51
|
Tajsic T, Morrell NW. Smooth muscle cell hypertrophy, proliferation, migration and apoptosis in pulmonary hypertension. Compr Physiol 2013; 1:295-317. [PMID: 23737174 DOI: 10.1002/cphy.c100026] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Pulmonary hypertension is a multifactorial disease characterized by sustained elevation of pulmonary vascular resistance (PVR) and pulmonary arterial pressure (PAP). Central to the pathobiology of this disease is the process of vascular remodelling. This process involves structural and functional changes to the normal architecture of the walls of pulmonary arteries (PAs) that lead to increased muscularization of the muscular PAs, muscularization of the peripheral, previously nonmuscular, arteries of the respiratory acinus, formation of neointima, and formation of plexiform lesions. Underlying or contributing to the development of these lesions is hypertrophy, proliferation, migration, and resistance to apoptosis of medial cells and this article is concerned with the cellular and molecular mechanisms of these processes. In the first part of the article we focus on the concept of smooth muscle cell phenotype and the difficulties surrounding the identification and characterization of the cell/cells involved in the remodelling of the vessel media and we review the general mechanisms of cell hypertrophy, proliferation, migration and apoptosis. Then, in the larger part of the article, we review the factors identified thus far to be involved in PH intiation and/or progression and review and discuss their effects on pulmonary artery smooth muscle cells (PASMCs) the predominant cells in the tunica media of PAs.
Collapse
Affiliation(s)
- Tamara Tajsic
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | | |
Collapse
|
52
|
3D scaffolds in tissue engineering and regenerative medicine: beyond structural templates? ACTA ACUST UNITED AC 2013. [DOI: 10.4155/pbp.13.21] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
53
|
Ohashi N, Nomura W, Narumi T, Tamamura H. Peptide-based ligand screening and functional analysis of protein kinase C. Biopolymers 2013; 100:613-20. [PMID: 23897302 DOI: 10.1002/bip.22324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 04/11/2013] [Accepted: 06/04/2013] [Indexed: 11/06/2022]
Abstract
Protein kinase C (PKC) plays an important role in cell signaling pathways and is implicated in disorders ranging from cancer to Alzheimer's disease. Highly potent PKC ligands as therapeutic drugs have not been developed to date and useful methodologies for controlling PKC activation in defined areas are necessary to analyze precise PKC functions in cells. Studies focused on the development of PKC ligand screening systems and methods for regulation of PKC activation have been performed in our laboratory. In this review, our ligand screening methods involving synthetic peptides and solvatochromic fluorescent dye-labeled small compounds are summarized and the technique of spatio-temporal manipulation of PKC activation by caging strategies is introduced.
Collapse
Affiliation(s)
- Nami Ohashi
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, 101-0062, Japan
| | | | | | | |
Collapse
|
54
|
Gould DW, Lahart I, Carmichael AR, Koutedakis Y, Metsios GS. Cancer cachexia prevention via physical exercise: molecular mechanisms. J Cachexia Sarcopenia Muscle 2013; 4:111-24. [PMID: 23239116 PMCID: PMC3684702 DOI: 10.1007/s13539-012-0096-0] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 11/14/2012] [Indexed: 01/09/2023] Open
Abstract
Cancer cachexia is a debilitating consequence of disease progression, characterised by the significant weight loss through the catabolism of both skeletal muscle and adipose tissue, leading to a reduced mobility and muscle function, fatigue, impaired quality of life and ultimately death occurring with 25-30 % total body weight loss. Degradation of proteins and decreased protein synthesis contributes to catabolism of skeletal muscle, while the loss of adipose tissue results mainly from enhanced lipolysis. These mechanisms appear to be at least, in part, mediated by systemic inflammation. Exercise, by virtue of its anti-inflammatory effect, is shown to be effective at counteracting the muscle catabolism by increasing protein synthesis and reducing protein degradation, thus successfully improving muscle strength, physical function and quality of life in patients with non-cancer-related cachexia. Therefore, by implementing appropriate exercise interventions upon diagnosis and at various stages of treatment, it may be possible to reverse protein degradation, while increasing protein synthesis and lean body mass, thus counteracting the wasting seen in cachexia.
Collapse
Affiliation(s)
- Douglas W Gould
- School of Sport, Performing Arts and Leisure, Department of Physical Activity, Exercise and Health, University of Wolverhampton, Walsall, West Midlands, UK,
| | | | | | | | | |
Collapse
|
55
|
Wing SS. Deubiquitinases in skeletal muscle atrophy. Int J Biochem Cell Biol 2013; 45:2130-5. [PMID: 23680672 DOI: 10.1016/j.biocel.2013.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 05/03/2013] [Indexed: 11/29/2022]
Abstract
The ubiquitin proteasome system plays a critical role in skeletal muscle atrophy. A large body of research has revealed that many ubiquitin ligases are induced and play an important role in mediating the wasting. However, relatively little is known about the roles of deubiquitinases in this process. Although it might be expected that deubiquitinases would be downregulated in atrophying muscles to promote ubiquitination and degradation of muscle proteins, this has not to date been demonstrated. Instead several deubiquitinases are induced in atrophying muscle, in particular USP19 and USP14. USP19, USP2 and A20 are also implicated in myogenesis. USP19 has been most studied to date. Its expression is increased in both systemic and disuse forms of atrophy and can be regulated through a p38 MAP kinase signaling pathway. In cultured muscle cells, it decreases the expression of myofibrillar proteins by apparently suppressing their transcription indicating that the ubiquitin proteasome system may be activated in skeletal muscle to not only increase protein degradation, but also to suppress protein synthesis. Deubiquitinases may be upregulated in atrophy in order to maintain the pool of free ubiquitin required for the increased overall conjugation and degradation of muscle proteins as well as to regulate the stability and function of proteins that are essential in mediating the wasting. Although deubiquitinases are not well studied, these early insights indicate that some of these enzymes play important roles and may be therapeutic targets for the prevention and treatment of muscle atrophy. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
Affiliation(s)
- Simon S Wing
- Polypeptide Laboratory, Department of Medicine, McGill University and McGill University Health Centre, Canada.
| |
Collapse
|
56
|
Park JH, Chung EJ, Kwon HJ, Im SS, Lim JG, Song DK. Protective effect of melatonin on TNF-α-induced muscle atrophy in L6 myotubes. J Pineal Res 2013; 54:417-425. [PMID: 23278522 DOI: 10.1111/jpi.12036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 12/03/2012] [Indexed: 01/22/2023]
Abstract
Muscle atrophy, characterized by decreased cell number and size, is a serious concern for patients afflicted with inflammatory diseases. Mounting evidence indicates that tumor necrosis factor alpha (TNF-α) plays a critical role in muscle atrophy in a number of clinical settings. We hypothesize that reactive oxygen species (ROS) mediate TNF-α-induced muscle cell death and hypotrophy. Recently, melatonin has attracted attention because of its free-radical scavenging and antioxidant properties. The aim of the current study was to evaluate the possible protective role of melatonin in TNF-α-induced muscle cell death and hypotrophy in rat L6 myotubes. To examine this possible role, L6 myotubes were exposed to various concentrations of recombinant TNF-α for 24 hr. We found that TNF-α at a concentration of 100 ng/mL induced ROS generation and decreased cell viability. Further analysis revealed that apoptosis, but not autophagy, may be important for TNF-α-induced cell death. Melatonin significantly attenuated TNF-α-induced ROS generation and apoptosis. In addition, decreased muscle fiber diameter and increased muscle cell proteolysis by TNF-α was highly attenuated by treatment with melatonin. The effects of melatonin were mediated neither through its plasmalemmal receptors nor by modulating the nuclear factor kappa B pathway activated by TNF-α. Taken together, these results suggest that TNF-α may mediate ROS-induced muscle cell death and hypotrophy and that melatonin may be a useful tool for protecting against muscle atrophy stemming from inflammatory diseases.
Collapse
Affiliation(s)
- Jae-Hyung Park
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | | | | | | | | | | |
Collapse
|
57
|
Shen Q, Shi P, Gao M, Yu X, Liu Y, Luo L, Zhu Y. Progress on materials and scaffold fabrications applied to esophageal tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2013; 33:1860-6. [DOI: 10.1016/j.msec.2013.01.064] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 01/01/2013] [Accepted: 01/26/2013] [Indexed: 12/29/2022]
|
58
|
Stasko SA, Hardin BJ, Smith JD, Moylan JS, Reid MB. TNF signals via neuronal-type nitric oxide synthase and reactive oxygen species to depress specific force of skeletal muscle. J Appl Physiol (1985) 2013; 114:1629-36. [PMID: 23558387 DOI: 10.1152/japplphysiol.00871.2012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
TNF promotes skeletal muscle weakness, in part, by depressing specific force of muscle fibers. This is a rapid, receptor-mediated response, in which TNF stimulates cellular oxidant production, causing myofilament dysfunction. The oxidants appear to include nitric oxide (NO); otherwise, the redox mechanisms that underlie this response remain undefined. The current study tested the hypotheses that 1) TNF signals via neuronal-type NO synthase (nNOS) to depress specific force, and 2) muscle-derived reactive oxygen species (ROS) are essential co-mediators of this response. Mouse diaphragm fiber bundles were studied using live cell assays. TNF exposure increased general oxidant activity (P < 0.05; 2',7'-dichlorodihydrofluorescein diacetate assay) and NO activity (P < 0.05; 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate assay) and depressed specific force across the full range of stimulus frequencies (1-300 Hz; P < 0.05). These responses were abolished by pretreatment with N(ω)-nitro-L-arginine methyl ester (L-NAME; a nonspecific inhibitor of NOS activity), confirming NO involvement. Genetic nNOS deficiency replicated L-NAME effects on TNF-treated muscle, diminishing NO activity (-80%; P < 0.05) and preventing the decrement in specific force (P < 0.05). Comparable protection was achieved by selective depletion of muscle-derived ROS. Pretreatment with either SOD (degrades superoxide anion) or catalase (degrades hydrogen peroxide) depressed oxidant activity in TNF-treated muscle and abolished the decrement in specific force. These findings indicate that TNF signals via nNOS to depress contractile function, a response that requires ROS and NO as obligate co-mediators.
Collapse
Affiliation(s)
- Shawn A Stasko
- Department of Physiology and Center for Muscle Biology, University of Kentucky, Lexington, Kentucky 40356-0298, USA
| | | | | | | | | |
Collapse
|
59
|
Chen LJ, Wei SY, Chiu JJ. Mechanical regulation of epigenetics in vascular biology and pathobiology. J Cell Mol Med 2013; 17:437-48. [PMID: 23551392 PMCID: PMC3822644 DOI: 10.1111/jcmm.12031] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 01/09/2013] [Indexed: 12/16/2022] Open
Abstract
Vascular endothelial cells (ECs) and smooth muscle cells (VSMCs) are constantly exposed to haemodynamic forces, including blood flow-induced fluid shear stress and cyclic stretch from blood pressure. These forces modulate vascular cell gene expression and function and, therefore, influence vascular physiology and pathophysiology in health and disease. Epigenetics, including DNA methylation, histone modification/chromatin remodelling and RNA-based machinery, refers to the study of heritable changes in gene expression that occur without changes in the DNA sequence. The role of haemodynamic force-induced epigenetic modifications in the regulation of vascular gene expression and function has recently been elucidated. This review provides an introduction to the epigenetic concepts that relate to vascular physiology and pathophysiology. Through the studies of gene expression, cell proliferation, angiogenesis, migration and pathophysiological states, we present a conceptual framework for understanding how mechanical force-induced epigenetic modifications work to control vascular gene expression and function and, hence, the development of vascular disorders. This research contributes to our knowledge of how the mechanical environment impacts the chromatin state of ECs and VSMCs and the consequent cellular behaviours.
Collapse
Affiliation(s)
- Li-Jing Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 350, Taiwan
| | | | | |
Collapse
|
60
|
Boccafoschi F, Mosca C, Ramella M, Valente G, Cannas M. The effect of mechanical strain on soft (cardiovascular) and hard (bone) tissues: common pathways for different biological outcomes. Cell Adh Migr 2013; 7:165-73. [PMID: 23287581 PMCID: PMC3954035 DOI: 10.4161/cam.23020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mechanical stress plays a pivotal role in developing and maintaining tissues functionalities. Cells are constantly subjected to strain and compressive forces that are sensed by specialized membrane mechanosensors and converted in biochemical signals able to differently influence cellular behavior in terms of surviving, differentiation and extracellular matrix remodeling. This review focuses on the effects of mechanical strain on soft and hard tissues. Unexpectedly, different cells share almost the same membrane mechanosensors and the relative intracellular pathways, but to ultimately obtain very different biological effects. The events occurring in cardiovascular and bone tissues are treated in details, showing that integrins, cadherins, growth factor receptors and ions channels specifically expressed in the different tissues are the major actors of the sight. However, MAPkinases and RhoGTPases are mainly involved in the biochemical intracellular signaling directed to nuclear modifications.
Collapse
Affiliation(s)
- Francesca Boccafoschi
- Department of Health Sciences, University of Piemonte Orientale A. Avogadro, Novara, Italy.
| | | | | | | | | |
Collapse
|
61
|
Tang K, Murano G, Wagner H, Nogueira L, Wagner PD, Tang A, Dalton ND, Gu Y, Peterson KL, Breen EC. Impaired exercise capacity and skeletal muscle function in a mouse model of pulmonary inflammation. J Appl Physiol (1985) 2013; 114:1340-50. [PMID: 23449936 DOI: 10.1152/japplphysiol.00607.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Pulmonary TNFα has been linked to reduced exercise capacity in a subset of patients with moderate to severe chronic obstructive pulmonary disease (COPD). We hypothesized that prolonged, high expression of pulmonary TNFα impairs cardiac and skeletal muscle function, and both contribute to exercise limitation. Using a surfactant protein C promoter-TNFα construct, TNFα was overexpressed throughout life in mouse lungs (SP-C/TNFα+). TNFα levels in wild-type (WT) female serum and lung were two- and threefold higher than in WT male mice. In SP-C/TNFα+ mice, TNFα increased similarly in both sexes. Treadmill exercise was impaired only in male SP-C/TNFα+ mice. While increases in lung volume and airspace size induced by TNFα were comparable in both sexes, pulmonary hypertension along with lower body and muscle mass were evident only in male mice. Left ventricular (LV) function (cardiac output, stroke volume, LV maximal pressure, and LV maximal pressure dP/dt) was not altered by TNFα overexpression. Fatigue measured in isolated soleus and EDL was more rapid only in soleus of male SP-C/TNFα+ mice and accompanied by a loss of oxidative IIa fibers, citrate synthase activity, and PGC-1α mRNA and increase in atrogin-1 and MuRF1 expression also only in male mice. In situ gastrocnemius fatigue resistance, reflecting both oxygen availability and contractility, was decreased similarly in female and male SP-C/TNFα+ mice. These data indicate that male, but not female, mice overexpressing pulmonary TNFα are susceptible to exercise limitation, possibly due to muscle wasting and loss of the oxidative muscle phenotype, with protection in females possibly due to estrogen.
Collapse
Affiliation(s)
- Kechun Tang
- Department of Medicine, University of California, San Diego, La Jolla, California 92093-0623, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Transcriptional effects of E3 ligase atrogin-1/MAFbx on apoptosis, hypertrophy and inflammation in neonatal rat cardiomyocytes. PLoS One 2013; 8:e53831. [PMID: 23335977 PMCID: PMC3545877 DOI: 10.1371/journal.pone.0053831] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 12/03/2012] [Indexed: 12/24/2022] Open
Abstract
Atrogin-1/MAFbx is an ubiquitin E3 ligase that regulates myocardial structure and function through the ubiquitin-dependent protein modification. However, little is known about the effect of atrogin-1 activation on the gene expression changes in cardiomyocytes. Neonatal rat cardiomyocytes were infected with adenovirus atrogin-1 (Ad-atrogin-1) or GFP control (Ad-GFP) for 24 hours. The gene expression profiles were compared with microarray analysis. 314 genes were identified as differentially expressed by overexpression of atrogin-1, of which 222 were up-regulated and 92 were down-regulated. Atrogin-1 overexpression significantly modulated the expression of genes in 30 main functional categories, most genes clustered around the regulation of cell death, proliferation, inflammation, metabolism and cardiomyoctye structure and function. Moreover, overexpression of atrogin-1 significantly inhibited cardiomyocyte survival, hypertrophy and inflammation under basal condition or in response to lipopolysaccharide (LPS). In contrast, knockdown of atrogin-1 by siRNA had opposite effects. The mechanisms underlying these effects were associated with inhibition of MAPK (ERK1/2, JNK1/2 and p38) and NF-κB signaling pathways. In conclusion, the present microarray analysis reveals previously unappreciated atrogin-1 regulation of genes that could contribute to the effects of atrogin-1 on cardiomyocyte survival, hypertrophy and inflammation in response to endotoxin, and may provide novel insight into how atrogin-1 modulates the programming of cardiac muscle gene expression.
Collapse
|
63
|
Chen L, Cheng C, Zhang C, Yao Q, Zhao E. Ubiquitin-conjugating enzyme involved in the immune response caused by pathogens invasion. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/oji.2013.33013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
64
|
Henderson TMA, Ladewig K, Haylock DN, McLean KM, O'Connor AJ. Cryogels for biomedical applications. J Mater Chem B 2013; 1:2682-2695. [DOI: 10.1039/c3tb20280a] [Citation(s) in RCA: 193] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
65
|
Schmidt-Lauber C, Harrach S, Pap T, Fischer M, Victor M, Heitzmann M, Hansen U, Fobker M, Brand SM, Sindic A, Pavenstädt H, Edemir B, Schlatter E, Bertrand J, Ciarimboli G. Transport mechanisms and their pathology-induced regulation govern tyrosine kinase inhibitor delivery in rheumatoid arthritis. PLoS One 2012; 7:e52247. [PMID: 23284953 PMCID: PMC3527388 DOI: 10.1371/journal.pone.0052247] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 11/16/2012] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) are effective in treating malignant disorders and were lately suggested to have an impact on non-malignant diseases. However, in some inflammatory conditions like rheumatoid arthritis (RA) the in vivo effect seemed to be moderate. As most TKIs are taken up actively into cells by cell membrane transporters, this study aimed to evaluate the role of such transporters for the accumulation of the TKI Imatinib mesylates in RA synovial fibroblasts as well as their regulation under inflammatory conditions. METHODOLOGY/PRINCIPAL FINDINGS The transport and accumulation of Imatinib was investigated in transporter-transfected HEK293 cells and human RA synovial fibroblasts (hRASF). Transporter expression was quantified by qRT-PCR. In transfection experiments, hMATE1 showed the highest apparent affinity for Imatinib among all known Imatinib transporters. Experiments quantifying the Imatinib uptake in the presence of specific transporter inhibitors and after siRNA knockdown of hMATE1 indeed identified hMATE1 to mediate Imatinib transport in hRASF. The anti-proliferative effect of Imatinib on PDGF stimulated hRASF was quantified by cell counting and directly correlated with the uptake activity of hMATE1. Expression of hMATE1 was investigated by Western blot and immuno-fluorescence. Imatinib transport under disease-relevant conditions, such as an altered pH and following stimulation with different cytokines, was also investigated by HPLC. The uptake was significantly reduced by an acidic extracellular pH as well as by the cytokines TNFα, IL-1β and IL-6, which all decreased the expression of hMATE1-mRNA and protein. CONCLUSION/SIGNIFICANCE The regulation of Imatinib uptake via hMATE1 in hRASF and resulting effects on their proliferation may explain moderate in vivo effects on RA. Moreover, our results suggest that investigating transporter mediated drug processing under normal and pathological conditions is important for developing intracellular acting drugs used in inflammatory diseases.
Collapse
Affiliation(s)
- Christian Schmidt-Lauber
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Münster, Germany
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Saliha Harrach
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Münster, Germany
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Thomas Pap
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Meike Fischer
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Marion Victor
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Marianne Heitzmann
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Uwe Hansen
- Institute for Physiological Chemistry and Pathobiochemistry, University Hospital Münster, Münster, Germany
| | - Manfred Fobker
- Center of Laboratory Medicine, University Hospital Münster, Münster, Germany
| | - Stefan-Martin Brand
- Leibniz-Institute for Arteriosclerosis Research, University Hospital Münster, Münster, Germany
- Molecular Genetics of Cardiovascular Disease, Institute of Sports Medicine, University Hospital Münster, Münster, Germany
| | - Aleksandra Sindic
- Department of Physiology, Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
| | - Hermann Pavenstädt
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Bayram Edemir
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Eberhard Schlatter
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Jessica Bertrand
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Giuliano Ciarimboli
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Münster, Germany
- * E-mail:
| |
Collapse
|
66
|
The preventive effect of β-carotene on denervation-induced soleus muscle atrophy in mice. Br J Nutr 2012; 109:1349-58. [PMID: 23046823 DOI: 10.1017/s0007114512003297] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Muscle atrophy increases the production of reactive oxygen species and the expression of atrophy-related genes, which are involved in the ubiquitin-proteasome system. In the present study, we investigated the effects of β-carotene on oxidative stress (100 μM-H2O2)-induced muscle atrophy in murine C2C12 myotubes. β-Carotene (10 μM) restored the H2O2-induced decreased levels of myosin heavy chain and tropomyosin (P< 0·05, n 3) and decreased the H2O2-induced increased levels of ubiquitin conjugates. β-Carotene reduced the H2O2-induced increased expression levels of E3 ubiquitin ligases (Atrogin-1 and MuRF1) and deubiquitinating enzymes (USP14 and USP19) (P< 0·05, n 3) and attenuated the H2O2-induced nuclear localisation of FOXO3a. Furthermore, we determined the effects of β-carotene on denervation-induced muscle atrophy. Male ddY mice (8 weeks old, n 30) were divided into two groups and orally pre-administered micelle with or without β-carotene (0·5 mg once daily) for 2 weeks, followed by denervation in the right hindlimb. β-Carotene was further administered once daily until the end of the experiment. At day 3 after denervation, the ratio of soleus muscle mass in the denervated leg to that in the sham leg was significantly higher in β-carotene-administered mice than in control vehicle-administered ones (P< 0·05, n 5). In the denervated soleus muscle, β-carotene administration significantly decreased the expression levels of Atrogin-1, MuRF1, USP14 and USP19 (P< 0·05, n 5) and the levels of ubiquitin conjugates. These results indicate that β-carotene attenuates soleus muscle loss, perhaps by repressing the expressions of Atrogin-1, MuRF1, USP14 and USP19, at the early stage of soleus muscle atrophy.
Collapse
|
67
|
Interferon-γ causes cardiac myocyte atrophy via selective degradation of myosin heavy chain in a model of chronic myocarditis. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:2038-46. [PMID: 23058369 DOI: 10.1016/j.ajpath.2012.08.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 08/09/2012] [Accepted: 08/30/2012] [Indexed: 11/23/2022]
Abstract
Interferon-γ (IFN-γ), a proinflammatory cytokine, has been implicated in the pathogenesis of a number of forms of heart disease including myocarditis and congestive heart failure. In fact, overexpression of IFN-γ in mice causes dilated cardiomyopathy. However, the direct effects of IFN-γ on cardiac myocytes and the mechanism by which it causes cardiac dysfunction have not been described. Here, we present the molecular pathology of IFN-γ exposure and its effect on myofibrillar proteins in isolated neonatal rat ventricular myocytes. Treatment with IFN-γ caused cardiac myocyte atrophy attributable to a specific decrease in myosin heavy chain protein. This selective degradation of myosin heavy chain was not accompanied by a decrease in total protein synthesis or by an increase in total protein degradation. IFN-γ increased both proteasome and immunoproteasome activity in cardiac myocytes and their inhibition blocked myosin heavy chain loss and myocyte atrophy, whereas inhibition of the lysosome or autophagosome did not. Collectively, these results provide a mechanism by which IFN-γ causes cardiac pathology in the setting of chronic inflammatory diseases.
Collapse
|
68
|
Núñez-Acuña G, Aguilar-Espinoza A, Chávez-Mardones J, Gallardo-Escárate C. Ubiquitin-conjugating enzyme E2-like gene associated to pathogen response in Concholepas concholepas: SNP identification and transcription expression. FISH & SHELLFISH IMMUNOLOGY 2012; 33:1065-1068. [PMID: 22971731 DOI: 10.1016/j.fsi.2012.08.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/21/2012] [Accepted: 08/24/2012] [Indexed: 06/01/2023]
Abstract
Ubiquitin-conjugated E2 enzyme (UBE2) is one of the main components of the proteasome degradation cascade. Previous studies have shown an increase of expression levels in individuals challenged to some pathogen organism such as virus and bacteria. The study was to characterize the immune response of UBE2 gene in the gastropod Concholepas concholepas through expression analysis and single nucleotide polymorphisms (SNP) discovery. Hence, UBE2 was identified from a cDNA library by 454 pyrosequencing, while SNP identification and validation were performed using De novo assembly and high resolution melting analysis. Challenge trials with Vibrio anguillarum was carried out to evaluate the relative transcript abundance of UBE2 gene from two to thirty-three hours post-treatment. The results showed a partial UBE2 sequence of 889 base pair (bp) with a partial coding region of 291 bp. SNP variation (A/C) was observed at the 546th position. Individuals challenged by V. anguillarum showed an overexpression of the UBE2 gene, the expression being significantly higher in homozygous individuals (AA) than (CC) or heterozygous individuals (A/C). This study contributes useful information relating to the UBE2 gene and its association with innate immune response in marine invertebrates.
Collapse
Affiliation(s)
- Gustavo Núñez-Acuña
- Laboratorio de Biotecnología y Genómica Acuícola, Departamento de Oceanografía, Facultad de Ciencias Naturales y Oceanográficas, Universidad de Concepción, P.O. Box 160-C, Concepción, Chile
| | | | | | | |
Collapse
|
69
|
Saracino GAA, Cigognini D, Silva D, Caprini A, Gelain F. Nanomaterials design and tests for neural tissue engineering. Chem Soc Rev 2012; 42:225-62. [PMID: 22990473 DOI: 10.1039/c2cs35065c] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nanostructured scaffolds recently showed great promise in tissue engineering: nanomaterials can be tailored at the molecular level and scaffold morphology may more closely resemble features of extracellular matrix components in terms of porosity, framing and biofunctionalities. As a consequence, both biomechanical properties of scaffold microenvironments and biomaterial-protein interactions can be tuned, allowing for improved transplanted cell engraftment and better controlled diffusion of drugs. Easier said than done, a nanotech-based regenerative approach encompasses different fields of know-how, ranging from in silico simulations, nanomaterial synthesis and characterization at the nano-, micro- and mesoscales to random library screening methods (e.g. phage display), in vitro cellular-based experiments and validation in animal models of the target injury. All of these steps of the "assembly line" of nanostructured scaffolds are tightly interconnected both in their standard analysis techniques and in their most recent breakthroughs: indeed their efforts have to jointly provide the deepest possible analyses of the diverse facets of the challenging field of neural tissue engineering. The purpose of this review is therefore to provide a critical overview of the recent advances in and drawbacks and potential of each mentioned field, contributing to the realization of effective nanotech-based therapies for the regeneration of peripheral nerve transections, spinal cord injuries and brain traumatic injuries. Far from being the ultimate overview of such a number of topics, the reader will acknowledge the intrinsic complexity of the goal of nanotech tissue engineering for a conscious approach to the development of a regenerative therapy and, by deciphering the thread connecting all steps of the research, will gain the necessary view of its tremendous potential if each piece of stone is correctly placed to work synergically in this impressive mosaic.
Collapse
Affiliation(s)
- Gloria A A Saracino
- Center for Nanomedicine and Tissue Engineering, A.O. Ospedale Niguarda Cà Granda, Milan, 20162, Italy
| | | | | | | | | |
Collapse
|
70
|
Florin V, Cottencin AC, Delaporte E, Staumont-Sallé D. Body weight increment in patients treated with infliximab for plaque psoriasis. J Eur Acad Dermatol Venereol 2012; 27:e186-90. [PMID: 22621415 DOI: 10.1111/j.1468-3083.2012.04571.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Psoriasis is frequently associated with overweight and obesity. Anti-TNF-α therapies are effective in the treatment of psoriasis. TNF-α is highly involved in body weight regulation. OBJECTIVE Our objective was to evaluate the increase in weight throughout the treatment with infliximab and the association of weight gain with the body mass index (BMI). METHODS Thirty-five patients affected with severe plaque psoriasis receiving infliximab were included. A control group consisted of 16 patients affected with severe plaque psoriasis and treated with cyclosporine, methotrexate, or acitretin. Assessment of PASI score, body weight and BMI were performed at a 1 and 3-year follow-up. RESULTS We observed a body weight increment of 2.5 ± 4.4 kg (mean ± SD) (i.e. + 3.6% of baseline) and 0.1 ± 5 kg (i.e. + 1.2%) in patients treated with infliximab and the control group, respectively (P = 0.046), after 1 year of treatment. After 3 years of infliximab administration, weight gain was 4.8 ± 5 kg (n = 16) (i.e. + 6%) (P = 0.005). Moreover, as classified by BMI, normal weight patients experienced a 4 ± 3.7 kg weight gain (i.e. + 6%) whereas overweight and obese patients had gained 1.3 ± 4.8 kg (i.e. + 1.2%) (P = 0.039) after 1 year of anti-TNF-α therapy. Percentual changes in body weight were larger in normal weight patients at baseline than in overweight/obese counterparts (P = 0.0149). CONCLUSION All patients, including normal weight patients, should receive a dietary intervention.
Collapse
Affiliation(s)
- V Florin
- Department of Dermatology, Claude-Huriez Hospital, University Hospital of Lille, Lille Cedex, France
| | | | | | | |
Collapse
|
71
|
Bhatnagar S, Mittal A, Gupta SK, Kumar A. TWEAK causes myotube atrophy through coordinated activation of ubiquitin-proteasome system, autophagy, and caspases. J Cell Physiol 2012; 227:1042-51. [PMID: 21567392 DOI: 10.1002/jcp.22821] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Proinflammatory cytokine TWEAK has now emerged as a key mediator of skeletal muscle-wasting in many catabolic conditions. However, the mechanisms by which TWEAK induces muscle proteolysis remain poorly understood. Here, we have investigated the role of ubiquitin-proteasome system, autophagy, and caspases in TWEAK-induced muscle wasting. Addition of TWEAK to C2C12 myotubes stimulated the ubiquitination of myosin heavy chain (MyHC) and augmented the expression of E3 ubiquitin ligase MuRF1. Pretreatment of myotubes with proteasome inhibitors MG132 or lactacystin or knockdown of MuRF1 by RNAi blocked the TWEAK-induced degradation of MyHC and myotube atrophy. TWEAK increased the expression of several autophagy-related molecules. Moreover, the inhibitors of autophagy improved the levels of MyHC in TWEAK-treated myotubes. TWEAK also increased activity of caspases in C2C12 myotubes. Pan-caspase or caspase 3 inhibitory peptide inhibited the TWEAK-induced loss of MyHC and myotube diameter. Our study demonstrates that nuclear factor-kappa B (NF-κB) transcription factor is essential for TWEAK-induced expression of MuRF1 and Beclin1. Furthermore, our results suggest that caspases contribute, at least in part, to the activation of NF-κB in response to TWEAK treatment. Collectively, the present study provides novel insight into the mechanisms of action of TWEAK in skeletal muscle.
Collapse
Affiliation(s)
- Shephali Bhatnagar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | | | | | | |
Collapse
|
72
|
Bashur CA, Venkataraman L, Ramamurthi A. Tissue engineering and regenerative strategies to replicate biocomplexity of vascular elastic matrix assembly. TISSUE ENGINEERING PART B-REVIEWS 2012; 18:203-17. [PMID: 22224468 DOI: 10.1089/ten.teb.2011.0521] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cardiovascular tissues exhibit architecturally complex extracellular matrices, of which the elastic matrix forms a major component. The elastic matrix critically maintains native structural configurations of vascular tissues, determines their ability to recoil after stretch, and regulates cell signaling pathways involved in morphogenesis, injury response, and inflammation via biomechanical transduction. The ability to tissue engineer vascular replacements that incorporate elastic matrix superstructures unique to cardiac and vascular tissues is thus important to maintaining vascular homeostasis. However, the vascular elastic matrix is particularly difficult to tissue engineer due to the inherently poor ability of adult vascular cells to synthesize elastin precursors and organize them into mature structures in a manner that replicates the biocomplexity of elastic matrix assembly during development. This review discusses current tissue engineering materials (e.g., growth factors and scaffolds) and methods (e.g., dynamic stretch and contact guidance) used to promote cellular synthesis and assembly of elastic matrix superstructures, and the limitations of these approaches when applied to smooth muscle cells, the primary elastin-generating cell type in vascular tissues. The potential application of these methods for in situ regeneration of disrupted elastic matrix at sites of proteolytic vascular disease (e.g., abdominal aortic aneurysms) is also discussed. Finally, the review describes the potential utility of alternative cell types to elastic tissue engineering and regenerative matrix repair. Future progress in the field is contingent on developing a thorough understanding of developmental elastogenesis and then mimicking the spatiotemporal changes in the cellular microenvironment that occur during that phase. This will enable us to tissue engineer clinically applicable elastic vascular tissue replacements and to develop elastogenic therapies to restore homeostasis in de-elasticized vessels.
Collapse
Affiliation(s)
- Chris A Bashur
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | |
Collapse
|
73
|
Sun YS, Peng SW, Lin KH, Cheng JY. Electrotaxis of lung cancer cells in ordered three-dimensional scaffolds. BIOMICROFLUIDICS 2012; 6:14102-1410214. [PMID: 22288000 PMCID: PMC3267495 DOI: 10.1063/1.3671399] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 11/30/2011] [Indexed: 05/12/2023]
Abstract
In this paper, we report a new method to incorporate 3D scaffold with electrotaxis measurement in the microfluidic device. The electrotactic response of lung cancer cells in the 3D foam scaffolds which resemble the in vivo pulmonary alveoli may give more insight on cellular behaviors in vivo. The 3D scaffold consists of ordered arrays of uniform spherical pores in gelatin. We found that cell morphology in the 3D scaffold was different from that in 2D substrate. Next, we applied a direct current electric field (EF) of 338 mV/mm through the scaffold for the study of cells' migration within. We measured the migration directedness and speed of different lung cancer cell lines, CL1-0, CL1-5, and A549, and compared with those examined in 2D gelatin-coated and bare substrates. The migration direction is the same for all conditions but there are clear differences in cell morphology, directedness, and migration speed under EF. Our results demonstrate cell migration under EF is different in 2D and 3D environments and possibly due to different cell morphology and/or substrate stiffness.
Collapse
|
74
|
Renzo LDI, Saraceno R, Schipani C, Rizzo M, Bianchi A, Noce A, Esposito M, Tiberti S, Chimenti S, DE Lorenzo A. Prospective assessment of body weight and body composition changes in patients with psoriasis receiving anti-TNF-α treatment. Dermatol Ther 2012; 24:446-51. [PMID: 21910803 DOI: 10.1111/j.1529-8019.2011.01439.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Tumor necrosis factor (TNF)-α is a pro-inflammatory cytokine associated with psoriasis pathogenesis. Anti-TNF-α therapies are effective in psoriasis. A significant weight gain has been reported in patients treated with anti-TNF-α agents. The aim of the present study was to evaluate the body composition changes in psoriatic patients receiving anti-TNF-α therapies according with disease phenotype. Forty patients affected with psoriasis were followed up for 24 weeks and divided into two groups: psoriasis vulgaris (PsO) and psoriatic arthritis (PsA). Anthropometric, blood biochemical, body composition parameters, resting metabolic rate, and disease activity indexes were measured at baseline and at week 24. After 24 weeks of anti-TNF-α administration, the disease activity indexes and concentration of inflammatory markers were significantly decreased. Seventy-five percent of PsO and 60% of PsA patients had an increase in body weight. Weight changes correlated with fat mass gain in the PsO group, and with fat and lean mass gain in the PsA group. In the present study, we demonstrated that a blockage of TNF-α bioactivity is related with fat and lean mass gain in both PsO and PsA subjects. The anti-TNF-α therapies could play a key role in the cross talk between adipose tissue and skeletal muscle, mediated by the reduction of TNF-α and interleukin-6 production.
Collapse
Affiliation(s)
- Laura D I Renzo
- Division of Human Nutrition, Department of Neuroscience, University of Rome Tor Vergata, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Venkataraman L, Ramamurthi A. Induced elastic matrix deposition within three-dimensional collagen scaffolds. Tissue Eng Part A 2011; 17:2879-89. [PMID: 21702719 DOI: 10.1089/ten.tea.2010.0749] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The structural stability of a cyclically distending elastic artery and the healthy functioning of vascular smooth muscle cells (SMCs) within are maintained by the presence of an intact elastic matrix and its principal protein, elastin. The accelerated degradation of the elastic matrix, which occurs in several vascular diseases, coupled with the poor ability of adult SMCs to regenerate lost elastin, can therefore adversely impact vascular homeostasis. Similarly, efforts to tissue engineer elastic matrix structures are constrained by our inability to induce adult cells to synthesize tropoelastin precursors and to crosslink them into architectural mimics of native elastic matrices, especially within engineered constructs where SMCs/fibroblasts primarily deposit collagen in abundance. In this study, we have shown that transforming growth factor-beta1 (TGF-β1) and hyaluronan oligomers (HA-o) synergistically enhance elastic matrix deposition by adult rat aortic SMCs (RASMCs) seeded within nonelastogenic, statically loaded three-dimensional gels, composed of nonelastogenic type-I collagen. While there was no substantial increase in production of tropoelastin within experimental cases compared to the nonadditive control cultures over 3 weeks, we observed significant increases in matrix elastin deposition; soluble matrix elastin in constructs that received the lowest doses of TGF-β1 with respective doses of HA-o, and insoluble matrix at the highest doses that corresponded with elevated lysyl-oxidase protein quantities. However, despite elastogenic induction, overall matrix yields remained poor in all experimental cases. At all provided doses, the factors reduced the production of matrix metalloproteinases (MMP)-9, especially the active enzyme, though MMP-2 levels were lowered only in constructs cultured with the higher doses of TGF-β1. Immuno-fluorescence showed elastic fibers within the collagen constructs to be discontinuous, except at the edges of the constructs. Von Kossa staining revealed no calcific deposits in any of the cases. This study confirms the benefits of utilizing TGF-β1 and HA-o in inducing matrix elastin synthesis by adult RASMCs over nonadditive controls, within a collagenous environment, that is not inherently conducive to elastogenesis.
Collapse
|
76
|
Caron AZ, Haroun S, Leblanc E, Trensz F, Guindi C, Amrani A, Grenier G. The proteasome inhibitor MG132 reduces immobilization-induced skeletal muscle atrophy in mice. BMC Musculoskelet Disord 2011; 12:185. [PMID: 21843349 PMCID: PMC3173404 DOI: 10.1186/1471-2474-12-185] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 08/15/2011] [Indexed: 11/10/2022] Open
Abstract
Background Skeletal muscle atrophy is a serious concern for the rehabilitation of patients afflicted by prolonged limb restriction. This debilitating condition is associated with a marked activation of NFκB activity. The ubiquitin-proteasome pathway degrades the NFκB inhibitor IκBα, enabling NFκB to translocate to the nucleus and bind to the target genes that promote muscle atrophy. Although several studies showed that proteasome inhibitors are efficient to reduce atrophy, no studies have demonstrated the ability of these inhibitors to preserve muscle function under catabolic condition. Methods We recently developed a new hindlimb immobilization procedure that induces significant skeletal muscle atrophy and used it to show that an inflammatory process characterized by the up-regulation of TNFα, a known activator of the canonical NFκB pathway, is associated with the atrophy. Here, we used this model to investigate the effect of in vivo proteasome inhibition on the muscle integrity by histological approach. TNFα, IL-1, IL-6, MuRF-1 and Atrogin/MAFbx mRNA level were determined by qPCR. Also, a functional measurement of locomotors activity was performed to determine if the treatment can shorten the rehabilitation period following immobilization. Results In the present study, we showed that the proteasome inhibitor MG132 significantly inhibited IκBα degradation thus preventing NFκB activation in vitro. MG132 preserved muscle and myofiber cross-sectional area by downregulating the muscle-specific ubiquitin ligases atrogin-1/MAFbx and MuRF-1 mRNA in vivo. This effect resulted in a diminished rehabilitation period. Conclusion These finding demonstrate that proteasome inhibitors show potential for the development of pharmacological therapies to prevent muscle atrophy and thus favor muscle rehabilitation.
Collapse
Affiliation(s)
- Annabelle Z Caron
- Centre de Recherche Clinique Étienne-Lebel, 3001-12th Avenue North, Sherbrooke, QC J1H5N4, Canada
| | | | | | | | | | | | | |
Collapse
|
77
|
Wing SS, Lecker SH, Jagoe RT. Proteolysis in illness-associated skeletal muscle atrophy: from pathways to networks. Crit Rev Clin Lab Sci 2011; 48:49-70. [PMID: 21699435 DOI: 10.3109/10408363.2011.586171] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Improvements in health in the past decades have resulted in increased numbers of the elderly in both developed and developing regions of the world. Advances in therapy have also increased the prevalence of patients with chronic and degenerative diseases. Muscle wasting, a feature of most chronic diseases, is prominent in the elderly and contributes to both morbidity and mortality. A major research goal has been to identify the proteolytic system(s) that is responsible for the degradation of proteins that occurs in muscle atrophy. Findings over the past 20 years have clearly confirmed an important role of the ubiquitin proteasome system in mediating muscle proteolysis, particularly that of myofibrillar proteins. However, recent observations have provided evidence that autophagy, calpains and caspases also contribute to the turnover of muscle proteins in catabolic states, and furthermore, that these diverse proteolytic systems interact with each other at various levels. Importantly, a number of intracellular signaling pathways such as the IGF1/AKT, myostatin/Smad, PGC1, cytokine/NFκB, and AMPK pathways are now known to interact and can regulate some of these proteolytic systems in a coordinated manner. A number of loss of function studies have identified promising therapeutic approaches to the prevention and treatment of wasting. However, additional biomarkers and other approaches to improve early identification of patients who would benefit from such treatment need to be developed. The current data suggests a network of interacting proteolytic and signaling pathways in muscle. Future studies are needed to improve understanding of the nature and control of these interactions and how they work to preserve muscle function under various states of growth and atrophy.
Collapse
Affiliation(s)
- Simon S Wing
- Departments of Medicine, McGill University and McGill University Health Centre Research Institute, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
78
|
Li JJ, Zhang TP, Meng Y, Du J, Li HH. Stability of F-box protein atrogin-1 is regulated by p38 mitogen-activated protein kinase pathway in cardiac H9c2 cells. Cell Physiol Biochem 2011; 27:463-70. [PMID: 21691063 DOI: 10.1159/000329967] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2011] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Atrogin-1/MAFbx is a major atrophy-related E3 ubiquitin ligase that functions as a negative regulator of cardiac hypertrophy. The mRNA expression of atrogin-1 is induced by oxidative stress via p38 mitogen-activated protein kinase (p38 MAPK). However, the molecular mechanisms that regulate the stability of atrogin-1 protein remain unclear. METHODS 293T and cardiac H9c2 cells were transfected with plasmids as indicated. The in vivo and in vitro ubiquitination assay and pulse-chase analysis were performed to detect the ubiquitination and stability of atrogin-1. The protein levels were measured by Western blot analysis. RESULTS We found that atrogin-1 underwent ubiquitin-mediated degradation by proteasome. The F-box motif of atrogin-1 and Skp1-Cul1-Roc1-F-box (SCF) complex are required for ubiquitination and degradation of atrogin-1. Furthermore, p38 MAPK signaling plays critical roles in regulating the ubiquitination and degradation of atrogin-1 as well as serum starvation-induced expression of atrogin-1 and reduction of H9c2 cell size. CONCLUSION These findings may define a new mechanism for regulating the stability of atrogin-1 partially by p38 MAPK signaling.
Collapse
Affiliation(s)
- Jun-Jie Li
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, No.10 Xitoutiao, You An Men, Beijing, China
| | | | | | | | | |
Collapse
|
79
|
Lin S, Sandig M, Mequanint K. Three-dimensional topography of synthetic scaffolds induces elastin synthesis by human coronary artery smooth muscle cells. Tissue Eng Part A 2011; 17:1561-71. [PMID: 21284555 DOI: 10.1089/ten.tea.2010.0593] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Due to the important structural and signaling roles of elastin in vascular stability, engineered human vascular tissues must incorporate elastin. However, despite considerable progress toward engineering of elastin-containing vascular tissues from animal cells, currently engineered vascular tissues using human cells largely lack elastin. In this study, we evaluated the effect of scaffold topography (two dimensional [2D] vs. three dimensional [3D]) on elastogenesis in adult human coronary artery smooth muscle cells (HCASMCs). We report that elastin gene expression by HCASMCs was increased by twofold after 4 days of culture in porous 3D polyurethane scaffolds. Transforming growth factor β1 (TGF-β1) further increased elastin gene expression in 3D cultures but not in 2D cultures. To evaluate if gene expression is translated into elastin synthesis, both 2D and 3D cultures were analyzed using Western blots. We show that only HCASMCs in 3D scaffolds produced elastin, suggesting that scaffold geometry itself is an important cue for elastogenesis. Moreover, TGF-β1 enhanced elastin synthesis in 3D, but had no effect on cells grown on 2D surfaces. TGF-β1, known to induce vascular smooth muscle cells (VSMC) differentiation, upregulated contractile VSMC marker proteins smooth muscle-α-actin and calponin in cells on 2D surfaces. Interestingly, in 3D scaffolds, TGF-β1 failed to upregulate these differentiation marker proteins for at least 7 days, but did so in cells cultured for 14 days, whereas elastin synthesis was not affected. To our knowledge this study is the first to successfully demonstrate that adult human VSMC can produce elastin when seeded on 3D scaffolds and to directly compare the effect of scaffold topography on elastin synthesis. Knowledge about the conditions required to regulate the phenotype of human VSMCs is paramount to engineer elastin-containing autologous human vascular substitutes.
Collapse
Affiliation(s)
- Shigang Lin
- Department of Chemical and Biochemical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario, Canada
| | | | | |
Collapse
|
80
|
Nomura W, Narumi T, Ohashi N, Serizawa Y, Lewin NE, Blumberg PM, Furuta T, Tamamura H. Synthetic caged DAG-lactones for photochemically controlled activation of protein kinase C. Chembiochem 2011; 12:535-9. [PMID: 22238145 PMCID: PMC8259317 DOI: 10.1002/cbic.201000670] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Indexed: 11/07/2022]
Abstract
Switching on kinases: Synthetic caged DAG-lactones have been developed and showed decreases of two orders of magnitude, relative to the corresponding parent compounds, in their binding affinities towards PKC. The caged compounds had no effect on the translocation of PKC until after photoactivation. This approach is a potentially powerful tool for probing the PKC signaling cascade.
Collapse
Affiliation(s)
- Wataru Nomura
- Department of Medicinal Chemistry, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kandasurugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Beamish JA, He P, Kottke-Marchant K, Marchant RE. Molecular regulation of contractile smooth muscle cell phenotype: implications for vascular tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2011; 16:467-91. [PMID: 20334504 DOI: 10.1089/ten.teb.2009.0630] [Citation(s) in RCA: 297] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The molecular regulation of smooth muscle cell (SMC) behavior is reviewed, with particular emphasis on stimuli that promote the contractile phenotype. SMCs can shift reversibly along a continuum from a quiescent, contractile phenotype to a synthetic phenotype, which is characterized by proliferation and extracellular matrix (ECM) synthesis. This phenotypic plasticity can be harnessed for tissue engineering. Cultured synthetic SMCs have been used to engineer smooth muscle tissues with organized ECM and cell populations. However, returning SMCs to a contractile phenotype remains a key challenge. This review will integrate recent work on how soluble signaling factors, ECM, mechanical stimulation, and other cells contribute to the regulation of contractile SMC phenotype. The signal transduction pathways and mechanisms of gene expression induced by these stimuli are beginning to be elucidated and provide useful information for the quantitative analysis of SMC phenotype in engineered tissues. Progress in the development of tissue-engineered scaffold systems that implement biochemical, mechanical, or novel polymer fabrication approaches to promote contractile phenotype will also be reviewed. The application of an improved molecular understanding of SMC biology will facilitate the design of more potent cell-instructive scaffold systems to regulate SMC behavior.
Collapse
Affiliation(s)
- Jeffrey A Beamish
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106-7207, USA
| | | | | | | |
Collapse
|
82
|
Peterson JM, Bakkar N, Guttridge DC. NF-κB Signaling in Skeletal Muscle Health and Disease. Curr Top Dev Biol 2011; 96:85-119. [DOI: 10.1016/b978-0-12-385940-2.00004-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
83
|
|
84
|
Bhatnagar S, Panguluri SK, Gupta SK, Dahiya S, Lundy RF, Kumar A. Tumor necrosis factor-α regulates distinct molecular pathways and gene networks in cultured skeletal muscle cells. PLoS One 2010; 5:e13262. [PMID: 20967264 PMCID: PMC2953497 DOI: 10.1371/journal.pone.0013262] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 09/14/2010] [Indexed: 11/18/2022] Open
Abstract
Background Skeletal muscle wasting is a debilitating consequence of large number of disease states and conditions. Tumor necrosis factor-α (TNF-α) is one of the most important muscle-wasting cytokine, elevated levels of which cause significant muscular abnormalities. However, the underpinning molecular mechanisms by which TNF-α causes skeletal muscle wasting are less well-understood. Methodology/Principal Findings We have used microarray, quantitative real-time PCR (QRT-PCR), Western blot, and bioinformatics tools to study the effects of TNF-α on various molecular pathways and gene networks in C2C12 cells (a mouse myoblastic cell line). Microarray analyses of C2C12 myotubes treated with TNF-α (10 ng/ml) for 18h showed differential expression of a number of genes involved in distinct molecular pathways. The genes involved in nuclear factor-kappa B (NF-kappaB) signaling, 26s proteasome pathway, Notch1 signaling, and chemokine networks are the most important ones affected by TNF-α. The expression of some of the genes in microarray dataset showed good correlation in independent QRT-PCR and Western blot assays. Analysis of TNF-treated myotubes showed that TNF-α augments the activity of both canonical and alternative NF-κB signaling pathways in myotubes. Bioinformatics analyses of microarray dataset revealed that TNF-α affects the activity of several important pathways including those involved in oxidative stress, hepatic fibrosis, mitochondrial dysfunction, cholesterol biosynthesis, and TGF-β signaling. Furthermore, TNF-α was found to affect the gene networks related to drug metabolism, cell cycle, cancer, neurological disease, organismal injury, and abnormalities in myotubes. Conclusions TNF-α regulates the expression of multiple genes involved in various toxic pathways which may be responsible for TNF-induced muscle loss in catabolic conditions. Our study suggests that TNF-α activates both canonical and alternative NF-κB signaling pathways in a time-dependent manner in skeletal muscle cells. The study provides novel insight into the mechanisms of action of TNF-α in skeletal muscle cells.
Collapse
Affiliation(s)
- Shephali Bhatnagar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Siva K. Panguluri
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Sanjay K. Gupta
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Saurabh Dahiya
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Robert F. Lundy
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
85
|
Abstract
Both cachexia and cardiovascular disease are strongly associated with rheumatoid arthritis (RA) and linked to the chronic inflammatory process. Typically, rheumatoid cachexia occurs in individuals with normal or increased BMI (reduced muscle mass and increased fat mass). Classic cachexia (reduced muscle mass and reduced fat mass) is rare in RA but is associated with high inflammatory activity and aggressive joint destruction in patients with a poor cardiovascular prognosis. Conversely, obesity is linked to hypertension and dyslipidemia but, paradoxically, lower RA disease activity and less cardiovascular disease-related mortality. Rheumatoid cachexia might represent the 'worst of both worlds' with respect to cardiovascular outcome, but until diagnostic criteria for this condition are agreed upon, its effect on cardiovascular disease risk remains controversial.
Collapse
|
86
|
Bonetto A, Penna F, Minero VG, Reffo P, Costamagna D, Bonelli G, Baccino FM, Costelli P. Glutamine prevents myostatin hyperexpression and protein hypercatabolism induced in C2C12 myotubes by tumor necrosis factor-α. Amino Acids 2010; 40:585-94. [DOI: 10.1007/s00726-010-0683-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 06/29/2010] [Indexed: 01/13/2023]
|
87
|
Macqueen DJ, Kristjánsson BK, Johnston IA. Salmonid genomes have a remarkably expanded akirin family, coexpressed with genes from conserved pathways governing skeletal muscle growth and catabolism. Physiol Genomics 2010; 42:134-48. [PMID: 20388840 PMCID: PMC2888561 DOI: 10.1152/physiolgenomics.00045.2010] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 04/09/2010] [Indexed: 12/17/2022] Open
Abstract
Metazoan akirin genes regulate innate immunity, myogenesis, and carcinogenesis. Invertebrates typically have one family member, while most tetrapod and teleost vertebrates have one to three. We demonstrate an expanded repertoire of eight family members in genomes of four salmonid fishes, owing to paralog preservation after three tetraploidization events. Retention of paralogs secondarily lost in other teleosts may be related to functional diversification and posttranslational regulation. We hypothesized that salmonid akirins would be transcriptionally regulated in fast-twitch skeletal muscle during activation of conserved pathways governing catabolism and growth. The in vivo nutritional state of Arctic charr (Salvelinus alpinus L.) was experimentally manipulated, and transcript levels for akirin family members and 26 other genes were measured by quantitative real-time PCR (qPCR), allowing the establishment of a similarity network of expression profiles. In fasted muscle, a class of akirins was upregulated, with one family member showing high coexpression with catabolic genes coding the NF-kappaB p65 subunit, E2 ubiquitin-conjugating enzymes, E3 ubiquitin ligases, and IGF-I receptors. Another class of akirin was upregulated with subsequent feeding, coexpressed with 14-3-3 protein genes. There was no similarity between expression profiles of akirins with IGF hormones or binding protein genes. The level of phylogenetic relatedness of akirin family members was not a strong predictor of transcriptional responses to nutritional state, or differences in transcript abundance levels, indicating a complex pattern of regulatory evolution. The salmonid akirins epitomize the complexity linking the genome to physiological phenotypes of vertebrates with a history of tetraploidization.
Collapse
Affiliation(s)
- Daniel J Macqueen
- Scottish Ocean Institute, School of Biology, University of St Andrews, St Andrews, United Kingdom.
| | | | | |
Collapse
|
88
|
C2 and C2C12 murine skeletal myoblast models of atrophic and hypertrophic potential: Relevance to disease and ageing? J Cell Physiol 2010; 225:240-50. [DOI: 10.1002/jcp.22252] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
89
|
Rhoads MG, Kandarian SC, Pacelli F, Doglietto GB, Bossola M. Expression of NF-kappaB and IkappaB proteins in skeletal muscle of gastric cancer patients. Eur J Cancer 2010; 46:191-7. [PMID: 19857958 DOI: 10.1016/j.ejca.2009.10.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 09/26/2009] [Accepted: 02/10/2009] [Indexed: 11/17/2022]
Abstract
The mechanisms eliciting cancer cachexia are not well understood. Wasting of skeletal muscle is problematic because it is responsible for the clinical deterioration in cancer patients and for the ability to tolerate cancer treatment. Studies done on animals suggest that nuclear factor of kappa B (NF-kappaB) signalling is important in the progression of muscle wasting due to several types of tumours. However, there are no published studies in humans on the role of NF-kappaB in cancer cachexia. In this project, we studied the rectus abdominis muscle in patients with gastric tumours (n=14) and in age-matched control subjects (n=10) for markers of NF-kappaB activation. Nuclear levels of p65, p50 and Bcl-3 were the same in both groups of subjects. However, phospho-p65 was elevated by 25% in the muscles of cancer patients. In addition, expression of the inhibitor of kappa B alpha (IkappaBalpha) was decreased by 25% in cancer patients. Decreased expression of IkappaBalpha reflects its degradation by one of the IkappaBalpha kinases and is a marker of NF-kappaB activation. Interestingly, there was no correlation between the stage of cancer and the extent of IkappaBalpha decrease, nor was there a correlation between the degree of cachexia and decreased IkappaBalpha levels. This suggests that the activation of NF-kappaB is an early and sustained event in gastric cancer. The work implicates the NF-kappaB signalling in the initiation and progression of cancer cachexia in humans and demonstrates the need for additional study of this pathway; it also recommends NF-kappaB signalling as a therapeutic target for the amelioration of cachexia as has been suggested from studies done on rodents.
Collapse
Affiliation(s)
- Mary G Rhoads
- Department of Health Sciences, Boston University, Boston, MA, USA
| | | | | | | | | |
Collapse
|
90
|
Cassano M, Quattrocelli M, Crippa S, Perini I, Ronzoni F, Sampaolesi M. Cellular mechanisms and local progenitor activation to regulate skeletal muscle mass. J Muscle Res Cell Motil 2010; 30:243-53. [PMID: 20195710 DOI: 10.1007/s10974-010-9204-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 02/05/2010] [Indexed: 12/21/2022]
Abstract
Skeletal muscle hypertrophy is a result of increased load, such as functional and stretch-overload. Activation of satellite cells and proliferation, differentiation and fusion are required for hypertrophy of overloaded skeletal muscles. On the contrary, a dramatic loss of skeletal muscle mass determines atrophy settings. The epigenetic changes involved in gene regulation at DNA and chromatin level are critical for the opposing phenomena, muscle growth and atrophy. Physiological properties of skeletal muscle tissue play a fundamental role in health and disease since it is the most abundant tissue in mammals. In fact, protein synthesis and degradation are finely modulated to maintain an appropriate muscle mass. When the molecular signaling is altered muscle wasting and weakness occurred, and this happened in most common inherited and acquired disorders such as muscular dystrophies, cachexia, and age-related wasting. To date, there is no accepted treatment to improve muscle size and strength, and these conditions pose a considerable anxiety to patients as well as to public health. Several molecules, including Magic-F1, myostatin inhibitor, IGF, glucocorticoids and microRNAs are currently investigated to interfere positively in the blueprint of skeletal muscle growth and regeneration.
Collapse
Affiliation(s)
- Marco Cassano
- Translational Cardiomyology, SCIL Katholieke Universiteit Leuven, Herestraat 49 bus 814, Leuven 3000, Belgium
| | | | | | | | | | | |
Collapse
|
91
|
Dodd SL, Gagnon BJ, Senf SM, Hain BA, Judge AR. Ros-mediated activation of NF-kappaB and Foxo during muscle disuse. Muscle Nerve 2010; 41:110-3. [PMID: 19813194 DOI: 10.1002/mus.21526] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We examined reactive oxygen species as upstream activators of nuclear factor kappaB; (NF-kappaB) and forkhead box O (Foxo) in skeletal muscle during disuse atrophy. Catalase, an enzyme that degrades H2O2, was overexpressed in soleus muscles via plasmid injection prior to 7 days of hindlimb immobilization. The increased catalase activity abolished immobilization-induced transactivation of both NF-kappaB and Foxo and attenuated the loss of muscle mass. Thus, H2O2 may be an important initiator of these signaling pathways that lead to muscle atrophy.
Collapse
Affiliation(s)
- Stephen L Dodd
- Department of Applied Physiology and Kinesiology, University of Florida, P.O. Box 118205, Gainesville, Florida 32611, USA.
| | | | | | | | | |
Collapse
|
92
|
Smith IJ, Alamdari N, O'Neal P, Gonnella P, Aversa Z, Hasselgren PO. Sepsis increases the expression and activity of the transcription factor Forkhead Box O 1 (FOXO1) in skeletal muscle by a glucocorticoid-dependent mechanism. Int J Biochem Cell Biol 2010; 42:701-11. [PMID: 20079455 DOI: 10.1016/j.biocel.2010.01.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 12/18/2009] [Accepted: 01/06/2010] [Indexed: 12/18/2022]
Abstract
Sepsis-induced muscle wasting has severe clinical consequences, including muscle weakness, need for prolonged ventilatory support and stay in the intensive care unit, and delayed ambulation with risk for pulmonary and thromboembolic complications. Understanding molecular mechanisms regulating loss of muscle mass in septic patients therefore has significant clinical implications. Forkhead Box O (FOXO) transcription factors have been implicated in muscle wasting, partly reflecting upregulation of the ubiquitin ligases atrogin-1 and MuRF1. The influence of sepsis on FOXO transcription factors in skeletal muscle is poorly understood. We tested the hypothesis that sepsis upregulates expression and activity of FOXO transcription factors in skeletal muscle by a glucocorticoid-dependent mechanism. Sepsis in rats increased muscle FOXO1 and 3a mRNA and protein levels but did not influence FOXO4 expression. Nuclear FOXO1 levels and DNA binding activity were increased in septic muscle whereas FOXO3a nuclear levels were not increased during sepsis. Sepsis-induced expression of FOXO1 was reduced by the glucocorticoid receptor antagonist RU38486 and treatment of rats with dexamethasone increased FOXO1 mRNA levels suggesting that the expression of FOXO1 is regulated by glucocorticoids. Reducing FOXO1, but not FOXO3a, expression by siRNA in cultured L6 myotubes inhibited dexamethasone-induced atrogin-1 and MuRF1 expression, further supporting a role of FOXO1 in glucocorticoid-regulated muscle wasting. Results suggest that sepsis increases FOXO1 expression and activity in skeletal muscle by a glucocorticoid-dependent mechanism and that glucocorticoid-dependent upregulation of atrogin-1 and MuRF1 in skeletal muscle is regulated by FOXO1. The study is significant because it provides novel information about molecular mechanisms involved in sepsis-induced muscle wasting.
Collapse
Affiliation(s)
- Ira J Smith
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue ST919, Boston, MA, United States
| | | | | | | | | | | |
Collapse
|
93
|
Kim HC, Lee GD, Hwang YS. Skeletal Muscle Dysfunction in Patients with Chronic Obstructive Pulmonary Disease. Tuberc Respir Dis (Seoul) 2010. [DOI: 10.4046/trd.2010.68.3.125] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Ho Cheol Kim
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
- Gyeongsang Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Gi Dong Lee
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Young Sil Hwang
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
| |
Collapse
|
94
|
McNicol FJ, Hoyland JA, Cooper RG, Carlson GL. Skeletal muscle contractile properties and proinflammatory cytokine gene expression in human endotoxaemia. Br J Surg 2009; 97:434-42. [DOI: 10.1002/bjs.6868] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Abstract
Background
Muscle dysfunction associated with sepsis contributes to morbidity and mortality but the underlying mechanisms are unclear. This study examined whether muscle weakness relates to an intrinsic defect in contraction, or to central mechanisms associated with acute illness, and whether systemic endotoxaemia induces changes in gene expression for proinflammatory cytokines within human muscle in vivo.
Methods
In this experimental study, 12 healthy men received intravenous Escherichia coli lipopolysaccharide (LPS, 4 ng/kg) or saline (control). Voluntary and electrically stimulated quadriceps contraction, and tumour necrosis factor (TNF) α mRNA expression in quadriceps muscle biopsies were studied before and after the infusion.
Results
Endotoxaemia induced transient weakness of voluntary quadriceps contraction, equivalent to a 7·8 (95 per cent confidence interval 2·1 to 13·5) per cent reduction in contractile force at 180 min (P = 0·027) and a 9·0 (5·2 to 12·8) per cent reduction at 300 min (P = 0·008). Electrically stimulated contraction was unaffected. LPS administration resulted in an apparent fibre-specific induction of TNF-α mRNA.
Conclusion
Endotoxaemia results in a reduction in voluntary muscle contractile force without an apparent defect in stimulated muscle contraction. Loss of volition may be a more important factor than intrinsic dysfunction in acute sepsis-associated human muscle weakness.
Collapse
Affiliation(s)
- F J McNicol
- Infection, Injury and Inflammation Research Group, Salford Royal NHS Foundation Trust, Hope Hospital, Salford, UK
| | - J A Hoyland
- University of Manchester School of Clinical and Laboratory Sciences, Manchester, UK
| | - R G Cooper
- Infection, Injury and Inflammation Research Group, Salford Royal NHS Foundation Trust, Hope Hospital, Salford, UK
| | - G L Carlson
- Infection, Injury and Inflammation Research Group, Salford Royal NHS Foundation Trust, Hope Hospital, Salford, UK
| |
Collapse
|
95
|
Lausen J, Pless O, Leonard F, Kuvardina ON, Koch B, Leutz A. Targets of the Tal1 transcription factor in erythrocytes: E2 ubiquitin conjugase regulation by Tal1. J Biol Chem 2009; 285:5338-46. [PMID: 20028976 DOI: 10.1074/jbc.m109.030296] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Tal1 transcription factor is essential for the development of the hematopoietic system and plays a role during definitive erythropoiesis in the adult. Despite the importance of Tal1 in erythropoiesis, only a small number of erythroid differentiation target genes are known. A chromatin precipitation and cloning approach was established to uncover novel Tal1 target genes in erythropoiesis. The BirA tag/BirA ligase biotinylation system in combination with streptavidin chromatin precipitation (Strep-CP) was used to co-precipitate genomic DNA bound to Tal1. Tal1 was found to bind in the vicinity of 31 genes including the E2-ubiquitin conjugase UBE2H gene. Binding of Tal1 to UBE2H was confirmed by chromatin immunoprecipitation. UBE2H expression is increased during erythroid differentiation of hCD34(+) cells. Tal1 expression activated UBE2H expression, whereas Tal1 knock-down reduced UBE2H expression and ubiquitin transfer activity. This study identifies parts of the ubiquitinylation machinery as a cellular target downstream of the transcription factor Tal1 and provides novel insights into Tal1-regulated erythropoiesis.
Collapse
Affiliation(s)
- Jörn Lausen
- Georg-Speyer-Haus, Institute for Biomedical Research, D-60596 Frankfurt (Main), Germany.
| | | | | | | | | | | |
Collapse
|
96
|
DI RENZO L, SARACENO R, SCHIPANI C, CHIMENTI S, DE LORENZO A. Why 3âmg/kg instead of 5âmg/kg of infliximab should work in psoriatic arthritis? J Dermatol 2009; 36:666-8. [DOI: 10.1111/j.1346-8138.2009.00730.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
97
|
|
98
|
Li W, Moylan JS, Chambers MA, Smith J, Reid MB. Interleukin-1 stimulates catabolism in C2C12 myotubes. Am J Physiol Cell Physiol 2009; 297:C706-14. [PMID: 19625606 DOI: 10.1152/ajpcell.00626.2008] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Interleukin-1 (IL-1) is an inflammatory cytokine that has been linked to muscle catabolism, a process regulated by muscle-specific E3 proteins of the ubiquitin-proteasome pathway. To address cellular mechanism, we tested the hypothesis that IL-1 induces myofibrillar protein loss by acting directly on muscle to increase expression of two critical E3 proteins, atrogin1/muscle atrophy F-box (MAFbx) and muscle RING-finger 1 (MuRF1). Experiments were conducted using mature C2C12 myotubes to eliminate systemic cytokine effects and avoid paracrine signaling by nonmuscle cell types. Time-course protocols were used to define the sequence of cellular responses. We found that atrogin1/MAFbx mRNA and MuRF1 mRNA are elevated 60-120 min after myotube exposure to either IL-1alpha or IL-1beta. These responses are preceded by signaling events that promote E3 expression. Both IL-1 isoforms stimulate phosphorylation of p38 mitogen-activated protein kinase and stimulate nuclear factor-kappaB (NF-kappaB) signaling; I-kappaB levels fall and NF-kappaB DNA binding activity increases. Other regulators of E3 expression are unaffected by IL-1 [cytosolic oxidant activity, Forkhead-O (Foxo) activity] or respond paradoxically (AKT). Chronic exposure of C2C12 myotubes over 48 h resulted in reduced myotube width and loss of sarcomeric actin. We conclude that IL-1alpha and IL-1beta act via an oxidant- and AKT/Foxo-independent mechanism to activate p38 MAPK, stimulate NF-kappaB signaling, increase expression of atrogin1/MAFbx and MuRF1, and reduce myofibrillar protein in differentiated myotubes.
Collapse
Affiliation(s)
- Wei Li
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536-0298, USA
| | | | | | | | | |
Collapse
|
99
|
Attenuation of proteolysis and muscle wasting by curcumin c3 complex in MAC16 colon tumour-bearing mice. Br J Nutr 2009; 102:967-75. [PMID: 19393114 DOI: 10.1017/s0007114509345250] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Muscle wasting or cachexia is caused by accelerated muscle protein breakdown via the ubiquitin-proteasome complex. We investigated the effect of curcumin c3 complex (curcumin c3) on attenuation of muscle proteolysis using in vitro and in vivo models. Our in vitro data indicate that curcumin c3 as low as 0.50 microg/ml was very effective in significantly inhibiting (30 %; P < 0.05) tyrosine release from human skeletal muscle cells, which reached a maximum level of inhibition of 60 % (P < 0.05) at 2.5 microg/ml. Curcumin c3 at 2.5 microg/ml also inhibited chymotrypsin-like 20S proteasome activity in these cells by 25 % (P < 0.05). For in vivo studies, we induced progressive muscle wasting in mice by implanting the MAC16 colon tumour. The in vivo data indicate that low doses of curcumin c3 (100 mg/kg body weight) was able to prevent weight loss in mice bearing MAC16 tumours whereas higher doses of curcumin c3 (250 mg/kg body weight) resulted in approximately 25 % (P < 0.05) weight gain as compared with the placebo-treated animals. Additionally, the effect of curcumin c3 on preventing and/or reversing cachexia was also evident by gains in the weight of the gastrocnemius muscle (30-58 %; P < 0.05) and with the increased size of the muscle fibres (30-65 %; P < 0.05). Furthermore, curcumin inhibited proteasome complex activity and variably reduced expression of muscle-specific ubiquitin ligases: atrogin-1/muscle atrophy F-box (MAFbx) and muscle RING finger 1 (MURF-1). In conclusion, oral curcumin c3 results in the prevention and reversal of weight loss. The data imply that curcumin c3 may be an effective adjuvant therapy against cachexia.
Collapse
|
100
|
Supinski GS, Vanags J, Callahan LA. Effect of proteasome inhibitors on endotoxin-induced diaphragm dysfunction. Am J Physiol Lung Cell Mol Physiol 2009; 296:L994-L1001. [PMID: 19376888 DOI: 10.1152/ajplung.90404.2008] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Infections produce severe respiratory muscle dysfunction. It is known that the proteasome proteolytic system is activated in skeletal muscle in sepsis, and it has been postulated that this degradative pathway is responsible for inducing skeletal muscle weakness and wasting. The objective of this study was to determine if administration of proteasomal inhibitors (MG132, epoxomicin, bortezomib) can prevent sepsis-induced diaphragm weakness. Rats were given either 1) saline (0.5 ml ip), 2) endotoxin (12 mg/kg ip), 3) endotoxin plus MG132 (2.5 mg/kg), 4) endotoxin plus epoxomicin (1 micromol/kg), or 5) endotoxin plus bortezomib (0.05 mg/kg). Animals were killed either 48 or 96 h after injections, and assessments were made of diaphragm proteolysis, force-frequency relationships, mass, protein content, and caspase activation. Endotoxin increased proteolysis (P <0.001). MG132, epoxomicin, and bortezomib each prevented the endotoxin-induced increase in proteolysis (P <0.01). Endotoxin induced severe reductions in diaphragm force generation by 48 h (P <0.01); none of the proteasomal inhibitors prevented loss of force. Endotoxin induced significant reductions in diaphragm mass and protein content by 96 h (P <0.01); neither MG132 nor epoxomicin prevented loss of mass or protein, but bortezomib attenuated the reduction in protein content (P <0.05). Endotoxin increased diaphragm caspase-3 activity (P <0.01); caspase-3 activity remained high when either MG132, epoxomicin, or bortezomib were given. These data suggest proteasomal inhibitors are not an adequate treatment to prevent endotoxin-induced diaphragmatic dysfunction.
Collapse
Affiliation(s)
- G S Supinski
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Kentucky, Lexington, KY 40536-0284, USA.
| | | | | |
Collapse
|