9 research outputs found

    The E3 ubiquitin ligase TRIM62 and inflammation-induced skeletal muscle atrophy

    Get PDF
    Introduction: Intensive care unit (ICU)-acquired weakness (ICUAW) complicates the disease course of critically ill patients. Inflammation and acute-phase response occur directly within myocytes and contribute to ICUAW. We observed that TRIM62, an E3-ubiquitin ligase and modifier of inflammation, is increased in skeletal muscle of ICUAW patients. We investigated regulation and function of muscular TRIM62 in critical illness. Methods: Twenty-six critically ill patients with Sequential Organ Failure Assessment scores more than or equal to 8 underwent two skeletal muscle biopsies from the vastus lateralis at median days 5 and 15 in ICU. Four patients undergoing elective orthopedic surgery served as controls. TRIM62 expression and protein content was analyzed in these biopsies. Kinetics of Trim62, Atrogin1 and MuRF1 expression were determined in the gastrocnemius/plantaris and tibialis anterior from mouse models of inflammation, denervation and starvation induced muscle atrophy to differentiate between these contributors of ICUAW. Cultured myocytes were used for mechanistic analyses. Results: TRIM62 expression and protein content was increased early and remained elevated in muscle from critically ill patients. In all three animal models muscular Trim62 expression was early and continuously increased. Trim62 was expressed in myocytes and its overexpression activated the atrophy-inducing activator protein 1 signal transduction pathway. Knockdown of Trim62 by siRNA inhibited lipopolysaccharide induced interleukin-6 expression. Conclusions: TRIM62 is activated in muscle of critically ill patients. It could play a role in the pathogenesis of ICUAW by activating and maintaining inflammation in myocytes. Trial registration: Current Controlled Trials, ISRCTN77569430. Registered 13 February 2008

    Muscle RING-finger 2 and 3 maintain striated-muscle structure and function

    Get PDF
    Background: The Muscle-specific RING-finger (MuRF) protein family of E3 ubiquitin ligases is important for maintenance of muscular structure and function. MuRF proteins mediate adaptation of striated muscles to stress. MuRF2 and MuRF3 bind to microtubules and are implicated in sarcomere formation with noticeable functional redundancy. However, if this redundancy is important for muscle function in vivo is unknown. Our objective was to investigate cooperative function of MuRF2 and MuRF3 in the skeletal muscle and the heart in vivo. Methods: MuRF2 and MuRF3 double knockout mice (DKO) were generated and phenotypically characterized. Skeletal muscle and the heart were investigated by morphological measurements, histological analyses, electron microscopy, immunoblotting, and real-time PCR. Isolated muscles were subjected to in vitro force measurements. Cardiac function was determined by echocardiography and working heart preparations. Function of cardiomyocytes was measured in vitro. Cell culture experiments and mass-spectrometry were used for mechanistic analyses. Results: DKO mice showed a protein aggregate myopathy in skeletal muscle. Maximal force development was reduced in DKO soleus and extensor digitorum longus. Additionally, a fibre type shift towards slow/type I fibres occurred in DKO soleus and extensor digitorum longus. MuRF2 and MuRF3-deficient hearts showed decreased systolic and diastolic function. Further analyses revealed an increased expression of the myosin heavy chain isoform beta/slow and disturbed calcium handling as potential causes for the phenotype in DKO hearts. Conclusions: The redundant function of MuRF2 and MuRF3 is important for maintenance of skeletal muscle and cardiac structure and function in vivo

    CSF2-dependent monocyte education in the pathogenesis of ANCA-induced glomerulonephritis

    Get PDF
    OBJECTIVES: Myeloid cell activation by antineutrophil cytoplasmic antibody (ANCA) is pivotal for necrotising vasculitis, including necrotising crescentic glomerulonephritis (NCGN). In contrast to neutrophils, the contribution of classical monocyte (CM) and non-classical monocyte (NCM) remains poorly defined. We tested the hypothesis that CMs contribute to antineutrophil cytoplasmic antibody-associated vasculitis (AAV) and that colony-stimulating factor-2 (CSF2, granulocyte-macrophage colony-stimulating factor (GM-CSF)) is an important monocyte-directed disease modifier.METHODS: Myeloperoxidase (MPO)-immunised MPO(-/-) mice were transplanted with haematopoietic cells from wild-type (WT) mice, C-C chemokine receptor 2 (CCR2)(-/-) mice to abrogate CM, or transcription factor CCAAT-enhancer-binding protein beta (C/EBPß)(-/-) mice to reduce NCM, respectively. Monocytes were stimulated with CSF2, and CSF2 receptor subunit beta (CSF2rb)-deficient mice were used. Urinary monocytes and CSF2 were quantified and kidney expression was analysed. CSF2-blocking antibody was used in the nephrotoxic nephritis (NTN) model. RESULTS: Compared with WT mice, CCR2(-/-) chimeric mice showed reduced circulating CM and were protected from NCGN. C/EBPß(-/-) chimeric mice lacked NCM but developed NCGN similar to WT chimeric mice. Kidney and urinary CSF2 were upregulated in AAV mice. CSF2 increased the ability of ANCA-stimulated monocytes to generate interleukin-1ß and to promote T17(H) effector cell polarisation. CSF2rb(-/-) chimeric mice harboured reduced numbers of kidney T17(H) cells and were protected from NCGN. CSF2 neutralisation reduced renal damage in the NTN model. Finally, patients with active AAV displayed increased urinary CM numbers, CSF2 levels and expression of GM-CSF in infiltrating renal cells. CONCLUSIONS: CMs but not NCMs are important for inducing kidney damage in AAV. CSF2 is a crucial pathological factor by modulating monocyte proinflammatory functions and thereby T17(H) cell polarisation

    Angiotensin II induces skeletal muscle atrophy by activating TFEB-mediated MuRF1 expression

    No full text
    RATIONALE: Skeletal-muscle wasting with accompanying cachexia is a life threatening complication in congestive heart failure (CHF). The molecular mechanisms are imperfectly understood, although an activated renin-angiotensin aldosterone system (RAAS) has been implicated. Angiotensin (Ang) II induces skeletal muscle atrophy in part by increased muscle-enriched E3 ubiquitin ligase muscle RING-finger-1 (MuRF1) expression, which may involve protein kinase-D (PKD1). OBJECTIVE: To elucidate the molecular mechanism of Ang II-induced skeletal muscle wasting. METHODS AND RESULTS: A cDNA expression screen identified the lysosomal hydrolase-coordinating transcription factor EB (TFEB) as novel regulator of the human MuRF1 promoter. TFEB played a key role in regulating Ang II-induced skeletal muscle atrophy by transcriptional control of MuRF1 via conserved E-box elements. Inhibiting TFEB with siRNA prevented Ang II induced MuRF1 expression and atrophy. The histone deactylase-5 (HDAC5), which was directly bound to and colocalized with TFEB, inhibited TFEB-induced MuRF1 expression. The inhibition of TFEB by HDAC5 was reversed by PKD1, which was associated with HDAC5 and mediated its nuclear export. Mice lacking PKD1 in skeletal myocytes were resistant to Ang II-induced muscle wasting. CONCLUSIONS: We propose that elevated Ang II serum concentrations, as occur in CHF patients, could activate the PKD1/HDAC5/TFEB/MuRF1 pathway to induce skeletal muscle wasting

    Early type II fiber atrophy in intensive care unit patients with nonexcitable muscle membrane

    No full text
    OBJECTIVE: Intensive care unit-acquired weakness indicates increased morbidity and mortality. Nonexcitable muscle membrane after direct muscle stimulation develops early and predicts intensive care unit-acquired weakness in sedated, mechanically ventilated patients. A comparison of muscle histology at an early stage in intensive care unit-acquired weakness has not been done. We investigated whether nonexcitable muscle membrane indicates fast-twitch myofiber atrophy during the early course of critical illness. DESIGN, SETTING, AND PATIENTS: We studied patients at increased risk for development of intensive care unit-acquired weakness with Sepsis-related Organ Failure Assessment scores ≥8 on 3 of 5 consecutive days within the first week in the intensive care unit. Electrophysiological compound muscle action potentials after direct muscle stimulation and muscle biopsies were obtained at median days 7 and 5, respectively. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Patients with nonexcitable muscle membranes (n = 15) showed smaller median type II cross-sectional areas (p < .05), whereas type I muscle fibers did not compared with patients with preserved muscle membrane excitability (compound muscle action potentials after direct muscle stimulation ≥3.0 mV; n = 9). We also observed decreased mRNA transcription levels of myosin heavy chain isoform IIa and a lower densitometric ratio of fast-to-slow myosin heavy chain protein content. CONCLUSION:: We suggest that electrophysiological nonexcitable muscle membrane predicts preferential type II fiber atrophy in intensive care unit patients during early critical illness

    Dynamics of myosin degradation in intensive care unit-acquired weakness during severe critical illness

    No full text
    IMPORTANCE: Intensive care unit (ICU)-acquired muscle wasting is a devastating complication leading to persistent weakness and functional disability. The mechanisms of this myopathy are unclear, but a disturbed balance of myosin heavy chain (MyHC) is implicated. OBJECTIVE: To investigate pathways of myosin turnover in severe critically ill patients at high risk of ICU-acquired weakness. DESIGN: Prospective, mechanistic, observational study. SETTING: Interdisciplinary ICUs of a university hospital. PARTICIPANTS: Twenty-nine patients with Sequential Organ Failure Assessment (SOFA) scores of at least 8 on three consecutive days within the first 5 days in ICU underwent two consecutive open skeletal muscle biopsies from the vastus lateralis at median days 5 and 15. Control biopsy specimens were from healthy subjects undergoing hip-replacement surgery. INTERVENTIONS: None. MAIN OUTCOME(S) AND MEASURE(S): Time-dependent changes in myofiber architecture, MyHC synthesis, and degradation were determined and correlated with clinical data. RESULTS: ICU-acquired muscle wasting was characterized by early, disrupted myofiber ultrastructure followed by atrophy of slow- and fast-twitch myofibers at later time points. A rapid decrease in MyHC mRNA and protein expression occurred by day 5 and persisted at day 15 (P < 0.05). Expression of the atrophy genes MuRF-1 and Atrogin1 was increased at day 5 (P < 0.05). Early MuRF-1 protein content was closely associated with late myofiber atrophy and the severity of weakness. CONCLUSIONS AND RELEVANCE: Decreased synthesis and increased degradation of MyHCs contribute to ICU-acquired muscle wasting. The rates and time frames suggest that pathogenesis of muscle failure is initiated very early during critical illness. The persisting reduction of MyHC suggests that sustained treatment is required
    corecore