GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: The Journal of Pathology, Wiley, Vol. 231, No. 2 ( 2013-10), p. 199-209
    Type of Medium: Online Resource
    ISSN: 0022-3417
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2013
    detail.hit.zdb_id: 3119-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Mass Spectrometry Reviews, Wiley, Vol. 43, No. 1 ( 2024-01), p. 90-105
    Abstract: The dystrophin‐associated protein complex (DAPC) is a highly organized multiprotein complex that plays a pivotal role in muscle fiber structure integrity and cell signaling. The complex is composed of three distinct interacting subgroups, intracellular peripheral proteins, transmembrane glycoproteins, and extracellular glycoproteins subcomplexes. Dystrophin protein nucleates the DAPC and is important for connecting the intracellular actin cytoskeletal filaments to the sarcolemma glycoprotein complex that is connected to the extracellular matrix via laminin, thus stabilizing the sarcolemma during muscle fiber contraction and relaxation. Genetic mutations that lead to lack of expression or altered expression of any of the DAPC proteins are associated with different types of muscle diseases. Hence characterization of this complex in healthy and dystrophic muscle might bring insights into its role in muscle pathogenesis. This review highlights the role of mass spectrometry in characterizing the DAPC interactome as well as post‐translational glycan modifications of some of its components such as α‐dystroglycan. Detection and quantification of dystrophin using targeted mass spectrometry are also discussed in the context of healthy versus dystrophic skeletal muscle.
    Type of Medium: Online Resource
    ISSN: 0277-7037 , 1098-2787
    URL: Issue
    RVK:
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2024
    detail.hit.zdb_id: 787244-6
    detail.hit.zdb_id: 1491946-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: The Journal of Pathology, Wiley, Vol. 248, No. 3 ( 2019-07), p. 339-351
    Abstract: Exon skipping is a promising genetic therapeutic strategy for restoring dystrophin expression in the treatment of Duchenne muscular dystrophy (DMD). The potential for newly synthesized dystrophin to trigger an immune response in DMD patients, however, is not well established. We have evaluated the effect of chronic phosphorodiamidate morpholino oligomer (PMO) treatment on skeletal muscle pathology and asked whether sustained dystrophin expression elicits a dystrophin‐specific autoimmune response. Here, two independent cohorts of dystrophic mdx mice were treated chronically with either 800 mg/kg/month PMO for 6 months ( n  = 8) or 100 mg/kg/week PMO for 12 weeks ( n  = 11). We found that significant muscle inflammation persisted after exon skipping in skeletal muscle. Evaluation of humoral responses showed serum‐circulating antibodies directed against de novo dystrophin in a subset of mice, as assessed both by Western blotting and immunofluorescent staining; however, no dystrophin‐specific antibodies were observed in the control saline‐treated mdx cohorts ( n  = 8) or in aged (12‐month‐old) mdx mice with expanded ‘revertant’ dystrophin‐expressing fibers. Reactive antibodies recognized both full‐length and truncated exon‐skipped dystrophin isoforms in mouse skeletal muscle. We found more antigen‐specific T‐cell cytokine responses (e.g. IFN‐g, IL‐2) in dystrophin antibody‐positive mice than in dystrophin antibody‐negative mice. We also found expression of major histocompatibility complex class I on some of the dystrophin‐expressing fibers along with CD8+ and perforin‐positive T cells in the vicinity, suggesting an activation of cell‐mediated damage had occurred in the muscle. Evaluation of complement membrane attack complex (MAC) deposition on the muscle fibers further revealed lower MAC deposition on muscle fibers of dystrophin antibody‐negative mice than on those of dystrophin antibody‐positive mice. Our results indicate that de novo dystrophin expression after exon skipping can trigger both cell‐mediated and humoral immune responses in mdx mice. Our data highlights the need to further investigate the autoimmune response and its long‐term consequences after exon‐skipping therapy. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
    Type of Medium: Online Resource
    ISSN: 0022-3417 , 1096-9896
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2019
    detail.hit.zdb_id: 3119-7
    detail.hit.zdb_id: 1475280-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Arthritis & Rheumatism, Wiley, Vol. 64, No. 11 ( 2012-11), p. 3750-3759
    Abstract: It is generally believed that muscle weakness in patients with polymyositis and dermatomyositis is due to autoimmune and inflammatory processes. However, it has been observed that there is a poor correlation between the suppression of inflammation and a recovery of muscle function in these patients. This study was undertaken to examine whether nonimmune mechanisms also contribute to muscle weakness. In particular, it has been suggested that an acquired deficiency of AMP deaminase 1 (AMPD1) may be responsible for muscle weakness in myositis. Methods We performed comprehensive functional, behavioral, histologic, molecular, enzymatic, and metabolic assessments before and after the onset of inflammation in a class I major histocompatibility complex (MHC)–transgenic mouse model of autoimmune inflammatory myositis. Results Muscle weakness and metabolic disturbances were detectable in the mice prior to the appearance of infiltrating mononuclear cells. Force contraction analysis of muscle function revealed that weakness was correlated with AMPD1 expression and was myositis specific. Decreasing AMPD1 expression resulted in decreased muscle strength in healthy mice. Fiber typing suggested that fast‐twitch muscles were converted to slow‐twitch muscles as myositis progressed, and microarray results indicated that AMPD1 and other purine nucleotide pathway genes were suppressed, along with genes essential to glycolysis. Conclusion These data suggest that an AMPD1 deficiency is acquired prior to overt muscle inflammation and is responsible, at least in part, for the muscle weakness that occurs in the mouse model of myositis. AMPD1 is therefore a potential therapeutic target in myositis.
    Type of Medium: Online Resource
    ISSN: 0004-3591 , 1529-0131
    URL: Issue
    RVK:
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2012
    detail.hit.zdb_id: 2014367-9
    detail.hit.zdb_id: 127294-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Muscle & Nerve, Wiley, Vol. 39, No. 5 ( 2009-05), p. 674-682
    Abstract: Expression of major histocompatibility complex (MHC) class I in skeletal muscle fibers is an early and consistent finding in inflammatory myopathies. To test if MHC class I has a primary role in muscle impairment, we used transgenic mice with inducible overexpression of MHC class I in their skeletal muscle cells. Contractile function was studied in isolated extensor digitorum longus (EDL, fast‐twitch) and soleus (slow‐twitch) muscles. We found that EDL was smaller, whereas soleus muscle was slightly larger. Both muscles generated less absolute force in myopathic compared with control mice; however, when force was expressed per cross‐sectional area, only soleus muscle generated less force. Inflammation was markedly increased, but no changes were found in the activities of key mitochondrial and glycogenolytic enzymes in myopathic mice. The induction of MHC class I results in muscle atrophy and an intrinsic decrease in force‐generation capacity. These observations may have important implications for our understanding of the pathophysiological processes of muscle weakness seen in inflammatory myopathies. Muscle Nerve, 2008
    Type of Medium: Online Resource
    ISSN: 0148-639X , 1097-4598
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2009
    detail.hit.zdb_id: 1476641-3
    detail.hit.zdb_id: 438353-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Journal of Inherited Metabolic Disease, Wiley, Vol. 42, No. 6 ( 2019-11), p. 1088-1096
    Abstract: Abundance of urea cycle enzymes in the liver is regulated by dietary protein intake. Although urea cycle enzyme levels rise in response to a high‐protein (HP) diet, signaling networks that sense dietary protein intake and trigger changes in expression of urea cycle genes have not been identified. The aim of this study was to identify signaling pathway(s) that respond to changes in protein intake and regulate expression of urea cycle genes in mice and human hepatocytes. Mice were adapted to either HP or low‐protein diets followed by isolation of liver protein and mRNA and integrated analysis of the proteomic and transcriptomic data. HP diet led to increased expression of mRNA and enzymes in amino acid degradation pathways and decreased expression of mRNA and enzymes in carbohydrate and fat metabolism, which implicated adenosine monophosphate‐activated protein kinase (AMPK) as a possible regulator. Primary human hepatocytes, treated with 5‐aminoimidazole‐4‐carboxamide ribonucleotide (AICAR) an activator of AMPK, were used to test whether AMPK regulates expression of urea cycle genes. The abundance of carbamoylphosphate synthetase 1 and ornithine transcarbamylase mRNA increased in hepatocytes treated with AICAR, which supports a role for AMPK signaling in regulation of the urea cycle. Because AMPK is either a target of drugs used to treat type‐2 diabetes, these drugs might increase the expression of urea cycle enzymes in patients with partial urea cycle disorders, which could be the basis of a new therapeutic approach.
    Type of Medium: Online Resource
    ISSN: 0141-8955 , 1573-2665
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2019
    detail.hit.zdb_id: 2006875-X
    detail.hit.zdb_id: 438341-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Muscle & Nerve, Wiley, Vol. 39, No. 5 ( 2009-05), p. 591-602
    Abstract: The availability of animal models for Duchenne muscular dystrophy has led to extensive preclinical research on potential therapeutics. Few studies have focused on reliability and sensitivity of endpoints for mdx mouse drug trials. Therefore, we sought to compare a wide variety of reported and novel endpoint measures in exercised mdx and normal control mice at 10, 20, and 40 weeks of age. Statistical analysis as well as power calculations for expected effect sizes in mdx preclinical drug trials across different ages showed that body weight, normalized grip strength, horizontal activity, rest time, cardiac function measurements, blood pressure, total central/peripheral nuclei per fiber, and serum creatine kinase are the most effective measurements for detecting drug‐induced changes. These data provide an experimental basis upon which standardization of preclinical drug testing can be developed. Muscle Nerve, 2008
    Type of Medium: Online Resource
    ISSN: 0148-639X , 1097-4598
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2009
    detail.hit.zdb_id: 1476641-3
    detail.hit.zdb_id: 438353-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: The Journal of Clinical Pharmacology, Wiley, Vol. 59, No. 7 ( 2019-07), p. 979-988
    Abstract: Duchenne muscular dystrophy (DMD) is an inherited neuromuscular disorder occurring in boys and caused by mutations in the dystrophin gene. Vamorolone is a first‐generation delta‐9,11 compound that has favorable efficacy and side effect profiles relative to classical glucocorticoids. The pharmacokinetics (PK) of oral vamorolone were assessed in parallel‐group studies in healthy men (phase 1, n = 86) and boys with DMD (phase 2a, n = 48) during 14 days of once‐daily dosing with a range of doses. Vamorolone exhibited moderate variability in PK, with the maximum plasma concentration usually occurring at 2‐4 hours and a half‐life of approximately 2 hours for all doses and days examined. Population PK modeling of all data together indicated that the PK of vamorolone can be well described by a 1‐compartment model with zero‐order absorption. Both men and boys showed a dose‐linearity of PK parameters for the doses examined, with no accumulation of the drug during daily dosing. Ingestion with food resulted in markedly enhanced absorption of the drug, as tested in healthy men. There were similar PK of vamorolone in healthy men and DMD boys with apparent clearance averaging 2.0 L/h/kg in men and 1.7 L/h/kg in boys. Overall, vamorolone exhibited well‐behaved linear PK, with similar profiles in healthy men and boys with DMD, moderate variability in PK parameters, and absorption and disposition profiles similar to those of classical glucocorticoids.
    Type of Medium: Online Resource
    ISSN: 0091-2700 , 1552-4604
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2019
    detail.hit.zdb_id: 2010253-7
    detail.hit.zdb_id: 188980-1
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Arthritis & Rheumatology, Wiley, Vol. 72, No. 7 ( 2020-07), p. 1170-1183
    Abstract: Muscle inflammation is a feature in myositis and Duchenne muscular dystrophy ( DMD ). Autoimmune mechanisms are thought to contribute to muscle weakness in patients with myositis. However, a lack of correlation between the extent of inflammatory cell infiltration and muscle weakness indicates that nonimmune pathologic mechanisms may play a role. The present study focused on 2 micro RNA (mi RNA ) sets previously identified as being elevated in the muscle of patients with DMD —an “inflammatory” mi RNA set that is dampened with glucocorticoids, and a “dystrophin‐targeting” mi RNA set that inhibits dystrophin translation—to test the hypothesis that these mi RNA s are similarly dysregulated in the muscle of patients with myositis, and could contribute to muscle weakness and disease severity. Methods A major histocompatibility complex class I–transgenic mouse model of myositis was utilized to study gene and mi RNA expression and histologic features in the muscle tissue, with the findings validated in human muscle biopsy tissue from 6 patients with myositis. Mice were classified as having mild or severe myositis based on transgene expression, body weight, histologic disease severity, and muscle strength/weakness. Results In mice with severe myositis, muscle tissue showed mononuclear cell infiltration along with elevated expression of type I interferon and NF ‐κB–regulated genes, including Tlr7 (3.8‐fold increase, P 〈 0.05). Furthermore, mice with severe myositis showed elevated expression of inflammatory mi RNA s (miR‐146a, miR‐142‐3p, miR‐142‐5p, miR‐455‐3p, and miR‐455‐5p; ~3–40‐fold increase, P 〈 0.05) and dystrophin‐targeting mi RNA s (miR‐146a, miR‐146b, miR‐31, and miR‐223; ~3–38‐fold increase, P 〈 0.05). Bioinformatics analyses of chromatin immunoprecipitation sequencing (ChIP‐seq) data identified at least one NF ‐κB consensus element within the promoter/enhancer regions of these mi RNA s. Western blotting and immunofluorescence analyses of the muscle tissue from mice with severe myositis demonstrated reduced levels of dystrophin. In addition, elevated levels of NF ‐κB–regulated genes, TLR 7 , and mi RNA s along with reduced dystrophin levels were observed in muscle biopsy tissue from patients with histologically severe myositis. Conclusion These data demonstrate that an acquired dystrophin deficiency may occur through NF ‐κB–regulated mi RNA s in myositis, thereby suggesting a unifying theme in which muscle injury, inflammation, and weakness are perpetuated both in myositis and in DMD .
    Type of Medium: Online Resource
    ISSN: 2326-5191 , 2326-5205
    URL: Issue
    RVK:
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2020
    detail.hit.zdb_id: 127294-9
    detail.hit.zdb_id: 2754614-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Arthritis & Rheumatism, Wiley, Vol. 65, No. 12 ( 2013-12), p. 3248-3258
    Abstract: Myositis is characterized by severe muscle weakness. We and others have previously shown that endoplasmic reticulum (ER) stress plays a role in the pathogenesis of myositis. The present study was undertaken to identify perturbed pathways and assess their contribution to muscle disease in a mouse myositis model. Methods Stable isotope labeling with amino acids in cell culture (SILAC) was used to identify alterations in the skeletal muscle proteome of myositic mice in vivo. Differentially altered protein levels identified in the initial comparisons were validated using a liquid chromatography tandem mass spectrometry spike‐in strategy and further confirmed by immunoblotting. In addition, we evaluated the effect of a proteasome inhibitor, bortezomib, on the disease phenotype, using well‐standardized functional, histologic, and biochemical assessments. Results With the SILAC technique we identified significant alterations in levels of proteins belonging to the ER stress response, ubiquitin proteasome pathway (UPP), oxidative phosphorylation, glycolysis, cytoskeleton, and muscle contractile apparatus categories. We validated the myositis‐related changes in the UPP and demonstrated a significant increase in the ubiquitination of muscle proteins as well as a specific increase in ubiquitin carboxyl‐terminal hydrolase isozyme L1 (UCHL‐1) in myositis, but not in muscle affected by other dystrophies or normal muscle. Inhibition of the UPP with bortezomib significantly improved muscle function and also significantly reduced tumor necrosis factor α expression in the skeletal muscle of mice with myositis. Conclusion Our findings indicate that ER stress activates downstream UPPs and contributes to muscle degeneration and that UCHL‐1 is a potential biomarker for disease progression. UPP inhibition offers a potential therapeutic strategy for myositis.
    Type of Medium: Online Resource
    ISSN: 0004-3591 , 1529-0131
    URL: Issue
    RVK:
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2013
    detail.hit.zdb_id: 2014367-9
    detail.hit.zdb_id: 127294-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...