analysis , Dystrophin/deficiency , Muscular Dystrophy, Animal/pathology , Purkinje Fibers/pathology , Utrophin/metabolism , Vacuoles/pathology , Animals , Arrhythmias, Cardiac , Dogs , Dystrophin/genetics , Electrocardiography , Purkinje Fibers/ultrastructure , Up-Regulation
11.
BMC Musculoskelet Disord ; 9: 1, 2008 Jan 09.
Article in English | MEDLINE | ID: mdl-18182116

ABSTRACT

BACKGROUND: Skeletal muscles are composed of heterogeneous collections of muscle fiber types, the arrangement of which contributes to a variety of functional capabilities in many muscle types. Furthermore, skeletal muscles can adapt individual myofibers under various circumstances, such as disease and exercise, by changing fiber types. This study was performed to examine the influence of dystrophin deficiency on fiber type composition of skeletal muscles in canine X-linked muscular dystrophy in Japan (CXMDJ), a large animal model for Duchenne muscular dystrophy. METHODS: We used tibialis cranialis (TC) muscles and diaphragms of normal dogs and those with CXMDJ at various ages from 1 month to 3 years old. For classification of fiber types, muscle sections were immunostained with antibodies against fast, slow, or developmental myosin heavy chain (MHC), and the number and size of these fibers were analyzed. In addition, MHC isoforms were detected by gel electrophoresis. RESULTS: In comparison with TC muscles of CXMDJ, the number of fibers expressing slow MHC increased markedly and the number of fibers expressing fast MHC decreased with growth in the affected diaphragm. In populations of muscle fibers expressing fast and/or slow MHC(s) but not developmental MHC of CXMDJ muscles, slow MHC fibers were predominant in number and showed selective enlargement. Especially, in CXMDJ diaphragms, the proportions of slow MHC fibers were significantly larger in populations of myofibers with non-expression of developmental MHC. Analyses of MHC isoforms also indicated a marked increase of type I and decrease of type IIA isoforms in the affected diaphragm at ages over 6 months. In addition, expression of developmental (embryonic and/or neonatal) MHC decreased in the CXMDJ diaphragm in adults, in contrast to continuous high-level expression in affected TC muscle. CONCLUSION: The CXMDJ diaphragm showed marked changes in fiber type composition unlike TC muscles, suggesting that the affected diaphragm may be effectively adapted toward dystrophic stress by switching to predominantly slow fibers. Furthermore, the MHC expression profile in the CXMDJ diaphragm was markedly different from that in mdx mice, indicating that the dystrophic dog is a more appropriate model than a murine one, to investigate the mechanisms of respiratory failure in DMD.


Subject(s)
Diaphragm/metabolism , Dystrophin/deficiency , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Myosin Heavy Chains/metabolism , Animals , Diaphragm/pathology , Disease Models, Animal , Dogs , Fluorescent Antibody Technique, Direct , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/pathology , Myosin Heavy Chains/classification
12.
BMC Musculoskelet Disord ; 8: 54, 2007 Jun 28.
Article in English | MEDLINE | ID: mdl-17598883

ABSTRACT

BACKGROUND: Matrix metalloproteinases (MMPs) are key regulatory molecules in the formation, remodeling and degradation of all extracellular matrix (ECM) components in both physiological and pathological processes in various tissues. The aim of this study was to examine the involvement of gelatinase MMP family members, MMP-2 and MMP-9, in dystrophin-deficient skeletal muscle. Towards this aim, we made use of the canine X-linked muscular dystrophy in Japan (CXMDJ) model, a suitable animal model for Duchenne muscular dystrophy. METHODS: We used surgically biopsied tibialis cranialis muscles of normal male dogs (n = 3) and CXMDJ dogs (n = 3) at 4, 5 and 6 months of age. Muscle sections were analyzed by conventional morphological methods and in situ zymography to identify the localization of MMP-2 and MMP-9. MMP-2 and MMP-9 activity was examined by gelatin zymography and the levels of the respective mRNAs in addition to those of regulatory molecules, including MT1-MMP, TIMP-1, TIMP-2, and RECK, were analyzed by semi-quantitative RT-PCR. RESULTS: In CXMDJ skeletal muscle, multiple foci of both degenerating and regenerating muscle fibers were associated with gelatinolytic MMP activity derived from MMP-2 and/or MMP-9. In CXMDJ muscle, MMP-9 immunoreactivity localized to degenerated fibers with inflammatory cells. Weak and disconnected immunoreactivity of basal lamina components was seen in MMP-9-immunoreactive necrotic fibers of CXMDJ muscle. Gelatinolytic MMP activity observed in the endomysium of groups of regenerating fibers in CXMDJ did not co-localize with MMP-9 immunoreactivity, suggesting that it was due to the presence of MMP-2. We observed increased activities of pro MMP-2, MMP-2 and pro MMP-9, and levels of the mRNAs encoding MMP-2, MMP-9 and the regulatory molecules, MT1-MMP, TIMP-1, TIMP-2, and RECK in the skeletal muscle of CXMDJ dogs compared to the levels observed in normal controls. CONCLUSION: MMP-2 and MMP-9 are likely involved in the pathology of dystrophin-deficient skeletal muscle. MMP-9 may be involved predominantly in the inflammatory process during muscle degeneration. In contrast, MMP-2, which was activated in the endomysium of groups of regenerating fibers, may be associated with ECM remodeling during muscle regeneration and fiber growth.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Muscle, Skeletal/enzymology , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Animals , Disease Models, Animal , Dogs , Dystrophin/genetics , Enzyme Activation , Gene Expression Regulation, Enzymologic , Immunohistochemistry , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Myositis/genetics , Myositis/metabolism , Myositis/pathology , Regeneration , Reverse Transcriptase Polymerase Chain Reaction
13.
BMC Cardiovasc Disord ; 6: 47, 2006 Dec 04.
Article in English | MEDLINE | ID: mdl-17140458

ABSTRACT

BACKGROUND: Cardiac mortality in Duchenne muscular dystrophy (DMD) has recently become important, because risk of respiratory failure has been reduced due to widespread use of the respirator. The cardiac involvement is characterized by distinctive electrocardiographic abnormalities or dilated cardiomyopathy, but the pathogenesis has remained obscure. In research on DMD, Golden retriever-based muscular dystrophy (GRMD) has attracted much attention as an animal model because it resembles DMD, but GRMD is very difficult to maintain because of their severe phenotypes. We therefore established a line of dogs with Beagle-based canine X-linked muscular dystrophy in Japan (CXMDJ) and examined the cardiac involvement. METHODS: The cardiac phenotypes of eight CXMDJ and four normal male dogs 2 to 21 months of age were evaluated using electrocardiography, echocardiography, and histopathological examinations. RESULTS: Increases in the heart rate and decreases in PQ interval compared to a normal littermate were detected in two littermate CXMDJ dogs at 15 months of age or older. Distinct deep Q-waves and increase in Q/R ratios in leads II, III, and aVF were detected by 6-7 months of age in all CXMDJ dogs. In the echocardiogram, one of eight of CXMDJ dogs showed a hyperechoic lesion in the left ventricular posterior wall at 5 months of age, but the rest had not by 6-7 months of age. The left ventricular function in the echocardiogram indicated no abnormality in all CXMDJ dogs by 6-7 months of age. Histopathology revealed myocardial fibrosis, especially in the left ventricular posterobasal wall, in three of eight CXMDJ dogs by 21 months of age. CONCLUSION: Cardiac involvement in CXMDJ dogs is milder and has slower progression than that described in GRMD dogs. The distinct deep Q-waves have been ascribed to myocardial fibrosis in the posterobasal region of the left ventricle, but our data showed that they precede the lesion on echocardiogram and histopathology. These findings imply that studies of CXMDJ may reveal not only another causative mechanism of the deep Q-waves but also more information on the pathogenesis in the dystrophin-deficient heart.


Subject(s)
Disease Models, Animal , Dogs , Genetic Linkage , Heart Diseases/etiology , Muscular Dystrophy, Animal/complications , Muscular Dystrophy, Animal/genetics , X Chromosome , Animals , Disease Progression , Echocardiography , Electrocardiography , Fibrosis , Heart Diseases/diagnosis , Heart Diseases/physiopathology , Heart Rate , Heart Ventricles , Male , Myocardium/pathology , Ventricular Function, Left
14.
Acta Myol ; 24(2): 145-54, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16550932

ABSTRACT

Canine X-linked muscular dystrophy (CXMD), which was found in a colony of golden retriever, is caused by a mutation in the dystrophin gene and it is a useful model of Duchenne muscular dystrophy (DMD). To investigate the pathogenesis and to develop therapy of DMD, we have established a beagle-based CXMD colony in Japan (CXMDJ) and examined their phenotypes. The mortality by 3 days of age in the third generation (G3) of CXMDJ dogs, 32.3%, was considerably higher than that in normal G3 littermates, 13.3%. Serum creatine kinase (CK) levels of G3 CXMDJ were significantly higher than that of normal male dogs with two peaks: at shortly after birth and around 2 months of age. Diaphragm muscle involvement occurred shortly after birth and was more severe than that of limb muscles. Stress during whelping might be associated with the neonatal death and respiratory muscle involvement. Gait disturbance was also noticed after 2 months of age. The involvement of limb and temporal muscles was observed from 2 months of age, which corresponded with the second peak of serum CK. Macroglossia, dysphagia, drooling and jaw joint contracture were overt from 4 months of age. We noticed severe macroglossia and hypertrophy of the sublingual muscles at the age of 12 months, and these were important features of this model, because dysphagia is one of major symptoms in older DMD patients. Overall, the phenotypes of CXMDJ were roughly identical to those of CXMD dogs in the literature. Beagle-based CXMDJ is smaller and easier to handle than golden retriever, therefore they are a useful model for DMD.


Subject(s)
Muscular Dystrophy, Animal/diagnosis , Animals , Body Weight , Creatine Kinase/blood , Disease Models, Animal , Dog Diseases , Dogs , Facial Muscles/pathology , Female , Gait , Japan , Macroglossia/pathology , Male , Mouth Floor/pathology , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/blood , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/therapy , Phenotype
15.
Mol Ther ; 10(5): 821-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509500

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal disorder of skeletal muscle caused by mutations in the dystrophin gene. Adeno-associated virus (AAV) vector-mediated gene therapy is a promising approach to the disease. Although a rod-truncated microdystrophin gene has been proven to ameliorate dystrophic phenotypes, the level of microdystrophin expression required for effective gene therapy by an AAV vector has not been determined yet. Here, we constructed a recombinant AAV type 2 vector, AAV2-MCKDeltaCS1, expressing microdystrophin (DeltaCS1) under the control of a muscle-specific MCK promoter and injected it into TA muscles of 10-day-old and 5-week-old mdx mice. AAV2-MCKDeltaCS1-mediated gene transfer into 5-week-old mdx muscle resulted in extensive and long-term expression of microdystrophin and significantly improved force generation. Interestingly, 10-day-old injected muscle expressed microdystrophin in a limited number of myofibers but showed hypertrophy of microdystrophin-positive muscle fibers and considerable recovery of contractile force. Thus, we concluded that AAV2-MCKDeltaCS1 could be a powerful tool for gene therapy of DMD.


Subject(s)
Dependovirus/genetics , Dystrophin/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/therapy , Animals , Creatine Kinase/genetics , Dystrophin/metabolism , Gene Expression , Mice , Mice, Inbred mdx , Muscle Contraction/genetics , Muscle Contraction/physiology , Muscle Fibers, Skeletal/chemistry , Muscle, Skeletal/chemistry , Muscle, Skeletal/cytology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Phenotype , Promoter Regions, Genetic/genetics , Sarcolemma/chemistry , Sarcolemma/metabolism
16.
J Alzheimers Dis ; 6(5): 483-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15505369

ABSTRACT

A new oral vaccine for Alzheimer's disease was developed using recombinant adeno-associated virus vector carrying Abeta cDNA (AAV/Abeta). Oral administration of the vaccine without adjuvant induced the expression and secretion of Abeta1-43 or Abeta1-21 in the epithelial cell layer of the intestine in amyloid precursor protein transgenic mice. Serum antibody levels were elevated for more than six months, while T cell proliferative responses to Abeta was not detected. Brain Abeta burden was significantly decreased compared to the control without inflammatory changes. This oral AAV/Abeta vaccine seems to be promising for prevention and treatment of Alzheimer's disease.


Subject(s)
Alzheimer Disease/prevention & control , Alzheimer Vaccines/administration & dosage , Dependovirus/genetics , Genetic Vectors/genetics , Administration, Oral , Alzheimer Disease/immunology , Alzheimer Vaccines/immunology , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Antigens, CD/immunology , Blotting, Western , DNA Primers/genetics , DNA, Complementary/metabolism , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Mice, Transgenic
17.
Exp Anim ; 52(2): 93-7, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12806883

ABSTRACT

The purpose of this study was to develop a strain of canine X-linked muscular dystrophy (CXMD), a model of Duchenne muscular dystrophy, in Japan. A female beagle was artificially inseminated with frozen-thawed spermatozoa derived from an affected golden retriever. Subsequently, two carrier female dogs (G1 carriers) and four normal male littermates were produced. Thereafter, the two G1 carriers were mated with beagle sires. As a result, each bitch whelped three times, and out of 54 pups, 17 affected male descendants, and 11 carrier female descendants (G2 carriers) were detected. One G2 carrier was then mated with a beagle sire and 15 pups in two whelpings were produced, including five affected males and four carrier females (G3 carriers). A total of 10 female beagles were artificially inseminated to evaluate the fertility of the frozen-thawed spermatozoa from the two affected dogs. The whelping rates of the two affected dogs were 4/5 and the litter sizes were 5.0 +/- 1.41 and 6.0 +/- 0.82, respectively. These results indicate that a canine X-linked muscular dystrophy colony has been established in Japan. We called them CXMDJ.


Subject(s)
Genetic Linkage , Muscular Dystrophies/genetics , X Chromosome , Animals , Dogs , Female , Genotype , Japan , Male , Pedigree
18.
J Cell Biol ; 158(6): 1097-107, 2002 Sep 16.
Article in English | MEDLINE | ID: mdl-12221071

ABSTRACT

Alpha1-syntrophin is a member of the family of dystrophin-associated proteins; it has been shown to recruit neuronal nitric oxide synthase and the water channel aquaporin-4 to the sarcolemma by its PSD-95/SAP-90, Discs-large, ZO-1 homologous domain. To examine the role of alpha1-syntrophin in muscle regeneration, we injected cardiotoxin into the tibialis anterior muscles of alpha1-syntrophin-null (alpha1syn-/-) mice. After the treatment, alpha1syn-/- muscles displayed remarkable hypertrophy and extensive fiber splitting compared with wild-type regenerating muscles, although the untreated muscles of the mutant mice showed no gross histological change. In the hypertrophied muscles of the mutant mice, the level of insulin-like growth factor-1 transcripts was highly elevated. Interestingly, in an early stage of the regeneration process, alpha1syn-/- mice showed remarkably deranged neuromuscular junctions (NMJs), accompanied by impaired ability to exercise. The contractile forces were reduced in alpha1syn-/- regenerating muscles. Our results suggest that the lack of alpha1-syntrophin might be responsible in part for the muscle hypertrophy, abnormal synapse formation at NMJs, and reduced force generation during regeneration of dystrophin-deficient muscle, all of which are typically observed in the early stages of Duchenne muscular dystrophy patients.


Subject(s)
Membrane Proteins/physiology , Muscle Proteins/physiology , Muscle, Skeletal/physiology , Neuromuscular Junction/anatomy & histology , Regeneration , Animals , Aquaporin 4 , Aquaporins/analysis , Calcium-Binding Proteins , Cobra Cardiotoxin Proteins/administration & dosage , Cobra Cardiotoxin Proteins/toxicity , Hypertrophy , Injections, Intramuscular , Membrane Proteins/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , Muscle Contraction/drug effects , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Fast-Twitch/metabolism , Muscle Proteins/genetics , Muscle, Skeletal/anatomy & histology , Myosin Heavy Chains/metabolism , Physical Exertion/drug effects , Protein Isoforms/metabolism , Somatomedins/analysis , Time Factors
19.
Hum Mol Genet ; 11(15): 1719-30, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12095914

ABSTRACT

The ability to transfer the dystrophin gene stably to the skeletal muscle of DMD patients is a major confounding issue in establishing an effective gene therapy for this disease. To overcome this problem, we have examined the ability of muscle fibres from mdx mice to act as in situ factories of retroviral vector production. Tibialis anterior (TA) muscles from 4-week-old mdx mice were injected with an adenoviral vector expressing LacZ within a retroviral expression cassette (AdLZIN). Retroviral vector production was induced by the inclusion of two additional adenoviral vectors expressing retroviral gag-pol (AdGagPol) and 10A1 env genes (Ad10A1). Upon introduction of infected muscles into cell culture, colonies of beta-galactosidase-expressing myotubes formed only in cultures where the muscle was injected with AdLZIN, AdGagPol and Ad10A1, but not from muscle injected with AdLZIN only. Muscles from mdx/nude mice producing retroviral vector displayed a 4.6-fold increase in beta-galactosidase-positive myofibres after 1 month, compared with contralateral muscle in the same animal injected with AdLZIN and AdGagPol only. By constructing a hybrid adeno-retroviral vector expressing a truncated micro-dystrophin construct (AdmicroDyIN), we were able to partially correct the mdx dystrophic phenotype. AdmicroDyIN-mediated expression of micro-dystrophin in mdx TA muscle restored the formation of the dystrophin-associated glycoprotein complex and significantly reduced the level of muscle degeneration over uninjected controls. By stimulating in situ production of retroviral vector expressing micro-dystrophin, we achieved 92%+/-6% transduction of myofibres in the TA muscle by 4 weeks. Strikingly, by 3 months post injection, micro-dystrophin was still expressed to high levels in nearly all the myofibres of the TA muscle. By comparison, there was a pronounced drop in the levels of micro-dystrophin expressed by muscles injected with AdmicroDyIN only. Finally, using a novel PCR approach, we detected reverse-transcribed, integrated proviral sequences in TA muscle genomic DNA by 4 weeks post injection, the levels of which were found to increase after 3 months.


Subject(s)
Adenoviridae/genetics , Dystrophin/genetics , Gene Transfer Techniques , Genetic Vectors , Muscular Dystrophy, Duchenne/therapy , Retroviridae/genetics , Animals , Disease Models, Animal , Genetic Therapy , Lac Operon , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Transduction, Genetic
20.
Biochem Biophys Res Commun ; 293(4): 1265-72, 2002 May 17.
Article in English | MEDLINE | ID: mdl-12054513

ABSTRACT

The adeno-associated virus vector is a good tool for gene transfer into skeletal muscle, but the length of a gene that can be incorporated is limited. To develop a gene therapy for Duchenne muscular dystrophy, we generated a series of rod-truncated micro-dystrophin cDNAs: M3 (one rod repeat, 3.9 kb), AX11 (three rod repeats, 4.4 kb), and CS1 (four rod repeats, 4.9 kb). These micro-dystrophins, driven by a CAG promoter, were used to produce transgenic (Tg) mdx mice and all three micro-dystrophins were shown to localize at the sarcolemma together with the expression of dystrophin-associated proteins. Among them, CS1 greatly improved dystrophic phenotypes of mdx mice and contractile force of the diaphragm in particular was restored to the level of normal C57BL/10 mice. AX11 modestly ameliorated the dystrophic pathology, but, importantly, M3-Tg mdx mice still showed severe dystrophic phenotypes. These data suggest that the rod structure, and its length in particular, is crucial for the function of micro-dystrophin.


Subject(s)
Dystrophin/biosynthesis , Dystrophin/genetics , Mice, Inbred mdx , Transgenes , Animals , Blotting, Western , Creatine Kinase/blood , DNA, Complementary/metabolism , Diaphragm/metabolism , Dystrophin/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/therapy , Phenotype , Promoter Regions, Genetic , Sarcolemma/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...