Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 93
Filtrer
1.
Gene Ther ; 20(5): 497-503, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-22855092

RÉSUMÉ

Gene delivery vectors derived from adeno-associated virus (AAV) have great potential as therapeutic agents. rAAV1 and rAAV6, efficiently target striated muscle, but the mechanisms that determine their tropism remain unclear. It is known that AAV6, but not AAV1, interacts with heparin-sulfate proteoglycans (HSPG). HSPGs are not primary receptors for AAV6, but heparin interactions may affect tissue tropism and transduction. To investigate these possibilities, we generated rAAV1 and rAAV6 capsids that do or do not bind heparin. We evaluated the transduction profile of these vectors in vivo across multiple routes of administration, and found that heparin-binding capability influences tissue transduction in striated muscle and neuronal tissues. Heparin-binding capsids transduce striated muscle more efficiently than non-binding capsids, via both intramuscular and intravenous injection. However, rAAV6 achieved greater muscle transduction than the heparin-binding rAAV1 variant, suggesting that there are additional factors that influence differences in transduction efficiency between AAV1 and AAV6. Interestingly, the opposite trend was found when vectors were delivered via intracranial injection. Non-binding vectors achieved robust and widespread gene expression, whereas transduction via heparin-binding serotypes was substantially reduced. These data indicate that heparin-binding capability is an important determinant of transduction that should be considered in the design of rAAV-mediated gene therapies.


Sujet(s)
Dependovirus/génétique , Techniques de transfert de gènes , Héparine/métabolisme , Muscle strié/métabolisme , Animaux , Capside/métabolisme , Dependovirus/métabolisme , Thérapie génétique , Protéoglycanes à sulfate d'héparane , Héparine/génétique , Humains , Souris , Transduction génétique , Tropisme/génétique
2.
Biophys J ; 98(4): 606-16, 2010 Feb 17.
Article de Anglais | MEDLINE | ID: mdl-20159157

RÉSUMÉ

Progressive force loss in Duchenne muscular dystrophy is characterized by degeneration/regeneration cycles and fibrosis. Disease progression may involve structural remodeling of muscle tissue. An effect on molecular motorprotein function may also be possible. We used second harmonic generation imaging to reveal vastly altered subcellular sarcomere microarchitecture in intact single dystrophic mdx muscle cells (approximately 1 year old). Myofibril tilting, twisting, and local axis deviations explain at least up to 20% of force drop during unsynchronized contractile activation as judged from cosine angle sums of myofibril orientations within mdx fibers. In contrast, in vitro motility assays showed unaltered sliding velocities of single mdx fiber myosin extracts. Closer quantification of the microarchitecture revealed that dystrophic fibers had significantly more Y-shaped sarcomere irregularities ("verniers") than wild-type fibers (approximately 130/1000 microm(3) vs. approximately 36/1000 microm(3)). In transgenic mini-dystrophin-expressing fibers, ultrastructure was restored (approximately 38/1000 microm(3) counts). We suggest that in aged dystrophic toe muscle, progressive force loss is reflected by a vastly deranged micromorphology that prevents a coordinated and aligned contraction. Second harmonic generation imaging may soon be available in routine clinical diagnostics, and in this work we provide valuable imaging tools to track and quantify ultrastructural worsening in Duchenne muscular dystrophy, and to judge the beneficial effects of possible drug or gene therapies.


Sujet(s)
Moteurs moléculaires/composition chimique , Moteurs moléculaires/métabolisme , Muscles squelettiques/métabolisme , Dystrophies musculaires/métabolisme , Vieillissement , Algorithmes , Animaux , Phénomènes biomécaniques , Dystrophine/métabolisme , Régulation de l'expression des gènes , Humains , Imagerie tridimensionnelle , Souris , Souris de lignée mdx , Souris transgéniques , Microscopie , Imagerie moléculaire , Mouvement , Fibres musculaires squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Muscles squelettiques/physiologie , Dystrophies musculaires/physiopathologie , Sarcomères/métabolisme
3.
Biophys J ; 94(12): 4751-65, 2008 Jun.
Article de Anglais | MEDLINE | ID: mdl-18424498

RÉSUMÉ

Skeletal muscle unloaded shortening has been indirectly determined in the past. Here, we present a novel high-speed optical tracking technique that allows recording of unloaded shortening in single intact, voltage-clamped mammalian skeletal muscle fibers with 2-ms time resolution. L-type Ca(2+) currents were simultaneously recorded. The time course of shortening was biexponential: a fast initial phase, tau(1), and a slower successive phase, tau(2,) with activation energies of 59 kJ/mol and 47 kJ/mol. Maximum unloaded shortening speed, v(u,max), was faster than that derived using other techniques, e.g., approximately 14.0 L(0) s(-1) at 30 degrees C. Our technique also allowed direct determination of shortening acceleration. We applied our technique to single fibers from C57 wild-type, dystrophic mdx, and minidystrophin-expressing mice to test whether unloaded shortening was affected in the pathophysiological mechanism of Duchenne muscular dystrophy. v(u,max) and a(u,max) values were not significantly different in the three strains, whereas tau(1) and tau(2) were increased in mdx fibers. The results were complemented by myosin heavy and light chain (MLC) determinations that showed the same myosin heavy chain IIA profiles in the interossei muscles from the different strains. In mdx muscle, MLC-1f was significantly increased and MLC-2f and MLC-3f somewhat reduced. Fast initial active shortening seems almost unaffected in mdx muscle.


Sujet(s)
Dystrophine/métabolisme , Vidéomicroscopie/méthodes , Fibres musculaires squelettiques/cytologie , Fibres musculaires squelettiques/physiologie , Muscles squelettiques/cytologie , Muscles squelettiques/physiologie , Animaux , Cellules cultivées , Dystrophine/génétique , Mâle , Souris , Souris de lignée BALB C , Souris de lignée mdx , Souris transgéniques , Vidéomicroscopie/instrumentation , Techniques de patch-clamp , Traitement du signal assisté par ordinateur/instrumentation
4.
Gene Ther ; 12(14): 1099-108, 2005 Jul.
Article de Anglais | MEDLINE | ID: mdl-15759015

RÉSUMÉ

Gene therapy for Duchenne muscular dystrophy (DMD) will require sustained expression of therapeutic dystrophins in striated muscles. Lentiviral vectors have a relatively large transgene carrying capacity and can integrate into nondividing cells. We therefore explored the use of lentiviral vectors for transferring genes into mouse skeletal muscle cells. These vectors successfully transferred a minidystrophin expression cassette into mdx muscles, and minidystrophin expression persisted and prevented subsequent muscle fiber degeneration for at least 6 months. However, only low to moderate levels of skeletal muscle transduction could be obtained by intramuscular injection of the highest currently available lentiviral doses. Using cultured cells, the lentiviral vectors effectively transduced proliferating and terminally differentiated muscle cells, indicating that cell cycling is not essential for transduction of myogenic cells. We further showed that lentiviral vectors efficiently transduced both primary myoblasts and multipotent adult progenitor cells (MAPCs) in vitro, and the cells persistently expressed transgenes without any obvious toxicity. When mdx primary myoblasts were genetically modified with minidystrophin vectors and transplanted into mdx skeletal muscles, significant numbers of dystrophin-expressing myofibers formed. Finally, we showed that a short, highly active CK6 regulatory cassette directed muscle-specific activity in the context of the lentiviral vectors. The ability of lentiviral vectors to transduce myogenic progenitors using a minidystrophin cassette regulated by a muscle-specific promoter suggests that this system could be useful for ex vivo gene therapy of muscular dystrophy.


Sujet(s)
Dystrophine/métabolisme , Thérapie génétique/méthodes , Vecteurs génétiques , Myopathie de Duchenne/thérapie , Animaux , Différenciation cellulaire , Prolifération cellulaire , Cellules cultivées , Dystrophine/génétique , Expression des gènes , Lentivirus/génétique , Souris , Souris de lignée C57BL , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/anatomopathologie , Myoblastes/métabolisme , Myoblastes/anatomopathologie , Cellules souches/métabolisme , Cellules souches/anatomopathologie , Transduction génétique , Transgènes
5.
Acta Myol ; 24(3): 184-93, 2005 Dec.
Article de Anglais | MEDLINE | ID: mdl-16629052

RÉSUMÉ

Over the past decade, adeno-associated virus (AAV) has become an extremely promising vector for gene therapy of many genetic disorders. This review summarizes the specific challenges that must be overcome to apply AAV gene therapy to Duchenne muscular dystrophy. Many of these challenges have been met successfully in animal studies, but further work is needed to translate these results into an effective clinical treatment.


Sujet(s)
Dependovirus , Thérapie génétique , Vecteurs génétiques , Myopathie de Duchenne/thérapie , Dystrophine/génétique , Humains , Myopathie de Duchenne/génétique , Myopathie de Duchenne/immunologie , Transgènes
6.
J Physiol ; 555(Pt 1): 251-65, 2004 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-14594987

RÉSUMÉ

L-type calcium currents (iCa) were recorded using the two-microelectrode voltage-clamp technique in single short toe muscle fibres of three different mouse strains: (i) C57/SV129 wild-type mice (wt); (ii) mdx mice (an animal model for Duchenne muscular dystrophy; and (iii) transgenically engineered mini-dystrophin (MinD)-expressing mdx mice. The activation and inactivation properties of iCa were examined in 2- to 18-month-old animals. Ca2+ current densities at 0 mV in mdx fibres increased with age, but were always significantly smaller compared to age-matched wild-type fibres. Time-to-peak (TTP) of iCa was prolonged in mdx fibres compared to wt fibres. MinD fibres always showed similar TTP and current amplitudes compared to age-matched wt fibres. In all three genotypes, the voltage-dependent inactivation and deactivation of iCa were similar. Intracellular resting calcium concentration ([Ca2+]i) and the distribution of dihydropyridine binding sites were also not different in young animals of all three genotypes, whereas iCa was markedly reduced in mdx fibres. We conclude, that dystrophin influences L-type Ca2+ channels via a direct or indirect linkage which may be disrupted in mdx mice and may be crucial for proper excitation-contraction coupling initiating Ca2+ release from the sarcoplasmic reticulum. This linkage seems to be fully restored in the presence of mini-dystrophin.


Sujet(s)
Canaux calciques de type L/biosynthèse , Dystrophine/biosynthèse , Muscles squelettiques/métabolisme , Dystrophies musculaires/métabolisme , Animaux , Calcium/métabolisme , Calcium/pharmacologie , Canaux calciques de type L/génétique , Relation dose-effet des médicaments , Dystrophine/génétique , Potentiels de membrane/effets des médicaments et des substances chimiques , Potentiels de membrane/physiologie , Souris , Souris de lignée C57BL , Souris de lignée mdx , Souris transgéniques , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Dystrophies musculaires/génétique , Liaison aux protéines/effets des médicaments et des substances chimiques , Liaison aux protéines/physiologie
7.
Hum Mol Genet ; 13(2): 213-21, 2004 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-14645204

RÉSUMÉ

Duchenne muscular dystrophy is an X-linked recessive disorder, primarily characterized by progressive muscle weakness and wasting. The disease results from the absence of dystrophin, however the precise molecular mechanisms leading to muscle pathology are poorly understood. Dystrophic muscles undergo increased oxidative stress and altered calcium homeostasis, which may contribute to myofiber loss by triggering both necrosis and apoptosis. Recent studies have identified ARC (apoptosis repressor with caspase recruitment domain) as an abundant protein in human muscle that can inhibit both hypoxia and caspase-8-induced apoptosis as well as protect cells from oxidative stress. To explore a potential role for ARC in protecting muscle fibers from dystrophic breakdown, we have cloned and characterized murine ARC and studied its expression in normal and dystrophic mouse muscle. ARC is expressed at high levels in striated muscle and displays fiber-type restricted expression patterns. ARC expression levels are normal in dystrophic mdx mice, although the intracellular localization pattern of ARC is slightly altered compared with normal muscles. Overexpression of ARC in transgenic mdx mice failed to alleviate the dystrophic pathology in skeletal muscles, suggesting that misregulation of the molecular pathways regulated by ARC does not significantly contribute to myofiber death.


Sujet(s)
Protéines de transport/métabolisme , Dystrophine/déficit , Protéines du muscle/métabolisme , Muscles squelettiques/métabolisme , Animaux , Protéines régulatrices de l'apoptose , Protéines de transport/génétique , Caspase-3 , Caspases/métabolisme , Perméabilité des membranes cellulaires , Cartographie chromosomique , Clonage moléculaire , Régulation de l'expression des gènes , Humains , Mâle , Souris , Souris de lignée mdx , Souris transgéniques , Mitochondries/métabolisme , Protéines du muscle/génétique , Muscles squelettiques/anatomopathologie , Myopathie de Duchenne/génétique , Myopathie de Duchenne/anatomopathologie , Spécificité d'organe , Valeurs de référence
8.
Gene Ther ; 9(23): 1576-88, 2002 Dec.
Article de Anglais | MEDLINE | ID: mdl-12424610

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is an X-linked, lethal muscular disorder caused by a defect in the DMD gene. AAV vector-mediated micro-dystrophin cDNA transfer is an attractive approach to treatment of DMD. To establish effective gene transfer into skeletal muscle, we examined the transduction efficiency of an AAV vector in skeletal muscles of dystrophin-deficient mdx mice. When an AAV vector encoding the LacZ gene driven by a CMV promoter (AAV-CMVLacZ) was introduced, beta-galactosidase expression markedly decreased in mdx muscle 4 weeks after injection due to immune responses against the transgene product. We also injected AAV-CMVLacZ into skeletal muscles of mini-dystrophin-transgenic mdx mice (CVBA3'), which show ameliorated phenotypes without overt signs of muscle degeneration. AAV vector administration, however, evoked substantial immune responses in CVBA3' muscle. Importantly, AAV vector using muscle-specific MCK promoter also elicited responses in mdx muscle, but at a considerably later period. These results suggested that neo-antigens introduced by AAV vectors could evoke immune reactions in mdx muscle, since increased permeability allowed a leakage of neo-antigens from the dystrophin-deficient sarcolemma of muscle fibers. However, resident antigen-presenting cells, such as myoblasts, myotubes and regenerating immature myofibers, might also play a role in the immune response.


Sujet(s)
Dystrophine/génétique , Thérapie génétique/méthodes , Muscles squelettiques/immunologie , Dystrophie musculaire de l'animal/thérapie , Transgènes/immunologie , Adenoviridae/génétique , Animaux , Production d'anticorps , Dystrophine/déficit , Femelle , Techniques de transfert de gènes , Vecteurs génétiques , Immunité cellulaire , Immunosuppression thérapeutique , Injections musculaires , Mâle , Souris , Souris de lignée C57BL , Muscles squelettiques/anatomopathologie , Dystrophie musculaire de l'animal/immunologie , beta-Galactosidase/métabolisme
9.
Mol Ther ; 4(6): 525-33, 2001 Dec.
Article de Anglais | MEDLINE | ID: mdl-11735336

RÉSUMÉ

Muscle tissue from Duchenne muscular dystrophy patients and the Dmd(mdx/mdx) (hereafter referred to as mdx) mouse is characterized by an abundance of necrotic myofibers and infiltrating macrophages. Both features may provide additional stimulus to the immune response directed against novel antigens, such as those delivered by gene therapy vectors. It has previously been shown that the immune evasion achieved by adeno-associated virus in healthy muscle fails in one model of muscular dystrophy. Here, we examined the immune response to adenoviral vectors and their transgenes in normal and mdx mice. We found that mdx mouse muscles contain 20 times more macrophages and 7 times more dendritic cells than healthy muscles. This higher professional antigen-presenting cell content results in a stronger immune response to antigens that can be directly presented by those cells, including viral antigens and constitutively expressed transgene products. However, we did not detect a significant immune response to beta-galactosidase expressed specifically in muscle, even at high expression levels. This result suggests that cross-presentation is not more effective in mdx mouse muscle, and that targeted vectors and tissue-specific promoters may be useful tools for evasion of the immune response in dystrophic muscle.


Sujet(s)
Cellules dendritiques/immunologie , Macrophages/immunologie , Muscles squelettiques/immunologie , Dystrophie musculaire de l'animal/immunologie , Adenoviridae/génétique , Animaux , Antigènes CD/immunologie , Amorces ADN/composition chimique , Dystrophine/génétique , Cytométrie en flux , Thérapie génétique , Interféron gamma/métabolisme , Souris , Souris de lignée C57BL , Souris de lignée mdx , Souris transgéniques , Dystrophie musculaire de l'animal/thérapie , Réaction de polymérisation en chaîne , Lymphocytes T/immunologie , Lymphocytes T/anatomopathologie , beta-Galactosidase/métabolisme
10.
Hum Mol Genet ; 10(24): 2745-50, 2001 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-11734539

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is characterized by progressive muscle degeneration that results from the absence of dystrophin. Despite null mutations in the dystrophin gene, many DMD patients display a low percentage of dystrophin-positive fibers. These "revertant fibers" are also present in the dystrophin-deficient mdx mouse and are believed to result from alternative splicing or second mutation events that bypass the mutation and restore an open reading frame. However, it is unclear what role dystrophin and the dystrophic pathology might play in revertant fiber formation and accumulation. We have analyzed the role of dystrophin expression and the dystrophic pathology in this process by monitoring revertant fibers in transgenic mdx mice that express truncated dystrophins. We found that newborn transgenic mice displayed approximately the same number of revertant fibers as newborn mdx mice, indicating that expression of a functional dystrophin does not suppress the initiation of revertant fiber formation. Surprisingly, when the transgene encoded a functional dystrophin, revertant fibers were not detected in adult or old mdx mice. In contrast, adult transgenic mice expressing a non-functional dystrophin accumulated increasing numbers of revertant fibers, similar to mdx mice, suggesting that positive selection is required for the persistence of revertant fibers. Finally, we provide evidence that the loss of revertant dystrophin in transgenic mdx muscle fibers overexpressing a functional dystrophin results from displacement of the revertant protein by the transgene-encoded dystrophin.


Sujet(s)
Dystrophine/physiologie , Fibres musculaires squelettiques/anatomopathologie , Animaux , Technique de Western , Régulation négative , Dystrophine/génétique , Exons , Technique d'immunofluorescence , Humains , Souris , Souris de lignée C57BL , Souris de lignée mdx , Souris transgéniques , Myopathie de Duchenne/génétique , Myopathie de Duchenne/anatomopathologie , ARN messager , RT-PCR
11.
J Physiol ; 535(Pt 2): 591-600, 2001 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-11533147

RÉSUMÉ

1. Differences in the effect of age on structure-function relationships of limb muscles of mdx (dystrophin null) and control mice have not been resolved. We tested the hypotheses that, compared with limb muscles from age-matched control mice, limb muscles of 6- to 17-month-old mdx mice are larger but weaker, with lower normalised force and power, whereas those from 24- to 28-month-old mdx mice are smaller and weaker. 2. The maximum isometric tetanic force (P(o)) and power output of limb muscles from 6-, 17-, 24- and 28-month-old mdx and control mice were measured in vitro at 25 degrees C and normalised with respect to cross-sectional area and muscle mass, respectively. 3. Body mass at 6 and 28 months was not significantly different in mdx and control mice, but that of control mice increased 16 % by 17 months and then declined 32 % by 28 months. The body masses of mdx mice declined linearly with age with a decrease of 25 % by 28 months. From 6 to 28 months of age, the range in the decline in the masses of EDL and soleus muscles of mdx and control mice was from 16 to 28 %. The muscle masses of mdx mice ranged from 9 % to 42 % greater than those of control mice at each of the four ages and, even at 28 months, the masses of EDL and soleus muscles of mdx mice were 17 % and 22 % greater than control values. 4. For mdx mice of all ages, muscle hypertrophy was highly effective in the maintenance of control values for absolute force for both EDL and soleus muscles and for absolute power of soleus muscles. Throughout their lifespan, muscles of mdx mice displayed significant weakness with values for specific P(o) and normalised power approximately 20 % lower than values for control mice at each age. For muscles of both strains, normalised force and power decreased approximately 28 % with age, and consequently weakness was more severe in muscles of old mdx than in those of old control mice.


Sujet(s)
Fibres musculaires à contraction rapide/physiologie , Fibres musculaires à contraction lente/physiologie , Dystrophie musculaire de l'animal/physiopathologie , Animaux , Dystrophine/génétique , Contraction isométrique/physiologie , Mâle , Souris , Souris de lignée C57BL , Souris de lignée mdx , Muscles squelettiques/cytologie , Muscles squelettiques/physiologie , Taille d'organe , Organismes exempts d'organismes pathogènes spécifiques
12.
Curr Biol ; 11(16): 1278-82, 2001 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-11525743

RÉSUMÉ

Duchenne Muscular Dystrophy (DMD) originates from deleterious mutations in the dystrophin gene, with a complete loss of the protein product. Subsequently, the disease is manifested in severe striated muscle wasting and death in early adulthood. Dystrophin provides a structural base for the assembly of an integral membrane protein complex. As such, dystrophin deficiency leads to an altered mechanical integrity of the myofiber and a predisposition to contraction-induced damage. However, the development of myofiber degeneration prior to an observed mechanical defect has been documented in various dystrophic models. Although activation of a detrimental signal transduction pathway has been suggested as a probable cause, a specific cellular cascade has yet to be defined. Here, it is shown that murine models of DMD displayed a muscle-specific activation of JNK1. Independent activation of JNK1 resulted in defects in myotube viability and integrity in vitro, similar to a dystrophic phenotype. In addition, direct muscle injection of an adenoviral construct containing the JNK1 inhibitory protein, JIP1, dramatically attenuated the progression of dystrophic myofiber destruction. Taken together, these results suggest that a JNK1-mediated signal cascade is a conserved feature of dystrophic muscle and contributes to the progression of the disease pathogenesis.


Sujet(s)
Mitogen-Activated Protein Kinases/métabolisme , Muscles squelettiques/anatomopathologie , Myopathie de Duchenne/enzymologie , Myopathie de Duchenne/anatomopathologie , Adenoviridae/génétique , Animaux , Cellules cultivées , Activation enzymatique , Protéines à fluorescence verte , Humains , Indicateurs et réactifs/métabolisme , Protéines luminescentes/métabolisme , Système de signalisation des MAP kinases , Souris , Souris de lignée mdx , Souris transgéniques , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/génétique , Muscles squelettiques/enzymologie , Muscles squelettiques/métabolisme , Dystrophie musculaire de l'animal/enzymologie , Dystrophie musculaire de l'animal/génétique , Dystrophie musculaire de l'animal/anatomopathologie , Myopathie de Duchenne/génétique , Myocarde/enzymologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Phosphorylation , Transfection
13.
Cancer Res ; 61(5): 2062-70, 2001 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-11280767

RÉSUMÉ

Secondary lymphoid tissue chemokine (SLC) is a CC chemokine that is selective in its recruitment of naive T cells and dendritic cells (DCs). In the lymph node, SLC is believed to play an important role in the initiation of an immune response by colocalizing naive T cells with DC-presenting antigen. Here, we used SLC as a treatment for tumors established from the poorly immunogenic B16 melanoma. Intratumoral injections of SLC inhibited tumor growth in a CD8+, T cell-dependent manner. SLC elicited a substantial infiltration of DCs and T cells into the tumor, coincident with the antitumor response. We next used SLC gene-modified DCs as a treatment of established tumors. Intratumoral injections of SLC-expressing DCs resulted in tumor growth inhibition that was significantly better than either control DCs or SLC alone. Distal site immunization of tumor-bearing mice with SLC gene-modified DCs pulsed with tumor lysate elicited an antitumor response whereas control DCs did not. We also found that s.c. injection of lysate-pulsed DCs expressing SLC promoted the migration of T cells to the immunization site. This report demonstrates that SLC can both induce antitumor responses and enhance the antitumor immunity elicited by DCs.


Sujet(s)
Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Chimiokines CC/immunologie , Cellules dendritiques/immunologie , Animaux , Chimiokine CCL21 , Chimiokines/génétique , Chimiokines/immunologie , Chimiokines/pharmacologie , Chimiokines CC/génétique , Chimiokines CC/pharmacologie , Cellules dendritiques/physiologie , Femelle , Thérapie génétique , Immunité cellulaire/effets des médicaments et des substances chimiques , Immunité cellulaire/immunologie , Immunothérapie adoptive , Injections intralésionnelles , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Souris , Souris de lignée BALB C , Souris de lignée C57BL
14.
J Muscle Res Cell Motil ; 22(5): 467-75, 2001.
Article de Anglais | MEDLINE | ID: mdl-11964072

RÉSUMÉ

Skeletal muscles of patients with Duchenne muscular dystrophy (DMD) and mdx mice lack dystrophin and are more susceptible to contraction-induced injury than control muscles. Our purpose was to develop an assay based on the high susceptibility to injury of limb muscles in mdx mice for use in evaluating therapeutic interventions. The assay involved two stretches of maximally activated tibialis anterior (TA) muscles in situ. Stretches of 40% strain relative to muscle fiber length were initiated from the plateau of isometric contractions. The magnitude of damage was assessed one minute later by the deficit in isometric force. At all ages (2-19 months), force deficits were four- to seven-fold higher for muscles in mdx compared with control mice. For control muscles, force deficits were unrelated to age, whereas force deficits increased dramatically for muscles in mdx mice after 8 months of age. The increase in susceptibility to injury of muscles from older mdx mice did not parallel similar adverse effects on muscle mass or force production. The in situ stretch protocol of TA muscles provides a valuable assay for investigations of the mechanisms of injury in dystrophic muscle and to test therapeutic interventions for reversing DMD.


Sujet(s)
Contraction musculaire , Muscles squelettiques/physiopathologie , Dystrophie musculaire de l'animal/physiopathologie , Facteurs âges , Animaux , Constitution physique , Stimulation électrique , Contraction isométrique , Souris , Souris de lignée C57BL , Souris de lignée mdx , Muscles squelettiques/anatomopathologie , Dystrophie musculaire de l'animal/anatomopathologie , Organismes exempts d'organismes pathogènes spécifiques , Contrainte mécanique , Facteurs temps
16.
Curr Biol ; 10(21): R795-7, 2000 Nov 02.
Article de Anglais | MEDLINE | ID: mdl-11084353

RÉSUMÉ

A new animal model for studying muscular dystrophy, a mutant form of the nematode Caenorhabditis elegans, brings the power of worm genetics to bear on the search for a cure for this disease; work on this worm has already led to the identification of a novel component that can suppress the mutant phenotype.


Sujet(s)
Caenorhabditis elegans/génétique , Dystrophine/génétique , Myopathie de Duchenne/génétique , Animaux , Modèles animaux de maladie humaine , Dystrophine/métabolisme , Humains , Modèles biologiques , Muscles squelettiques/physiologie , Myopathie de Duchenne/physiopathologie , Mutation
17.
Hum Mol Genet ; 9(17): 2507-15, 2000 Oct 12.
Article de Anglais | MEDLINE | ID: mdl-11030755

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by the lack of expression of the dystrophin protein in muscle tissues. We genetically engineered a mouse model (mdx) of DMD that allowed for the high level and inducible transcription of a dystrophin mini-gene. This was achieved via the tetracycline-responsive transactivator (tTA) system. Multiple analyses confirmed that dystrophin expression in the mice was: (i) tTA dependent; (ii) correctly localized to the sarcolemmal membranes; (iii) capable of preventing the onset of dystrophy; and (iv) effectively blocked by the oral administration of tetracyclines. The model allowed us to somatically extinguish or induce dystrophin gene transcription. Somatic induction of dystrophin transcription prevented the onset of muscular dystrophy in some muscle groups. The levels of phenotypic rescue were influenced, however, by the age of the animals at the time of dystrophin induction. We also found that despite somatic termination of dystrophin gene transcription, the dystrophin protein was found to be associated with the sarcolemmal membrane for at least 26 weeks. Persistent detection of dystrophin was also accompanied by a prolonged protection of the muscle cells from the onset of dystrophy. The findings demonstrated that somatic transfer of the dystrophin gene not only may allow for the prevention of muscular dystrophy in multiple muscle groups, but also may be accompanied by persistent efficacy, secondary to the long-term functional stability of the dystrophin protein in vivo. This model should be useful in future studies concerning the potential of genetic therapy for DMD, as well as other muscle disorders.


Sujet(s)
Dystrophine/génétique , Régulation de l'expression des gènes , Thérapie génétique , Souris de lignée mdx/génétique , Muscles squelettiques/métabolisme , Dystrophie musculaire de l'animal/thérapie , Myopathie de Duchenne/thérapie , Animaux , Croisements génétiques , Dystrophine/biosynthèse , Femelle , Mâle , Souris , Souris transgéniques , Fibres musculaires squelettiques/ultrastructure , Muscles squelettiques/anatomopathologie , Muscles squelettiques/ultrastructure , Dystrophie musculaire de l'animal/génétique , Dystrophie musculaire de l'animal/anatomopathologie , Myopathie de Duchenne/génétique , Myopathie de Duchenne/anatomopathologie , ARN messager/génétique , ARN messager/métabolisme , Tétracycline/pharmacologie , Transactivateurs/génétique , Transcription génétique , Transgènes
18.
J Cell Biol ; 150(6): 1399-410, 2000 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-10995444

RÉSUMÉ

Dystrophin is a multidomain protein that links the actin cytoskeleton to laminin in the extracellular matrix through the dystrophin associated protein (DAP) complex. The COOH-terminal domain of dystrophin binds to two components of the DAP complex, syntrophin and dystrobrevin. To understand the role of syntrophin and dystrobrevin, we previously generated a series of transgenic mouse lines expressing dystrophins with deletions throughout the COOH-terminal domain. Each of these mice had normal muscle function and displayed normal localization of syntrophin and dystrobrevin. Since syntrophin and dystrobrevin bind to each other as well as to dystrophin, we have now generated a transgenic mouse deleted for the entire dystrophin COOH-terminal domain. Unexpectedly, this truncated dystrophin supported normal muscle function and assembly of the DAP complex. These results demonstrate that syntrophin and dystrobrevin functionally associate with the DAP complex in the absence of a direct link to dystrophin. We also observed that the DAP complexes in these different transgenic mouse strains were not identical. Instead, the DAP complexes contained varying ratios of syntrophin and dystrobrevin isoforms. These results suggest that alternative splicing of the dystrophin gene, which naturally generates COOH-terminal deletions in dystrophin, may function to regulate the isoform composition of the DAP complex.


Sujet(s)
Protéines associées à la dystrophine , Dystrophine , Dystrophies musculaires/métabolisme , Actines/métabolisme , Animaux , Sites de fixation/physiologie , Noyau de la cellule/anatomopathologie , Protéines du cytosquelette/analyse , Protéines du cytosquelette/métabolisme , Modèles animaux de maladie humaine , Dystrophine/composition chimique , Dystrophine/génétique , Dystrophine/métabolisme , Exons , Protéines membranaires/analyse , Protéines membranaires/métabolisme , Souris , Souris de lignée C57BL , Souris de lignée mdx , Souris transgéniques , Microsomes/composition chimique , Microsomes/métabolisme , Contraction musculaire/génétique , Fibres musculaires squelettiques/composition chimique , Fibres musculaires squelettiques/métabolisme , Fibres musculaires squelettiques/anatomopathologie , Protéines du muscle/analyse , Protéines du muscle/métabolisme , Dystrophies musculaires/anatomopathologie , Neuropeptides/analyse , Neuropeptides/métabolisme , Structure tertiaire des protéines , Utrophine
19.
Am J Physiol Cell Physiol ; 279(4): C1290-4, 2000 Oct.
Article de Anglais | MEDLINE | ID: mdl-11003610

RÉSUMÉ

Muscle fibers of mdx mice that lack dystrophin are more susceptible to contraction-induced injury, particularly when stretched. In contrast, transgenic mdx (tg-mdx) mice, which overexpress dystrophin, show no morphological or functional signs of dystrophy. Permeabilization disrupts the sarcolemma of fibers from muscles of mdx, tg-mdx, and control mice. We tested the null hypothesis stating that, after single stretches of maximally activated single permeabilized fibers, force deficits do not differ among fibers from extensor digitorum longus muscles of mdx, tg-mdx, or control mice. Fibers were maximally activated by Ca(2+) (pCa 4.5) and then stretched through strains of 10%, 20%, or 30% of fiber length (L(f)) at a velocity of 0.5 L(f)/s. Immediately after each strain, the force deficits were not different for fibers from each of the three groups of mice. When collated with studies of membrane-intact fibers in whole muscles of mdx, tg-mdx, and control mice, these results indicate that dystrophic symptoms do not arise from factors within myofibrils but, rather, from disruption of the sarcolemmal integrity that normally provides protection from contraction-induced injury.


Sujet(s)
Perméabilité des membranes cellulaires/génétique , Contraction musculaire , Fibres musculaires squelettiques/métabolisme , Muscles squelettiques/physiopathologie , Dystrophie musculaire de l'animal/physiopathologie , Animaux , Calcium/métabolisme , Calcium/pharmacologie , Dystrophine/métabolisme , Techniques in vitro , Mâle , Souris , Souris de lignée C57BL , Souris de lignée mdx , Souris transgéniques , Contraction musculaire/génétique , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Sarcolemme/génétique , Sarcolemme/métabolisme , Contrainte mécanique
20.
J Gene Med ; 2(4): 250-9, 2000.
Article de Anglais | MEDLINE | ID: mdl-10953916

RÉSUMÉ

BACKGROUND: The inherent limitations of [E1-]Ad vectors as gene therapy vehicles suggest that further modifications may improve their overall performance profiles. However, Ad vector modifications can have untoward effects on their basic biology, e.g., some helper-virus dependent Ad vectors have been found to be unstable without the presence of preterminal protein (pTP) activities. Despite this concern, we generated a new class of helper-virus independent Ad vector that was multiply deleted for the E1, polymerase, and pTP genes, and investigated the ramifications of these deletions upon several vector performance parameters. METHODS: The construction and propagation of an [E1-, polymerase-, pTP-]Ad vector was achieved with the use of trans-complementing cells co-expressing the Ad E1, polymerase and pTP genes. RESULTS: High titer production of the [E1-, polymerase-, pTP-]Ad vector was successfully accomplished via conventional Ad purification techniques. This unique class of Ad vector was capable of long-term gene transfer in vivo (despite lacking pTP functions) that was concomitant with a significantly decreased hepatic toxicity. CONCLUSIONS: Previous studies had suggested that Ad genome persistence in vivo may be dependent upon the presence of low level vector genome replication and/or pTP functions. Our results suggest that [E1-, polymerase-, pTP-]Ad vectors can overcome these barriers. The further benefits afforded by the use of this class of Ad vector (increased cloning capacity, high level growth, decreased propensity to generate replication competent Ad (RCA), decreased toxicity) suggests that they will be highly beneficial for use in several aspects of human gene therapy.


Sujet(s)
Adenoviridae/génétique , Délétion de gène , Techniques de transfert de gènes , Génome viral , Phosphoprotéines/génétique , Précurseurs de protéines/génétique , Protéines virales/génétique , Protéines E1 d'adénovirus/génétique , Animaux , Lignée cellulaire , DNA-directed DNA polymerase/génétique , Vecteurs génétiques , Virus assistants/génétique , Humains , Souris , Souris de lignée C57BL , Souris SCID , Transduction génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE