151
|
Chokeshaiusaha K, Puthier D, Sananmuang T, Olanratmanee EO, Nguyen C, Kedkovid R. Differential DNA methylation analysis across the promoter regions using methylated DNA immunoprecipitation sequencing profiling of porcine loin muscle. Vet World 2020; 13:1113-1125. [PMID: 32801562 PMCID: PMC7396332 DOI: 10.14202/vetworld.2020.1113-1125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Background and Aim: Pork leanness and marbling are among the essential traits of consumer preference. To acquire knowledge about universal epigenetic regulations for improving breed selection, a meta-analysis of methylated DNA immunoprecipitation sequencing (MeDIP-seq) profiling data of mixed loin muscle types was performed in this study. Materials and Methods: MeDIP-seq profiling datasets of longissimus dorsi muscle and psoas major muscles from male and female pigs of Landrace and Tibetan breeds were preprocessed and aligned to the porcine genome. Analysis of differential methylated DNA regions (DMRs) between the breeds was performed by focusing on transcription start sites (TSSs) of known genes (−20,000-3000 bases from TSS). All associated genes were further reviewed for their functions and predicted for transcription factors (TF) possibly associated with their TSSs. Results: When the methylation levels of DMRs in TSS regions of Landrace breed were compared to those of Tibetan breed, 10 DMRs were hypomethylated (Landrace < Tibetan), and 19 DMRs were hypermethylated (Landrace > Tibetan), accordingly (p≤0.001). According to the reviews about gene functions, all associated genes were pieces of evidence for their roles in a variety of muscle and lipid metabolisms. Prediction of the binding TFs revealed the six most abundant binding TFs to such DMRs-associated TSS (p≤0.0001) as follows: ZNF384, Foxd3, IRF1, KLF9, EWSR1-FLI1, HES5, and TFAP2A. Conclusion: Common DMRs-associated TSS between the lean-type and the marbled-type loin muscles were identified in this study. Interestingly, the genes associated with such regions were strongly evidenced for their possible roles on the muscle trait characteristics by which further novel research topics could be focused on them in the future.
Collapse
Affiliation(s)
- Kaj Chokeshaiusaha
- Department of Veterinary Science, Faculty of Veterinary Medicine, Rajamangala University of Technology Tawan-OK, Chon Buri, Thailand
| | - Denis Puthier
- Aix-Marseille University, INSERM UMR 1090, TAGC, Marseille, France
| | - Thanida Sananmuang
- Department of Veterinary Science, Faculty of Veterinary Medicine, Rajamangala University of Technology Tawan-OK, Chon Buri, Thailand
| | - Em-On Olanratmanee
- Department of Veterinary Science, Faculty of Veterinary Medicine, Rajamangala University of Technology Tawan-OK, Chon Buri, Thailand
| | - Catherine Nguyen
- Aix-Marseille University, INSERM UMR 1090, TAGC, Marseille, France
| | - Roongtham Kedkovid
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Swine Reproduction Research Unit, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
152
|
Ramalho R, Rao M, Zhang C, Agrati C, Ippolito G, Wang FS, Zumla A, Maeurer M. Immunometabolism: new insights and lessons from antigen-directed cellular immune responses. Semin Immunopathol 2020; 42:279-313. [PMID: 32519148 PMCID: PMC7282544 DOI: 10.1007/s00281-020-00798-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023]
Abstract
Modulation of immune responses by nutrients is an important area of study in cellular biology and clinical sciences in the context of cancer therapies and anti-pathogen-directed immune responses in health and disease. We review metabolic pathways that influence immune cell function and cellular persistence in chronic infections. We also highlight the role of nutrients in altering the tissue microenvironment with lessons from the tumor microenvironment that shapes the quality and quantity of cellular immune responses. Multiple layers of biological networks, including the nature of nutritional supplements, the genetic background, previous exposures, and gut microbiota status have impact on cellular performance and immune competence against molecularly defined targets. We discuss how immune metabolism determines the differentiation pathway of antigen-specific immune cells and how these insights can be explored to devise better strategies to strengthen anti-pathogen-directed immune responses, while curbing unwanted, non-productive inflammation.
Collapse
Affiliation(s)
- Renata Ramalho
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM, U4585 FCT), Applied Nutrition Studies Group G.E.N.A.-IUEM), Instituto Universitário Egas Moniz, Egas Moniz Higher Education School, Monte de Caparica, Portugal
| | - Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Chao Zhang
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | | | | | - Fu-Sheng Wang
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London and NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.
- I Medizinische Klinik, Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
153
|
Kuo YK, Lin YC, Lee CY, Chen CY, Tani J, Huang TJ, Chang H, Wu MH. Novel Insights into the Pathogenesis of Spinal Sarcopenia and Related Therapeutic Approaches: A Narrative Review. Int J Mol Sci 2020; 21:E3010. [PMID: 32344580 PMCID: PMC7216136 DOI: 10.3390/ijms21083010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023] Open
Abstract
Spinal sarcopenia is a complex and multifactorial disorder associated with a loss of strength, increased frailty, and increased risks of fractures and falls. In addition, spinal sarcopenia has been associated with lumbar spine disorders and osteoporosis, which renders making decisions on treatment modalities difficult. Patients with spinal sarcopenia typically exhibit lower cumulative survival, a higher risk of in-hospital complications, prolonged hospital stays, higher postoperative costs, and higher rates of blood transfusion after thoracolumbar spine surgery. Several studies have focused on the relationships between spinal sarcopenia, appendicular muscle mass, and bone-related problems-such as osteoporotic fractures and low bone mineral density-and malnutrition and vitamin D deficiency. Although several techniques are available for measuring sarcopenia, each of them has its advantages and shortcomings. For treating spinal sarcopenia, nutrition, physical therapy, and medication have been proven to be effective; regenerative therapeutic options seem to be promising owing to their repair and regeneration potential. Therefore, in this narrative review, we summarize the characteristics, detection methodologies, and treatment options for spinal sarcopenia, as well as its role in spinal disorders.
Collapse
Affiliation(s)
- Yu-Kai Kuo
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Medicine, Keelung Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Yu-Ching Lin
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Keelung & Chang Gung University, College of Medicine, Keelung 20401, Taiwan;
| | - Ching-Yu Lee
- Department of Orthopedics, Taipei Medical University Hospital, Taipei 11031, Taiwan; (C.-Y.L.)
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chih-Yu Chen
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Stanford Byers Center for Biodesign, Stanford University, Stanford, CA 94305, USA;
| | - Jowy Tani
- Stanford Byers Center for Biodesign, Stanford University, Stanford, CA 94305, USA;
- Department of Neurology, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 11031, Taiwan
| | - Tsung-Jen Huang
- Department of Orthopedics, Taipei Medical University Hospital, Taipei 11031, Taiwan; (C.-Y.L.)
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Hsi Chang
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Pediatrics, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Meng-Huang Wu
- Department of Orthopedics, Taipei Medical University Hospital, Taipei 11031, Taiwan; (C.-Y.L.)
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Stanford Byers Center for Biodesign, Stanford University, Stanford, CA 94305, USA;
| |
Collapse
|
154
|
Ahmed OAA, Fahmy UA, Bakhaidar R, El-Moselhy MA, Okbazghi SZ, Ahmed ASF, Hammad ASA, Alhakamy NA. Omega-3 Self-Nanoemulsion Role in Gastroprotection against Indomethacin-Induced Gastric Injury in Rats. Pharmaceutics 2020; 12:E140. [PMID: 32045979 PMCID: PMC7076357 DOI: 10.3390/pharmaceutics12020140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/25/2020] [Accepted: 02/03/2020] [Indexed: 01/20/2023] Open
Abstract
Peptic ulcer disease is an injury of the alimentary tract that leads to a mucosal defect reaching the submucosa. This study aimed to formulate and optimize omega-3 oil as a self-nanoemulsifying drug delivery system (SNEDDS) to achieve oil dispersion in the nano-range in the stomach to augment omega-3 oil gastric ulcer protection efficacy. Three SNEDDS components were selected as the design factors: the concentrations of the oil omega-3 (X1, 10-30%), the surfactant tween 20 and Kolliphor mixture (X2, 20-40%), and the cosurfactant transcutol (X3, 40-60%). The mixture experimental design proposed twenty-three formulations with varying omega-3 SNEDDS formulation component percentages. The optimized omega-3 SNEDDS formula was investigated for gastric ulcer protective effects by evaluating the ulcer index and by the determination of gastric mucosa oxidative stress parameters. Results revealed that optimized omega-3-SNEDDS achieved significant improvement in the gastric ulcer index in comparison with pure omega-3 oil. Histopathological findings confirmed the protective effect of the formulated optimized omega-3 SNEDDS in comparison with omega-3 oil. These findings suggest that formulation of omega-3 in the form of a SNEDDS would be more effective in gastric ulcer protection than the administration of omega-3 as a crude oil.
Collapse
Affiliation(s)
- Osama A A Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Pharmaceutics & Industrial Pharmacy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Usama A Fahmy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rana Bakhaidar
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed A El-Moselhy
- Department of Pharmacology, School of Pharmacy, Ibn Sina national college, Jeddah 22413, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Solomon Z Okbazghi
- Global Analytical and Pharmaceutical Development, Alexion Pharmaceuticals, New Haven, CT 06510, USA
| | - Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Asmaa S A Hammad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
155
|
Graham ZA, Goldberger A, Azulai D, Conover CF, Ye F, Bauman WA, Cardozo CP, Yarrow JF. Contusion spinal cord injury upregulates p53 protein expression in rat soleus muscle at multiple timepoints but not key senescence cytokines. Physiol Rep 2020; 8:e14357. [PMID: 32026570 PMCID: PMC7002538 DOI: 10.14814/phy2.14357] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/12/2019] [Accepted: 12/27/2019] [Indexed: 01/04/2023] Open
Abstract
To determine whether muscle disuse after a spinal cord injury (SCI) produces elevated markers of cellular senescence and induces markers of the senescence-associated secretory phenotypes (SASPs) in paralyzed skeletal muscle. Four-month-old male Sprague-Dawley rats received a moderate-severe (250 kiloDyne) T-9 contusion SCI or Sham surgery and were monitored over 2 weeks, and 1-, 2-, or 3 months. Animals were sacrificed via isoflurane overdose and terminal exsanguination and the soleus was carefully excised and snap frozen. Protein expression of senescence markers p53, p27, and p16 was determined from whole soleus lysates using Western immunoblotting and RT-qPCR was used to determine the soleus gene expression of IL-1α, IL-1β, IL-6, CXCL1, and TNFα. SCI soleus muscle displayed 2- to 3-fold higher total p53 protein expression at 2 weeks, and at 1 and 2 months when compared with Sham. p27 expression was stable across all groups and timepoints. p16 protein expression was lower at 3 months in SCI versus Sham, but not earlier timepoints. Gene expression was relatively stable between groups at 2 weeks. There were Surgery x Time interaction effects for IL-6 and TNFα mRNA expression but not for IL-1α, IL-1β, or CXCL1. There were no main effects for time or surgery for IL-1α, IL-1β, or CXCL1, but targeted t tests showed reductions in IL-1α and CXCL1 in SCI animals compared to Sham at 3 months and IL-1β was reduced in SCI animals compared to Sham animals at the 2-month timepoint. The elevation in p53 does not appear consistent with the induction of SASP because mRNA expression of cytokines associated with senescence was not uniformly upregulated and, in some instances, was downregulated in the early chronic phase of SCI.
Collapse
Affiliation(s)
- Zachary A. Graham
- Research ServiceBirmingham VA Medical CenterBirminghamALUSA
- Department of Cell, Developmental and Integrative BiologyUniversity of Alabama‐BirminghamBirminghamALUSA
| | - Abigail Goldberger
- Center for the Medical Consequences of Spinal Cord InjuryJames J. Peters VA Medical CenterBronxNYUSA
| | - Daniella Azulai
- Center for the Medical Consequences of Spinal Cord InjuryJames J. Peters VA Medical CenterBronxNYUSA
| | - Christine F. Conover
- Research Service and Brain Rehabilitation Research CenterMalcolm Randall VA Medical CenterNorth Florida/South Georgia Veterans Health SystemGainesvilleFLUSA
| | - Fan Ye
- Research Service and Brain Rehabilitation Research CenterMalcolm Randall VA Medical CenterNorth Florida/South Georgia Veterans Health SystemGainesvilleFLUSA
| | - William A. Bauman
- Center for the Medical Consequences of Spinal Cord InjuryJames J. Peters VA Medical CenterBronxNYUSA
- Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Christopher P. Cardozo
- Center for the Medical Consequences of Spinal Cord InjuryJames J. Peters VA Medical CenterBronxNYUSA
- Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Joshua F. Yarrow
- Research Service and Brain Rehabilitation Research CenterMalcolm Randall VA Medical CenterNorth Florida/South Georgia Veterans Health SystemGainesvilleFLUSA
- Division of Endocrinology, Diabetes, and MetabolismUniversity of Florida College of MedicineGainesvilleFLUSA
| |
Collapse
|
156
|
Hu Y, Zhang T, Chen J, Cheng W, Chen J, Zheng Z, Lin J, Zhu G, Zhang Y, Bai X, Wang Y, Song B, Wang Q, Qin L, Zhang P. Downregulation of Hypoxia-Inducible Factor-1α by RNA Interference Alleviates the Development of Collagen-Induced Arthritis in Rats. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:1330-1342. [PMID: 32160704 PMCID: PMC7038004 DOI: 10.1016/j.omtn.2020.01.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/13/2022]
Abstract
Rheumatoid arthritis (RA) is the most common type of autoimmune arthritis. Hypoxia-inducible factor-1α (HIF-1α) as a transcription factor in response to hypoxia suggests that it could be a potential therapeutic target for the treatment of RA. In this study, we assessed whether the HIF pathway blockade attenuates the manifestations of RA in the collagen-induced arthritis (CIA) rat model. We constructed a short hairpin RNA (shRNA) lentiviral expression vector targeting HIF-1α (pLVX-shRNA-HIF-1α) and to achieve HIF-1α RNA interference. Quantitative RT-PCR, immunofluorescence staining, and western blot were used to detect the expressions of HIF-1α, vascular endothelial growth factor (VEGF), phsopho (p)-p65, and p-IКBɑ mRNA and protein, respectively. Micro-computed tomography was used to investigate joint morphology at different time points after CIA induction. Moreover, enzyme-linked immunosorbent assay (ELISA) was used to monitor the expression of inflammatory cytokines. In vitro analyses revealed that pLVX-shRNA-HIF-1α effectively inhibited the expression of HIF-1α and VEGF and led to the activation of p-65 and p-IКBɑ, as well as decreased proinflammatory cytokine expression in cell culture. Inhibition of HIF-1α in rats decreased signs of a systemic inflammatory condition, together with decreased pathological changes of RA. Moreover, downregulation of HIF-1α expression markedly reduced the synovitis and angiogenesis. In conclusion, we have shown that pharmacological inhibition of HIF-1 may improve the clinical manifestations of RA.
Collapse
Affiliation(s)
- Yiping Hu
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Tiantian Zhang
- Department of Rheumatology, People's Hospital of Bao'an District, Shenzhen, Guangdong 518128, China
| | - Jingqin Chen
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - WenXiang Cheng
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - Jianhai Chen
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - Zhengtan Zheng
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Jietao Lin
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - Guoyuan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Yong Zhang
- Shenzhen Pingle Orthopaedic Hospital, Guangdong 518000, China
| | - Xueling Bai
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Yan Wang
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Bing Song
- School of Dentistry, Cardiff University, Cardiff, Heath Park, CF23 6AL Wales, UK
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China.
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China; University of Chinese Academy of Sciences, Shenzhen Hospital, Shenzhen 518000, China.
| |
Collapse
|
157
|
M1 macrophage infiltration exacerbate muscle/bone atrophy after peripheral nerve injury. BMC Musculoskelet Disord 2020; 21:44. [PMID: 31959156 PMCID: PMC6971979 DOI: 10.1186/s12891-020-3069-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/15/2020] [Indexed: 01/15/2023] Open
Abstract
Background Peripheral nerve injury causes limb muscle/bone atrophy, leading to chronic pain. However, the mechanisms underlying muscle/bone atrophy after peripheral nerve injury remain unknown. It was recently reported that M1 macrophages are the main factors responsible for neurogenic inflammation after peripheral nerve injury. We hypothesized that M1 macrophages are important in muscle/bone atrophy after nerve injury. Therefore, we investigated the influence of M1 macrophages on muscle/bone atrophy after nerve injury in mice to prevent muscle/bone atrophy by suppressing M1 macrophages. Methods Hindlimb muscle weight and total bone density were measured in a chronic constriction injury (CCI) mouse model. Immunohistochemical analysis and intravital microscopy were performed to visualize hindlimb muscles/bones, and cells were quantified using flow cytometry. We compared M1 macrophage infiltration into muscles/bones and muscle/bone atrophy between macrophage depletion and untreated groups. We also investigated muscle/bone atrophy using administration models for anti-inflammatory and neuropathic pain drugs. Results Peripheral nerve injury caused significant reduction in muscle weight and total bone density at 1 and 3 weeks after CCI, respectively, compared with that in controls. Osteoclast numbers were significantly higher at 1 week after CCI in the CCI group than in the control group. M1 macrophage infiltration into muscles was observed from 2 h after CCI via intravital microscopy and 1 week after CCI, and it was significantly higher 1 week after CCI than in the control group. In the macrophage depletion group, dexamethasone, pregabalin, and loxoprofen groups, M1 macrophage infiltration into muscles/bones was significantly lower and muscle weight and total bone density were significantly higher than in the untreated group. Conclusions M1 macrophage infiltration exacerbates muscle/bone atrophy after peripheral nerve injury. By suppressing M1 macrophages at the neural injury local site, muscle/bone atrophy could be avoided.
Collapse
|
158
|
Nwaogu CJ, Galema A, Cresswell W, Dietz MW, Tieleman BI. A fruit diet rather than invertebrate diet maintains a robust innate immunity in an omnivorous tropical songbird. J Anim Ecol 2020; 89:867-883. [PMID: 31764994 PMCID: PMC7079115 DOI: 10.1111/1365-2656.13152] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 11/04/2019] [Indexed: 11/30/2022]
Abstract
Diet alteration may lead to nutrient limitations even in the absence of food limitation, and this may affect physiological functions, including immunity. Nutrient limitations may also affect the maintenance of body mass and key life‐history events that may affect immune function. Yet, variation in immune function is largely attributed to energetic trade‐offs rather than specific nutrient constraints. To test the effect of diet on life‐history traits, we tested how diet composition affects innate immune function, body mass and moult separately and in combination with each other, and then used path analyses to generate hypotheses about the mechanistic connections between immunity and body mass under different diet compositions. We performed a balanced parallel and crossover design experiment with omnivorous common bulbuls Pycnonotus barbatus in out‐door aviaries in Nigeria. We fed 40 wild‐caught bulbuls ad libitum on fruits or invertebrates for 24 weeks, switching half of each group between treatments after 12 weeks. We assessed innate immune indices (haptoglobin, nitric oxide and ovotransferrin concentrations, and haemagglutination and haemolysis titres), body mass and primary moult, fortnightly. We simplified immune indices into three principal components (PCs), but we explored mechanistic connections between diet, body mass and each immune index separately. Fruit‐fed bulbuls had higher body mass, earlier moult and showed higher values for two of the three immune PCs compared to invertebrate‐fed bulbuls. These effects were reversed when we switched bulbuls between treatments after 12 weeks. Exploring the correlations between immune function, body mass and moult, showed that an increase in immune function was associated with a decrease in body mass and delayed moult in invertebrate‐fed bulbuls, while fruit‐fed bulbuls maintained body mass despite variation in immune function. Path analyses indicated that diet composition was most likely to affect body mass and immune indices directly and independently from each other. Only haptoglobin concentration was indirectly linked to diet composition via body mass. We demonstrated a causal effect of diet composition on innate immune function, body mass and moult: bulbuls were in a better condition when fed on fruits than invertebrates, confirming that innate immunity is nutrient specific. Our results are unique because they show a reversible effect of diet composition on wild adult birds whose immune systems are presumably fully developed and adapted to wild conditions—demonstrating a short‐term consequence of diet alteration on life‐history traits.
Collapse
Affiliation(s)
- Chima J Nwaogu
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands.,School of Biology, University of St Andrews, St Andrews Fife, UK.,A.P. Leventis Ornithological Research Institute, Jos, Nigeria
| | - Annabet Galema
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Will Cresswell
- School of Biology, University of St Andrews, St Andrews Fife, UK.,A.P. Leventis Ornithological Research Institute, Jos, Nigeria
| | - Maurine W Dietz
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - B Irene Tieleman
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
159
|
Zhang HY, Yang HX, Liu Q, Xie MJ, Zhang J, Liu X, Liu XD, Yu SB, Lu L, Zhang M, Wang MQ. Injury responses of Sprague-Dawley rat jaw muscles to an experimental unilateral anterior crossbite prosthesis. Arch Oral Biol 2020; 109:104588. [DOI: 10.1016/j.archoralbio.2019.104588] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 10/14/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
|
160
|
Pacifico J, Geerlings MAJ, Reijnierse EM, Phassouliotis C, Lim WK, Maier AB. Prevalence of sarcopenia as a comorbid disease: A systematic review and meta-analysis. Exp Gerontol 2019; 131:110801. [PMID: 31887347 DOI: 10.1016/j.exger.2019.110801] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/28/2019] [Accepted: 11/28/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Sarcopenia shares risk factors with various other age-related diseases. This meta-analysis aimed to determine the prevalence of sarcopenia as a comorbid disease. METHODS Medline, EMBASE and Cochrane databases were searched for articles from inception to 8th June 2018, reporting the prevalence of sarcopenia in individuals with a diagnosis of cardiovascular disease (CVD), dementia, diabetes mellitus or respiratory disease and, if applicable their controls. No exclusion criteria were applied with regards to definition of sarcopenia, individuals' age, study design and setting. Meta-analyses were stratified by disease, definition of sarcopenia and continent. RESULTS The 63 included articles described 17,206 diseased individuals (mean age: 65.3 ± 1.6 years, 49.9% females) and 22,375 non-diseased controls (mean age: 54.6 ± 16.2 years, 53.8% females). The prevalence of sarcopenia in individuals with CVD was 31.4% (95% CI: 22.4-42.1%), no controls were available. The prevalence of sarcopenia was 26.4% (95% CI: 13.6-44.8%) in individuals with dementia compared to 8.3% (95% CI: 2.8-21.9%) in their controls; 31.1% (95% CI: 19.8-45.2%) in individuals with diabetes mellitus compared to 16.2% (95% CI: 9.5-26.2%) in controls; and 26.8% (95% CI: 17.8-38.1%) in individuals with respiratory diseases compared to 13.3% (95% CI: 8.3-20.7%) in controls. CONCLUSIONS Sarcopenia is highly prevalent in individuals with CVD, dementia, diabetes mellitus and respiratory disease.
Collapse
Affiliation(s)
- Jacob Pacifico
- Department of Medicine and Aged Care, @AgeMelbourne, The Royal Melbourne Hospital, The University of Melbourne, Victoria, Australia
| | - Milou A J Geerlings
- Department of Clinical Physical Therapy, VieCuri Medical Center, Venlo, the Netherlands; Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit, Amsterdam, the Netherlands
| | - Esmee M Reijnierse
- Department of Medicine and Aged Care, @AgeMelbourne, The Royal Melbourne Hospital, The University of Melbourne, Victoria, Australia
| | - Christina Phassouliotis
- Department of Medicine and Aged Care, @AgeMelbourne, The Royal Melbourne Hospital, The University of Melbourne, Victoria, Australia
| | - Wen Kwang Lim
- Department of Medicine and Aged Care, @AgeMelbourne, The Royal Melbourne Hospital, The University of Melbourne, Victoria, Australia
| | - Andrea B Maier
- Department of Medicine and Aged Care, @AgeMelbourne, The Royal Melbourne Hospital, The University of Melbourne, Victoria, Australia; Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit, Amsterdam, the Netherlands.
| |
Collapse
|
161
|
Bilen MA, Martini DJ, Liu Y, Shabto JM, Brown JT, Williams M, Khan AI, Speak A, Lewis C, Collins H, Kissick HT, Carthon BC, Akce M, Shaib WL, Alese OB, Pillai RN, Steuer CE, Wu CS, Lawson DH, Kudchadkar RR, El‐Rayes BF, Ramalingam SS, Owonikoko TK, Harvey RD, Master VA. Combined Effect of Sarcopenia and Systemic Inflammation on Survival in Patients with Advanced Stage Cancer Treated with Immunotherapy. Oncologist 2019; 25:e528-e535. [PMID: 32162807 PMCID: PMC7066707 DOI: 10.1634/theoncologist.2019-0751] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/31/2019] [Indexed: 12/27/2022] Open
Abstract
Background Sarcopenia and inflammation have been associated with poor survival in patients with cancer. We explored the combined effects of these variables on survival in patients with cancer treated with immunotherapy. Methods We performed a retrospective review of 90 patients enrolled on immunotherapy‐based phase I clinical trials at Emory University from 2009 to 2017. Baseline neutrophil‐to‐lymphocyte ratio, monocyte‐to‐lymphocyte ratio, and platelet‐to‐lymphocyte ratio (PLR) were used as surrogates of inflammation. The skeletal muscle index (SMI) was derived from the skeletal muscle density calculated from baseline abdominal computed tomography images. Optimal cutoffs for continuous inflammation biomarkers and SMI were determined by bias‐adjusted log‐rank test. A four‐level risk stratification was used to create low‐risk (PLR <242 and nonsarcopenic), intermediate‐risk (PLR <242 and sarcopenic), high‐risk (PLR ≥242 and nonsarcopenic), and very‐high‐risk (PLR ≥242 and sarcopenic) groups with subsequent association with survival. Results Most patients (59%) were male, and the most common cancers were melanoma (33%) and gastrointestinal (22%). Very high‐risk, high‐risk, and intermediate‐risk patients had significantly shorter overall survival (hazard ratio [HR], 8.46; 95% confidence interval [CI], 2.65–27.01; p < .001; HR, 5.32; CI, 1.96–14.43; p = .001; and HR, 4.01; CI, 1.66–9.68; p = .002, respectively) and progression‐free survival (HR, 12.29; CI, 5.15–29.32; p < .001; HR, 3.51; CI, 1.37–9.02; p = .009; and HR, 2.14; CI, 1.12–4.10; p = .022, respectively) compared with low‐risk patients. Conclusion Baseline sarcopenia and elevated inflammatory biomarkers may have a combined effect on decreasing survival in immunotherapy‐treated patients in phase I trials. These data may be immediately applicable for medical oncologists for the risk stratification of patients beginning immunotherapeutic agents. Implications for Practice Sarcopenia and inflammation have been associated with poor survival in patients with cancer, but it is unclear how to apply this information to patient care. The authors created a risk‐stratification system that combined sarcopenia and platelet‐to‐lymphocyte ratio as a marker of systemic inflammation. The presence of sarcopenia and systemic inflammation decreased progression‐free survival and overall survival in our cohort of 90 patients who received immunotherapy in phase I clinical trials. The data presented in this study may be immediately applicable for medical oncologists as a way to risk‐stratify patients who are beginning treatment with immunotherapy. The interaction between chronic inflammation and body composition is particularly important in the era of immunotherapy, considering that immune checkpoint inhibitors rely on the host immune system for their efficacy. This article reports on the combined effects of inflammation and sarcopenia on clinical outcomes in patients with solid tumors treated with immunotherapy‐based regimens.
Collapse
Affiliation(s)
- Mehmet Asim Bilen
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Dylan J. Martini
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Yuan Liu
- Departments of Biostatistics and Bioinformatics, Emory UniversityAtlantaGeorgiaUSA
| | - Julie M. Shabto
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Jacqueline T. Brown
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Milton Williams
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Amir I. Khan
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Alexandra Speak
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Colleen Lewis
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Hannah Collins
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Haydn T. Kissick
- Department of Urology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Bradley C. Carthon
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Mehmet Akce
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Walid L. Shaib
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Olatunji B. Alese
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Rathi N. Pillai
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Conor E. Steuer
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Christina S. Wu
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - David H. Lawson
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Ragini R. Kudchadkar
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Bassel F. El‐Rayes
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Suresh S. Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Taofeek K. Owonikoko
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - R. Donald Harvey
- Department of Hematology and Medical Oncology, Emory University School of MedicineAtlantaGeorgiaUSA
- Department of Pharmacology, Emory University School of MedicineAtlantaGeorgiaUSA
- Winship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Viraj A. Master
- Department of Urology, Emory University School of MedicineAtlantaGeorgiaUSA
| |
Collapse
|
162
|
Livshits G, Kalinkovich A. Inflammaging as a common ground for the development and maintenance of sarcopenia, obesity, cardiomyopathy and dysbiosis. Ageing Res Rev 2019; 56:100980. [PMID: 31726228 DOI: 10.1016/j.arr.2019.100980] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 12/12/2022]
Abstract
Sarcopenia, obesity and their coexistence, obese sarcopenia (OBSP) as well as atherosclerosis-related cardio-vascular diseases (ACVDs), including chronic heart failure (CHF), are among the greatest public health concerns in the ageing population. A clear age-dependent increased prevalence of sarcopenia and OBSP has been registered in CHF patients, suggesting mechanistic relationships. Development of OBSP could be mediated by a crosstalk between the visceral and subcutaneous adipose tissue (AT) and the skeletal muscle under conditions of low-grade local and systemic inflammation, inflammaging. The present review summarizes the emerging data supporting the idea that inflammaging may serve as a mutual mechanism governing the development of sarcopenia, OBSP and ACVDs. In support of this hypothesis, various immune cells release pro-inflammatory mediators in the skeletal muscle and myocardium. Subsequently, the endothelial structure is disrupted, and cellular processes, such as mitochondrial activity, mitophagy, and autophagy are impaired. Inflamed myocytes lose their contractile properties, which is characteristic of sarcopenia and CHF. Inflammation may increase the risk of ACVD events in a hyperlipidemia-independent manner. Significant reduction of ACVD event rates, without the lowering of plasma lipids, following a specific targeting of key pro-inflammatory cytokines confirms a key role of inflammation in ACVD pathogenesis. Gut dysbiosis, an imbalanced gut microbial community, is known to be deeply involved in the pathogenesis of age-associated sarcopenia and ACVDs by inducing and supporting inflammaging. Dysbiosis induces the production of trimethylamine-N-oxide (TMAO), which is implicated in atherosclerosis, thrombosis, metabolic syndrome, hypertension and poor CHF prognosis. In OBSP, AT dysfunction and inflammation induce, in concert with dysbiosis, lipotoxicity and other pathophysiological processes, thus exacerbating sarcopenia and CHF. Administration of specialized, inflammation pro-resolving mediators has been shown to ameliorate the inflammatory manifestations. Considering all these findings, we hypothesize that sarcopenia, OBSP, CHF and dysbiosis are inflammaging-oriented disorders, whereby inflammaging is common and most probably the causative mechanism driving their pathogenesis.
Collapse
Affiliation(s)
- Gregory Livshits
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel.; Adelson School of Medicine, Ariel University, Ariel, Israel..
| | - Alexander Kalinkovich
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
163
|
Straughn AR, Kakar SS. Withaferin A ameliorates ovarian cancer-induced cachexia and proinflammatory signaling. J Ovarian Res 2019; 12:115. [PMID: 31767036 PMCID: PMC6878639 DOI: 10.1186/s13048-019-0586-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Ovarian cancer is the fifth leading cause of cancer-related deaths amongst women in the United States. Cachexia is the primary cause of death in approximately 30% of cancer patients, and is often evidenced in ovarian cancer patients. We tested the steroidal lactone Withaferin A to examine if it could ameliorate ovarian cancer-induced cachexia. METHODS Six-week-old severely immunodeficient female mice were xenografted with the ovarian cancer cell line A2780 followed by treatment with Withaferin A or vehicle. Changes in functional grip strength were assessed on a weekly basis. Postmortem, H&E staining was performed on skeletal muscle sections and immunofluorescent immunohistochemistry was performed on skeletal muscle and tumor sections. The levels of NF-κB-related proinflammatory cytokines were assessed in the xenografted tumors and in resident host skeletal muscle. RESULTS Xenografting of the A2780 cell line resulted in a significant rate of mortality, which was attenuated by a therapeutic dosage of Withaferin A. Mice that received vehicle treatment following xenografting exhibited functional muscle decline over the course of the study. The therapeutic dosage Withaferin A treatment attenuated this reduction in grip strength, whereas the supratherapeutic dosage of Withaferin A was found to be toxic/lethal and demonstrated a further decline in functional muscle strength and an increased rate of mortality on par with vehicle treatment. At a histological level, the vehicle treated tumor-bearing mice exhibited a profound reduction in myofibrillar cross-sectional area compared to the vehicle treated tumor-free control group. The atrophic changes induced by the xenografted tumor were significantly ameliorated by treatment with Withaferin A. The combination of functional muscle weakening and induction of myofibrillar atrophy corroborate a cachectic phenotype, which was functionally rescued by Withaferin A. Further, treatment completely abolished the slow-to-fast myofiber type conversion observed in the settings of cancer-induced cachexia. In both host resident skeletal muscle and the xenografted tumors, we report an increase in NF-κB-related proinflammatory cytokines that was reversed by Withaferin A treatment. Finally, we demonstrated that Withaferin A significantly downregulates cytosolic and nuclear levels of phospho-p65, the active canonical NF-κB transcription factor, in xenografted tumors. CONCLUSIONS Cumulatively, our results demonstrate a previously overlooked role of Withaferin A in a xenograft model of ovarian cancer. We propose mechanisms by which Withaferin A reduces NF-κB-dependent pro-inflammatory cytokine production leading to an attenuation of the cachectic phenotype in an i.p. xenograft model of ovarian cancer.
Collapse
Affiliation(s)
- Alex R Straughn
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Sham S Kakar
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA.
- Department of Physiology, University of Louisville, School of Medicine, 500 South Floyd Street, Louisville, KY, 40202, USA.
| |
Collapse
|
164
|
Exploring the Interface between Inflammatory and Therapeutic Glucocorticoid Induced Bone and Muscle Loss. Int J Mol Sci 2019; 20:ijms20225768. [PMID: 31744114 PMCID: PMC6888251 DOI: 10.3390/ijms20225768] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 02/02/2023] Open
Abstract
Due to their potent immunomodulatory anti-inflammatory properties, synthetic glucocorticoids (GCs) are widely utilized in the treatment of chronic inflammatory disease. In this review, we examine our current understanding of how chronic inflammation and commonly used therapeutic GCs interact to regulate bone and muscle metabolism. Whilst both inflammation and therapeutic GCs directly promote systemic osteoporosis and muscle wasting, the mechanisms whereby they achieve this are distinct. Importantly, their interactions in vivo are greatly complicated secondary to the directly opposing actions of GCs on a wide array of pro-inflammatory signalling pathways that underpin catabolic and anti-anabolic metabolism. Several clinical studies have attempted to address the net effects of therapeutic glucocorticoids on inflammatory bone loss and muscle wasting using a range of approaches. These have yielded a wide array of results further complicated by the nature of inflammatory disease, underlying the disease management and regimen of GC therapy. Here, we report the latest findings related to these pathway interactions and explore the latest insights from murine models of disease aimed at modelling these processes and delineating the contribution of pre-receptor steroid metabolism. Understanding these processes remains paramount in the effective management of patients with chronic inflammatory disease.
Collapse
|
165
|
Cistanche tubulosa (Schenk) Wight Extract Enhances Hindlimb Performance and Attenuates Myosin Heavy Chain IId/IIx Expression in Cast-Immobilized Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9283171. [PMID: 31885674 PMCID: PMC6925718 DOI: 10.1155/2019/9283171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/13/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022]
Abstract
Skeletal muscle atrophy is encountered in many clinical conditions, but a pharmacological treatment has not yet been established. Cistanche tubulosa (Schenk) Wight is an herbal medicine used in traditional Japanese and Chinese medicine. In the current study, we investigated the effect of C. tubulosa extract (CTE) on atrophied muscle in vivo. We also investigated hindlimb cast immobilization in mice and devised a novel type of hindlimb-immobilizing cast, consisting of sponge-like tape and a thin plastic tube. Using this method, 3 out of 4 groups of mice (n = 11 for each group) were cast-immobilized in the hindlimbs and administered CTE or vehicle for 13 days. A sham procedure was performed in the mice of the fourth group to which the vehicle was administered. Next, the triceps surae muscles (TS) were excised. To analyze the effect of the novel cast system and CTE administration on muscle atrophy, we evaluated TS wet weight and myofiber cross-sectional area (CSA). We also determined MyHC IId/IIx expression levels by western blotting, since their increase is a hallmark of disuse muscle atrophy, suggesting slow-to-fast myofiber type shift. Moreover, we performed two tests of hindlimb performance. The novel cast immobilization method significantly reduced TS wet weight and myofiber CSA. This was accompanied by deterioration of hindlimb function and an increase in MyHC IId/IIx expression. CTE administration did not alter TS wet weight or myofiber CSA; however, it showed a trend of amelioration of the loss of hindlimb function and of suppression of the increased MyHC IId/IIx expression in cast-immobilized mice. Our novel hindlimb cast immobilization method effectively induced muscle atrophy. CTE did not affect muscle mass, but suppressed the shift from slow to fast myofiber type in cast-immobilized mice, ameliorating hindlimb function deterioration.
Collapse
|
166
|
Tran MP, Tsutsumi R, Erberich JM, Chen KD, Flores MD, Cooper KL. Evolutionary loss of foot muscle during development with characteristics of atrophy and no evidence of cell death. eLife 2019; 8:50645. [PMID: 31612857 PMCID: PMC6855805 DOI: 10.7554/elife.50645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 10/01/2019] [Indexed: 12/19/2022] Open
Abstract
Many species that run or leap across sparsely vegetated habitats, including horses and deer, evolved the severe reduction or complete loss of foot muscles as skeletal elements elongated and digits were lost, and yet the developmental mechanisms remain unknown. Here, we report the natural loss of foot muscles in the bipedal jerboa, Jaculus jaculus. Although adults have no muscles in their feet, newborn animals have muscles that rapidly disappear soon after birth. We were surprised to find no evidence of apoptotic or necrotic cell death during stages of peak myofiber loss, countering well-supported assumptions of developmental tissue remodeling. We instead see hallmarks of muscle atrophy, including an ordered disassembly of the sarcomere associated with upregulation of the E3 ubiquitin ligases, MuRF1 and Atrogin-1. We propose that the natural loss of muscle, which remodeled foot anatomy during evolution and development, involves cellular mechanisms that are typically associated with disease or injury. Intrinsic muscles are a group of muscles deep inside the hands and feet. They help to control the precise movements required, for example, for a pianist to play their instrument or for certain animals to climb with remarkable agility. Some animals, such as horses and deer, have evolved in such a way that they no longer grasp objects with hands and feet. Where intrinsic muscles were once present in the hands and feet of their ancestors, these animals now have strong ligaments that prevent over-extension of the wrist and ankle joints during hard landings. Given their size, it is difficult to study horses and deer in the laboratory and understand how they lost their intrinsic muscles during evolution. Tran et al. therefore focused on a small rodent called the lesser Egyptian jerboa, which also displays long legs with strong ligaments and no intrinsic muscles. Newborn jerboas have foot muscles that look very much like the intrinsic muscles found in mice, but these muscles disappear within 4 days of birth. A mechanism called programmed cell death is often responsible for specific tissues disappearing during development, but the experiments of Tran et al. revealed that this was not the case in jerboas. Instead, their intrinsic muscles were degraded by processes triggered by genes that disassemble underused muscles. In mice and humans, fasting, nerve injuries, or immobility trigger this type of muscle degradation, but in jerboas these processes appear to be a normal part of development. This unexpected discovery shows that development and disease-like processes are linked, and that more studies of nontraditional research animals may help scientists better understand these connections.
Collapse
Affiliation(s)
- Mai P Tran
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Rio Tsutsumi
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Joel M Erberich
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Kevin D Chen
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Michelle D Flores
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Kimberly L Cooper
- Division of Biological Sciences, Section of Cellular and Developmental Biology, University of California, San Diego, La Jolla, United States
| |
Collapse
|
167
|
Glutathione and Nitric Oxide: Key Team Players in Use and Disuse of Skeletal Muscle. Nutrients 2019; 11:nu11102318. [PMID: 31575008 PMCID: PMC6836164 DOI: 10.3390/nu11102318] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/11/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Glutathione (GSH) is the main non-enzymatic antioxidant playing an important role in detoxification, signal transduction by modulation of protein thiols redox status and direct scavenging of radicals. The latter function is not only performed against reactive oxygen species (ROS) but GSH also has a fundamental role in buffering nitric oxide (NO), a physiologically-produced molecule having-multifaceted functions. The efficient rate of GSH synthesis and high levels of GSH-dependent enzymes are characteristic features of healthy skeletal muscle where, besides the canonical functions, it is also involved in muscle contraction regulation. Moreover, NO production in skeletal muscle is a direct consequence of contractile activity and influences several metabolic myocyte pathways under both physiological and pathological conditions. In this review, we will consider the homeostasis and intersection of GSH with NO and then we will restrict the discussion on their role in processes related to skeletal muscle function and degeneration.
Collapse
|
168
|
Egawa T, Ohno Y, Yokoyama S, Yokokawa T, Tsuda S, Goto K, Hayashi T. The Protective Effect of Brazilian Propolis against Glycation Stress in Mouse Skeletal Muscle. Foods 2019; 8:E439. [PMID: 31557885 PMCID: PMC6836237 DOI: 10.3390/foods8100439] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/06/2023] Open
Abstract
We investigated the protective effect of Brazilian propolis, a natural resinous substance produced by honeybees, against glycation stress in mouse skeletal muscles. Mice were divided into four groups: (1) Normal diet + drinking water, (2) Brazilian propolis (0.1%)-containing diet + drinking water, (3) normal diet + methylglyoxal (MGO) (0.1%)-containing drinking water, and (4) Brazilian propolis (0.1%)-containing diet + MGO (0.1%)-containing drinking water. MGO treatment for 20 weeks reduced the weight of the extensor digitorum longus (EDL) muscle and tended to be in the soleus muscle. Ingestion of Brazilian propolis showed no effect on this change in EDL muscles but tended to increase the weight of the soleus muscles regardless of MGO treatment. In EDL muscles, Brazilian propolis ingestion suppressed the accumulation of MGO-derived advanced glycation end products (AGEs) in MGO-treated mice. The activity of glyoxalase 1 was not affected by MGO, but was enhanced by Brazilian propolis in EDL muscles. MGO treatment increased mRNA expression of inflammation-related molecules, interleukin (IL)-1β, IL-6, and toll-like receptor 4 (TLR4). Brazilian propolis ingestion suppressed these increases. MGO and/or propolis exerted no effect on the accumulation of AGEs, glyoxalase 1 activity, and inflammatory responses in soleus muscles. These results suggest that Brazilian propolis exerts a protective effect against glycation stress by inhibiting the accumulation of AGEs, promoting MGO detoxification, and reducing proinflammatory responses in the skeletal muscle. However, these anti-glycation effects does not lead to prevent glycation-induced muscle mass reduction.
Collapse
Affiliation(s)
- Tatsuro Egawa
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University, Kyoto 606-8501, Japan.
- Laboratory of Health and Exercise Sciences, Graduate School of Human and Environmental Studies, Kyoto University, Kyoto 606-8501, Japan.
| | - Yoshitaka Ohno
- Laboratory of Physiology, School of Health Sciences, Toyohashi SOZO University, Toyohashi 440-8511, Japan.
| | - Shingo Yokoyama
- Laboratory of Physiology, School of Health Sciences, Toyohashi SOZO University, Toyohashi 440-8511, Japan.
| | - Takumi Yokokawa
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University, Kyoto 606-8501, Japan.
| | - Satoshi Tsuda
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University, Kyoto 606-8501, Japan.
| | - Katsumasa Goto
- Laboratory of Physiology, School of Health Sciences, Toyohashi SOZO University, Toyohashi 440-8511, Japan.
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Toyohashi 440-8511, Japan.
| | - Tatsuya Hayashi
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
169
|
Abstract
PURPOSE OF REVIEW Cachexia is defined as ongoing loss of skeletal muscle mass, with or without depletion of adipose tissue and is a common syndrome in cancer patients, affecting 50% of those diagnosed. Cachexia, which cannot be fully reversed and causes significant functional impairment is caused by various mechanisms such as an altered energy balance and disruption of homeostatic control by the central nervous system. This central nervous system deregulation involves hypothalamic pituitary adrenal (HPA) axis stimulation, which can be triggered by IL-1R1 engagement on neuronal processes and endothelium in the microvasculature of the hypothalamus. This review will explore current evidence regarding both the importance of IL-1α in the various components of cancer cachexia and its potential as a therapeutic target. RECENT FINDINGS IL-1α, which signals through IL-1R1, has been identified as a key agonist in the IL-1 pathway. As such, IL-1α has been explored as a therapeutic target in cancer cachexia, leading to the development of bermekimab, a mAb which neutralizes IL-1α. With a limited array of medication currently available to treat cancer cachexia, bermekimab represents a possible therapy. SUMMARY IL-1α is a key mediator in cachexia development and targeting this may be a viable therapeutic target.
Collapse
|
170
|
Silva WJ, Graça FA, Cruz A, Silvestre JG, Labeit S, Miyabara EH, Yan CYI, Wang DZ, Moriscot AS. miR-29c improves skeletal muscle mass and function throughout myocyte proliferation and differentiation and by repressing atrophy-related genes. Acta Physiol (Oxf) 2019; 226:e13278. [PMID: 30943315 PMCID: PMC6900115 DOI: 10.1111/apha.13278] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 03/02/2019] [Accepted: 03/31/2019] [Indexed: 12/25/2022]
Abstract
AIM To identify microRNAs (miRs) involved in the regulation of skeletal muscle mass. For that purpose, we have initially utilized an in silico analysis, resulting in the identification of miR-29c as a positive regulator of muscle mass. METHODS miR-29c was electrotransferred to the tibialis anterior to address its morphometric and functional properties and to determine the level of satellite cell proliferation and differentiation. qPCR was used to investigate the effect of miR-29c overexpression on trophicity-related genes. C2C12 cells were used to determine the impact of miR-29c on myogenesis and a luciferase reporter assay was used to evaluate the ability of miR-29c to bind to the MuRF1 3'UTR. RESULTS The overexpression of miR-29c in the tibialis anterior increased muscle mass by 40%, with a corresponding increase in fibre cross-sectional area and force and a 30% increase in length. In addition, satellite cell proliferation and differentiation were increased. In C2C12 cells, miR-29c oligonucleotides caused increased levels of differentiation, as evidenced by an increase in eMHC immunostaining and the myotube fusion index. Accordingly, the mRNA levels of myogenic markers were also increased. Mechanistically, the overexpression of miR-29c inhibited the expression of the muscle atrophic factors MuRF1, Atrogin-1 and HDAC4. For the key atrogene MuRF1, we found that miR-29c can bind to its 3'UTR to mediate repression. CONCLUSIONS The results herein suggest that miR-29c can improve skeletal muscle size and function by stimulating satellite cell proliferation and repressing atrophy-related genes. Taken together, our results indicate that miR-29c might be useful as a future therapeutic device in diseases involving decreased skeletal muscle mass.
Collapse
Affiliation(s)
- William José Silva
- Department of Anatomy, Institute of Biomedical SciencesUniversity of Sao PauloSao PauloBrazil
| | - Flavia Aparecida Graça
- Department of Anatomy, Institute of Biomedical SciencesUniversity of Sao PauloSao PauloBrazil
| | - André Cruz
- Department of Anatomy, Institute of Biomedical SciencesUniversity of Sao PauloSao PauloBrazil
| | | | - Siegfried Labeit
- Faculty for Clinical Medicine Mannheim of the University of HeidelbergInstitute for Integrative Pathophysiology, Universitätsmedizin MannheimMannheimGermany
| | - Elen Haruka Miyabara
- Department of Anatomy, Institute of Biomedical SciencesUniversity of Sao PauloSao PauloBrazil
| | - Chao Yun Irene Yan
- Department of Cell Biology, Institute of biomedical SciencesUniversity of Sao PauloSao PauloBrazil
| | - Da Zhi Wang
- Department of CardiologyBoston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Anselmo Sigari Moriscot
- Department of Anatomy, Institute of Biomedical SciencesUniversity of Sao PauloSao PauloBrazil
| |
Collapse
|
171
|
Ramasamy DK, Dutta T, Kannan V, Chandramouleeswaran V. Amino acids in post-stroke rehabilitation. Nutr Neurosci 2019; 24:426-431. [PMID: 31328694 DOI: 10.1080/1028415x.2019.1641295] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Objectives: Patients with stroke are prone to disability due to muscle hypercatabolism. We aim to review this concept based on available data on benefits of amino acid supplementation in post-stroke rehabilitation.Method: The search was performed on Medline and Embase in January 2019. Randomized controlled studies, observational studies and case reports conducted in the last 15 years on the supplementation of amino acids in post-stroke rehabilitation patients were included.Result: Amino acids prevent muscle hypercatabolism in post stroke patients by suppressing myofibrillar protein and skeletal muscle degradation. Stroke patients supplemented with amino acids led to an improvement of functional and physical performance.Discussion: Muscle protein hypercatabolism and sequestration of amino acids from skeletal muscles occur cyclically in post-stroke patients to counter each other. There is a resultant deficit of amino acids which is unmet. Amino acids have antiproteolytic effect. Its supplementation prevents muscle wasting and improves rehabilitation by promoting physical performance, muscle strength, mass, and function.
Collapse
Affiliation(s)
| | - Trayambak Dutta
- Department of Medical & Scientific Affairs, Tablets India Ltd., Chennai, India
| | | | | |
Collapse
|
172
|
Wu C, Tang L, Ni X, Xu T, Fang Q, Xu L, Ma W, Yang X, Sun H. Salidroside Attenuates Denervation-Induced Skeletal Muscle Atrophy Through Negative Regulation of Pro-inflammatory Cytokine. Front Physiol 2019; 10:665. [PMID: 31293430 PMCID: PMC6604664 DOI: 10.3389/fphys.2019.00665] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/09/2019] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle atrophy is associated with pro-inflammatory cytokines. Salidroside is a biologically active ingredient of Rhodiola rosea, which exhibits anti-inflammatory property. However, there is little known about the effect of salidroside on denervation-induced muscle atrophy. Therefore, the present study aimed to determine whether salidroside could protect against denervation-induced muscle atrophy and to clarify potential molecular mechanisms. Denervation caused progressive accumulation of inflammatory factors in skeletal muscle, especially interleukin 6 (IL6) and its receptor, and recombinant murine IL6 (rmIL6) local infusion could induce target muscle atrophy, suggesting that denervation induced inflammation in target muscles and the inflammation may trigger muscle wasting. Salidroside alleviated denervation-induced muscle atrophy and inhibited the production of IL6. Furthermore, the inhibition of phosphorylation of signal transducer and activator of transcription 3 (STAT3), and the decreased levels of suppressor of cytokine signaling (SOCS3), muscle RING finger protein-1 (MuRF1), atrophy F-box (atrogin-1), microtubule-associated protein light chain 3 beta (LC3B) and PTEN-induced putative kinase (PINK1) were observed in denervated muscles that were treated with salidroside. Finally, all of these responses to salidroside were replicated in neutralizing antibody against IL6. Taken together, these results suggest that salidroside alleviates denervation-induced inflammation response, thereby inhibits muscle proteolysis and muscle atrophy. Therefore, it was assumed that salidroside might be a potential therapeutic candidate to prevent muscle wasting.
Collapse
Affiliation(s)
- Changyue Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,School of Medicine, Nantong University, Nantong, China
| | - Longhai Tang
- Departments of Blood Component Preparation, Suzhou Blood Center, Suzhou, China
| | - Xuejun Ni
- Departments of Ultrasound, Affiliated Hospital of Nantong University, Nantong, China
| | - Tongtong Xu
- School of Medicine, Nantong University, Nantong, China
| | - Qingqing Fang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Lai Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Wenjing Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
173
|
Smith L, Yang L, Hamer M. Handgrip strength, inflammatory markers, and mortality. Scand J Med Sci Sports 2019; 29:1190-1196. [PMID: 30972827 DOI: 10.1111/sms.13433] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/22/2019] [Accepted: 04/04/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE To investigate the extent to which inflammatory markers explain the association between handgrip strength and mortality. METHODS Analyses of data from The English Longitudinal Study of Ageing. Handgrip strength and inflammatory marker data (C-reactive protein and fibrinogen) were collected at baseline (2004/5) and inflammatory marker data at follow-up (2012/13). Participant data were linked with death records. General linear models were used to explore associations between handgrip strength and inflammatory markers at follow-up. Cox proportional hazards regression models were used to examine associations between grip strength and risk of death. Models were estimated with the covariates age, sex, wealth, physical activity, smoking, depressive symptoms, long-standing illness, and adiposity. RESULTS The sample comprised of 5,240 participants (mean age 65.9 (SD 9.4) years; 53.8% female). Over an average 9.7 ± 1.4 years follow-up, there were 650 deaths. Inverse associations were evident between handgrip strength and change in inflammatory markers in women only. There was an association between grip strength and lower risk of mortality in women (hazard ratio = 0.85; 95% CI, 0.74, 0.98) after adjusting for age and wealth. The association was attenuated after adjustment for clinical and behavioral risk factors (0.92; 0.79, 1.07), and further attenuated after adjusting for inflammatory markers (0.95; 0.82, 1.11). CONCLUSION Higher grip strength is associated with lower levels of inflammation at 8-year follow-up, and inflammatory markers partly explained the association between handgrip strength and mortality.
Collapse
Affiliation(s)
- Lee Smith
- The Cambridge Centre for Sport and Exercise Sciences, Anglia Ruskin University, Cambridge, UK
| | - Lin Yang
- Department of Epidemiology, Medical University of Vienna, Vienna, Austria
| | - Mark Hamer
- School Sport, Exercise Health Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
174
|
Goubert D, Meeus M, Willems T, De Pauw R, Coppieters I, Crombez G, Danneels L. The association between back muscle characteristics and pressure pain sensitivity in low back pain patients. Scand J Pain 2019; 18:281-293. [PMID: 29794309 DOI: 10.1515/sjpain-2017-0142] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 02/24/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS Some low back pain (LBP) patients recover after every pain episode whereas others develop chronicity. Research indicates that the amount of atrophy and fat infiltration differs between patients with LBP. Also enhanced pain sensitivity is present only in a subgroup of LBP patients. The relationship between pain sensitivity and muscular deformations in LBP, is however unexplored. This study examined the association between pressure pain sensitivity and the structural characteristics of the lumbar muscles in three different groups of non-specific LBP patients. METHODS This cross-sectional study examined the total cross-sectional area (CSA), fat CSA, muscle CSA and muscle fat index (MFI) of the lumbar multifidus (MF) and erector spinae (ES) at level L4 by magnetic resonance imaging in 54 patients with non-specific LBP (23 recurrent LBP, 15 non-continuous chronic LBP and 16 continuous chronic LBP). Pressure pain thresholds were measured at four locations (lower back, neck, hand and leg) by a manual pressure algometer and combined into one "pain sensitivity" variable. As a primary outcome measure, the association between pain sensitivity and muscle structure characteristics was investigated by multiple independent general linear regression models. Secondly, the influence of body mass index (BMI) and age on muscle characteristics was examined. RESULTS A positive association was found between pain sensitivity and the total CSA of the MF (p=0.006) and ES (p=0.001), and the muscle CSA of the MF (p=0.003) and ES (p=0.001), irrespective of the LBP group. No association was found between pain sensitivity and fat CSA or MFI (p>0.01). Furthermore, a positive association was found between BMI and the fat CSA of the MF (p=0.004) and ES (p=0.006), and the MFI of the MF (p<0.01) and ES (p=0.003). Finally, a positive association was found between age with the fat CSA of the MF (p=0.008) but not with the fat CSA of the ES (p>0.01), nor the MFI of the MF (p>0.01) and ES (p>0.01). CONCLUSIONS A higher pain sensitivity is associated with a smaller total and muscle CSA in the lumbar MF and ES, and vice versa, but results are independent from the LBP subgroup. On the other hand, the amount of fat infiltration in the lumbar muscles is not associated with pain sensitivity. Instead, a higher BMI is associated with more lumbar fat infiltration. Finally, older patients with LBP are associated with higher fat infiltration in the MF but not in the ES muscle. IMPLICATIONS These results imply that reconditioning muscular tissues might possibly decrease the pain sensitivity of LBP patients. Vice versa, therapy focusing on enhancement of pain sensitivity might also positively influence the CSA and that way contribute to the recovery of LBP. Furthermore, the amount of lumbar muscle fat seems not susceptible to pain sensitivity or vice versa, but instead a decrease in BMI might decrease the fat infiltration in the lumbar muscles and therefore improve the muscle structure quality in LBP. These hypothesis apply for all non-specific LBP patients, despite the type of LBP.
Collapse
Affiliation(s)
- Dorien Goubert
- Department of Rehabilitation Sciences and Physiotherapy, Faculty of Medicine and Health Sciences, Ghent University, Ghent Campus Heymans (UZ) 3 B3, Corneel Heymanslaan 10, Ghent, Belgium, Phone: +3293325374, Fax: +32 9 332 38 11; andPain in Motion International Research Group
| | - Mira Meeus
- Department of Rehabilitation Sciences and Physiotherapy, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium;Pain in Motion International Research Group.,Department of Rehabilitation Sciences and Physiotherapy, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Tine Willems
- Department of Rehabilitation Sciences and Physiotherapy, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Robby De Pauw
- Department of Physiotherapy, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Iris Coppieters
- Department of Physiotherapy, Human Physiology and Anatomy (KIMA), Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium; andPain in Motion International Research Group
| | - Geert Crombez
- Department of Experimental-Clinical and Health Psychology, Faculty of Psychology and Educational Sciences, Ghent University, Ghent, Belgium
| | - Lieven Danneels
- Department of Rehabilitation Sciences and Physiotherapy, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
175
|
Loh KP, Tooze JA, Nicklas BJ, Kritchevsky SB, Williamson JD, Ellis LR, Powell BL, Pardee TS, Goyal NG, Klepin HD. Inflammatory biomarkers, geriatric assessment, and treatment outcomes in acute myeloid leukemia. J Geriatr Oncol 2019; 11:410-416. [PMID: 30962090 DOI: 10.1016/j.jgo.2019.03.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 11/18/2022]
Abstract
OBJECTIVES To investigate changes in inflammatory biomarkers during induction therapy for older adults with acute myeloid leukemia (AML) and their associations with geriatric assessment (GA) measures and outcomes. METHODS This was a single institution ancillary study to a prospective observational study (N = 20 consecutive adults aged ≥60 with newly diagnosed AML who received induction chemotherapy). Biomarkers (Interleukin-6 [IL-6], IL-6 soluble receptor [IL-6 sR], tumor necrosis factor alpha [TNFα], TNFα soluble receptor 1 [TNFα sR1], interleukin-3 [IL-3], C-reactive protein [CRP]) were collected at start of induction, weekly for three weeks, and post-induction and were compared over time using paired t-tests. GA was administered at baseline and post-induction, and correlated with biomarker levels using Spearman correlations. Survival was estimated using Kaplan-Meier and compared by categorized biomarker level using Wilcoxon tests. RESULTS Biomarker levels were stable during induction, except for CRP and IL-6 sR. Declines in objectively measured physical function [Short Physical Performance Battery (SPPB); r = 0.71, p < 0.01] and increases in self-reported limitation in instrumental activities of daily living (r = 0.81, p < 0.01) were correlated with increased TNFα sR1. Declines in SPPB were correlated with increased CRP (r = -0.73, p < 0.01). Improvement in depression was correlated with increased IL-6 sR (r = -0.59 p = 0.02). Survival was shorter in those with baseline TNFα or CRP levels above the median (6.1 vs. 40.2 months and 5.5 vs. 27.6 months respectively, p = 0.04 for both). CONCLUSION Among older adults with AML, the relationships between TNFα sR1, CRP, and IL-6 sR with change in physical and emotional health during treatment warrants further investigation.
Collapse
Affiliation(s)
- Kah Poh Loh
- Division of Hematology/Oncology, James P. Wilmot Cancer Institute, University of Rochester/Strong Memorial Hospital, Rochester, NY, United States.
| | - Janet A Tooze
- Division of Public Health Sciences, Wake Forest Baptist Health, Winston-Salem, NC, United States
| | - Barbara J Nicklas
- Section Gerontology and Geriatric Medicine, Wake Forest Baptist Health, Winston-Salem, NC, United States
| | - Stephen B Kritchevsky
- Section Gerontology and Geriatric Medicine, Wake Forest Baptist Health, Winston-Salem, NC, United States
| | - Jeff D Williamson
- Section Gerontology and Geriatric Medicine, Wake Forest Baptist Health, Winston-Salem, NC, United States
| | - Leslie R Ellis
- Section on Hematology and Oncology, Wake Forest Baptist Health, Winston-Salem, NC, United States
| | - Bayard L Powell
- Section on Hematology and Oncology, Wake Forest Baptist Health, Winston-Salem, NC, United States
| | - Timothy S Pardee
- Section on Hematology and Oncology, Wake Forest Baptist Health, Winston-Salem, NC, United States
| | - Neha G Goyal
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, United States
| | - Heidi D Klepin
- Section on Hematology and Oncology, Wake Forest Baptist Health, Winston-Salem, NC, United States
| |
Collapse
|
176
|
Oh SSJ, Kim S, Moon S, Park DH, Kang JH. Lactate Overload Inhibits Myogenic Activity in C2C12 Myotubes. Open Life Sci 2019; 14:29-37. [PMID: 33817134 PMCID: PMC7874767 DOI: 10.1515/biol-2019-0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/10/2018] [Indexed: 01/09/2023] Open
Abstract
Lactate (LA), an endogenous metabolite produced from pyruvate, can accumulate in skeletal muscle in certain conditions including major diseases, as well as during intensive exercise. Using differentiated C2C12 myotubes, we evaluated the early (1-h) and delayed (24-h) effects of LA (8 mM) on mechanisms involved in myogenesis or muscle atrophy, including 5'-adenosine monophosphate-activated protein kinase (AMPK)-mediated inhibition of protein synthesis through the mTOR/P70-S6K pathway, Akt-mediated inhibition of expression of the MAFbx atrophic factor by FOXO3a and expression of the myogenic transcription factors, MyoD, myogenin and myosin heavy chain. Although the early effects of LA overload were not significant on myogenic or atrophic mechanisms, LA treatment for 24 h significantly activated atrophic mechanisms but suppressed myogenesis in myotubes. In addition, LA overload for 24 h significantly suppressed the expression of Sirtuin 1 and peroxisome proliferator-activated receptor gamma coactivator-1 alpha. Consistent with LA-induced activation of atrophic mechanisms, the diameter of C2C12 myotubes treated with LA for 24 h, but not for 1 h, was significantly lower than in control myotubes. Thus, a sustained, but not a transient, LA overload could induce muscle atrophy through the regulation of AMPK- and Akt-mediated pathways, although further in vivo studies are needed to confirm this.
Collapse
Affiliation(s)
| | - Sujin Kim
- Department of Kinesiology, Inha University Incheon, South Korea.,Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Sohee Moon
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Dong-Ho Park
- Department of Kinesiology, Inha University, 100, Inha-Ro, Nam-gu, Incheon 22212, Republic of Korea
| | - Ju-Hee Kang
- Department of Pharmacology and Medicinal Toxicology Research Center, Hypoxia-related Diseases Research Center, Inha University School of Medicine, Room 1015, 60th Anniversary Hall, 100, Inha-ro, Nam-gu, Incheon 22212, Republic of Korea
| |
Collapse
|
177
|
Creatine supplementation in Walker-256 tumor-bearing rats prevents skeletal muscle atrophy by attenuating systemic inflammation and protein degradation signaling. Eur J Nutr 2019; 59:661-669. [PMID: 30806774 DOI: 10.1007/s00394-019-01933-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/16/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE The aim of this study was to investigate the effects of creatine supplementation on muscle wasting in Walker-256 tumor-bearing rats. METHODS Wistar rats were randomly assigned into three groups (n = 10/group): control (C), tumor bearing (T), and tumor bearing supplemented with creatine (TCr). Creatine was provided in drinking water for a total of 21 days. After 11 days of supplementation, tumor cells were implanted subcutaneously into T and TCr groups. The animals' weight, food and water intake were evaluated along the experimental protocol. After 10 days of tumor implantation (21 total), animals were euthanized for inflammatory state and skeletal muscle cross-sectional area measurements. Skeletal muscle components of ubiquitin-proteasome pathways were also evaluated using real-time PCR and immunoblotting. RESULTS The results showed that creatine supplementation protected tumor-bearing rats against body weight loss and skeletal muscle atrophy. Creatine intake promoted lower levels of plasma TNF-α and IL-6 and smaller spleen morphology changes such as reduced size of white pulp and lymphoid follicle compared to tumor-bearing rats. In addition, creatine prevented increased levels of skeletal muscle Atrogin-1 and MuRF-1, key regulators of muscle atrophy. CONCLUSION Creatine supplementation prevents skeletal muscle atrophy by attenuating tumor-induced pro-inflammatory environment, a condition that minimizes Atrogin-1 and MuRF-1-dependent proteolysis.
Collapse
|
178
|
Limongi D, Baldelli S, Checconi P, Marcocci ME, De Chiara G, Fraternale A, Magnani M, Ciriolo MR, Palamara AT. GSH-C4 Acts as Anti-inflammatory Drug in Different Models of Canonical and Cell Autonomous Inflammation Through NFκB Inhibition. Front Immunol 2019; 10:155. [PMID: 30787932 PMCID: PMC6372722 DOI: 10.3389/fimmu.2019.00155] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 01/17/2019] [Indexed: 01/06/2023] Open
Abstract
An imbalance in GSH/GSSG ratio represents a triggering event in pro-inflammatory cytokine production and inflammatory response. However, the molecular mechanism(s) through which GSH regulates macrophage and cell autonomous inflammation remains not deeply understood. Here, we investigated the effects of a derivative of GSH, the N-butanoyl glutathione (GSH-C4), a cell permeable compound, on lipopolisaccharide (LPS)-stimulated murine RAW 264.7 macrophages, and human macrophages. LPS alone induces a significant production of pro-inflammatory cytokines, such as IL-1β, IL-6, and TNF-α and a significant decrement of GSH content. Such events were significantly abrogated by treatment with GSH-C4. Moreover, GSH-C4 was highly efficient in buffering cell autonomous inflammatory status of aged C2C12 myotubes and 3T3-L1 adipocytes by suppressing the production of pro-inflammatory cytokines. We found that inflammation was paralleled by a strong induction of the phosphorylated form of NFκB, which translocates into the nucleus; a process that was also efficiently inhibited by the treatment with GSH-C4. Overall, the evidence suggests that GSH decrement is required for efficient activation of an inflammatory condition and, at the same time, GSH-C4 can be envisaged as a good candidate to abrogate such process, expanding the anti-inflammatory role of this molecule in chronic inflammatory states.
Collapse
Affiliation(s)
- Dolores Limongi
- Department of Human Sciences and Promotion of the Quality of Life, IRCCS San Raffaele Pisana, San Raffaele Roma Open University, Rome, Italy
| | - Sara Baldelli
- Department of Human Sciences and Promotion of the Quality of Life, IRCCS San Raffaele Pisana, San Raffaele Roma Open University, Rome, Italy
| | - Paola Checconi
- Department of Human Sciences and Promotion of the Quality of Life, IRCCS San Raffaele Pisana, San Raffaele Roma Open University, Rome, Italy
| | - Maria Elena Marcocci
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Giovanna De Chiara
- Institute of Translational Pharmacology, National Research Council Rome, Rome, Italy
| | | | - Mauro Magnani
- University of Urbino Carlo Bo, Department of Biomolecular Sciences, Urbino, Italy
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy.,IRCCS San Raffaele Pisana, Rome, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.,IRCCS San Raffaele Pisana, Rome, Italy.,Institute Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
179
|
Hatabe Y, Shibata M, Ohara T, Oishi E, Yoshida D, Honda T, Hata J, Kanba S, Kitazono T, Ninomiya T. Decline in Handgrip Strength From Midlife to Late-Life is Associated With Dementia in a Japanese Community: The Hisayama Study. J Epidemiol 2018; 30:15-23. [PMID: 30531122 PMCID: PMC6908846 DOI: 10.2188/jea.je20180137] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The association between decline in handgrip strength from midlife to late life and dementia is unclear. METHODS Japanese community-dwellers without dementia aged 60 to 79 years (ie, individuals in late life; mean age, 68 years) were followed for 24 years (1988-2012) (n = 1,055); 835 of them had participated in a health examination in 1973-1974 (mean age, 53 years), and these earlier data were used for the midlife analysis. Using a Cox proportional hazards model, we estimated the risk conferred by a decline in handgrip strength over a 15-year period (1973-74 to 1988) from midlife to late life on the development of total dementia, Alzheimer's disease (AD), and vascular dementia (VaD) over the late-life follow-up period from 1988 to 2012. RESULTS During the follow-up, 368 subjects experienced total dementia. The age- and sex-adjusted incidence of total dementia increased significantly with greater decline in handgrip strength (increased or unchanged handgrip strength [≥+0%] 25.1, mildly decreased [-14 to -1%] 28.4, and severely decreased [≤-15%] 38.9 per 1,000 person-years). A greater decline in handgrip strength was significantly associated with higher risk of total dementia after adjusting for potential confounding factors; subjects with severely decreased handgrip strength had 1.51-fold (95% confidence interval, 1.14-1.99, P < 0.01) increased risk of total dementia compared to those with increased or unchanged handgrip strength. Similar significant findings were observed for AD, but not for VaD. CONCLUSIONS Our findings suggest that a greater decline in handgrip strength from midlife to late life is an important indicator for late-life onset of dementia.
Collapse
Affiliation(s)
- Yozo Hatabe
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University.,Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University
| | - Mao Shibata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University
| | - Tomoyuki Ohara
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University.,Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University
| | - Emi Oishi
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University
| | - Daigo Yoshida
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University
| | - Takanori Honda
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University
| | - Jun Hata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University
| | - Shigenobu Kanba
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University
| | - Takanari Kitazono
- Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University
| |
Collapse
|
180
|
Fernández-Nocelo S, Gallego R, Costoya JA, Arce VM. Expression of myostatin in human hematopoietic cells unveils novel autocrine/paracrine actions for the hormone. J Cell Physiol 2018; 234:7236-7246. [PMID: 30370618 DOI: 10.1002/jcp.27494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 09/05/2018] [Indexed: 12/15/2022]
Abstract
Myostatin is a member of the transforming growth factor β (TGFβ) superfamily that has a well-established role as a mediator of muscle growth and development. However, myostatin is now emerging as a pleiotropic hormone with multiple actions in the regulation of the metabolism as well as several aspects of both cardiac and smooth muscle cells physiology. In addition, myostatin is also expressed in several nonmuscular cells where its physiological role remains to be elucidated in most cases. In this report, we have shown that both myostatin and its receptor system are expressed in blood cells and in hematopoietic cell lines. Furthermore, myostatin treatment promotes differentiation of both HL60 and K562 cells through a mechanism that involves activation of extracellular signal-regulated kinases 1/2 and p38-mitogen-activated protein kinase, thus leading to the possibility that myostatin may be a paracrine/autocrine factor involved in the control of haematopoiesis. In addition, the presence of myostatin expression in immune cells could envisage a novel role for the hormone in the pathogenesis of inflammatory diseases.
Collapse
Affiliation(s)
- Susana Fernández-Nocelo
- Departamento de Fisioloxía, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Rosalía Gallego
- Departamento de Ciencias Morfolóxicas, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - José A Costoya
- Departamento de Fisioloxía, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,CIMUS, Universidade de Santiago de Compostela and Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Víctor M Arce
- Departamento de Fisioloxía, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,CIMUS, Universidade de Santiago de Compostela and Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
181
|
Marrelli K, Cheng AJ, Brophy JD, Power GA. Perceived Versus Performance Fatigability in Patients With Rheumatoid Arthritis. Front Physiol 2018; 9:1395. [PMID: 30364087 PMCID: PMC6191483 DOI: 10.3389/fphys.2018.01395] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/13/2018] [Indexed: 12/17/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, inflammatory disease that affects 1% of the general population. Fatigue is a common complaint of patients with RA, however their perceived fatigue may be more exacerbated than objective measures of fatigue may indicate. The assessment of fatigue is made complex due to inconsistent and vague terms used to define fatigue, and the task dependence of fatigability. Fatigue is defined as a state of exhaustion and decreased strength, while fatigability indicates an individual's susceptibility to fatigue. In order to offer some clarity to the manifestation of fatigue in clinical populations, in this review we outline that fatigue should be described with subsections that are related to the symptom, such as: perceived fatigability and performance fatigability. Where perceived fatigability indicates the subjective state of the individual and thus involves the individual's subjective measure of fatigue, performance fatigability would be measured through clinical and laboratory-based assessments that quantify the functional decline in performance. This review describes RA and the various neuromuscular changes associated with the disease that can lead to alterations in both perceived and performance fatigue. From there, we discuss fatigue and RA, how fatigue can be assessed, effects of exercise interventions on RA symptoms and fatigue, and recommendations for future studies investigating subjective and objective measures of fatigability.
Collapse
Affiliation(s)
- Kristina Marrelli
- Department of Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, ON, Canada
| | - Arthur J. Cheng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Julie D. Brophy
- Community Rheumatologist, Wellington Ortho and Rehab, Guelph, ON, Canada
| | - Geoffrey A. Power
- Department of Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
182
|
Majumder A, Singh M, Behera J, Theilen NT, George AK, Tyagi N, Metreveli N, Tyagi SC. Hydrogen sulfide alleviates hyperhomocysteinemia-mediated skeletal muscle atrophy via mitigation of oxidative and endoplasmic reticulum stress injury. Am J Physiol Cell Physiol 2018; 315:C609-C622. [PMID: 30110564 DOI: 10.1152/ajpcell.00147.2018] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although hyperhomocysteinemia (HHcy) occurs because of the deficiency in cystathionine-β-synthase (CBS) causing skeletal muscle dysfunction, it is still unclear whether this effect is mediated through oxidative stress, endoplasmic reticulum (ER) stress, or both. Nevertheless, there is no treatment option available to improve HHcy-mediated muscle injury. Hydrogen sulfide (H2S) is an antioxidant compound, and patients with CBS mutation do not produce H2S. In this study, we hypothesized that H2S mitigates HHcy-induced redox imbalance/ER stress during skeletal muscle atrophy via JNK phosphorylation. We used CBS+/- mice to study HHcy-mediated muscle atrophy, and treated them with sodium hydrogen sulfide (NaHS; an H2S donor). Proteins and mRNAs were examined by Western blots and quantitative PCR. Proinflammatory cytokines were also measured. Muscle mass and strength were studied via fatigue susceptibility test. Our data revealed that HHcy was detrimental to skeletal mass, particularly gastrocnemius and quadriceps muscle weight. We noticed that oxidative stress was reversed by NaHS in homocysteine (Hcy)-treated C2C12 cells. Interestingly, ER stress markers (GRP78, ATF6, pIRE1α, and pJNK) were elevated in vivo and in vitro, and NaHS mitigated these effects. Additionally, we observed that JNK phosphorylation was upregulated in C2C12 after Hcy treatment, but NaHS could not reduce this effect. Furthermore, inflammatory cytokines IL-6 and TNF-α were higher in plasma from CBS as compared with wild-type mice. FOXO1-mediated Atrogin-1 and MuRF-1 upregulation were attenuated by NaHS. Functional studies revealed that NaHS administration improved muscle fatigability in CBS+/- mice. In conclusion, our work provides evidence that NaHS is beneficial in mitigating HHcy-mediated skeletal injury incited by oxidative/ER stress responses.
Collapse
Affiliation(s)
- Avisek Majumder
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky.,Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine , Louisville, Kentucky
| | - Mahavir Singh
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Jyotirmaya Behera
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Nicholas T Theilen
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Akash K George
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Neetu Tyagi
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Naira Metreveli
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| |
Collapse
|
183
|
Mundi MS, Patel J, McClave SA, Hurt RT. Current perspective for tube feeding in the elderly: from identifying malnutrition to providing of enteral nutrition. Clin Interv Aging 2018; 13:1353-1364. [PMID: 30122907 PMCID: PMC6080667 DOI: 10.2147/cia.s134919] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
With the number of individuals older than 65 years expected to rise significantly over the next few decades, dramatic changes to our society and health care system will need to take place to meet their needs. Age-related changes in muscle mass and body composition along with medical comorbidities including stroke, dementia, and depression place elderly adults at high risk for developing malnutrition and frailty. This loss of function and decline in muscle mass (ie, sarcopenia) can be associated with reduced mobility and ability to perform the task of daily living, placing the elderly at an increased risk for falls, fractures, and subsequent institutionalization, leading to a decline in the quality of life and increased mortality. There are a number of modifiable factors that can mitigate some of the muscle loss elderly experience especially when hospitalized. Due to this, it is paramount for providers to understand the pathophysiology behind malnutrition and sarcopenia, be able to assess risk factors for malnutrition, and provide appropriate nutrition support. The present review describes the pathophysiology of malnutrition, identifies contributing factors to this condition, discusses tools to assess nutritional status, and proposes key strategies for optimizing enteral nutrition therapy for the elderly.
Collapse
Affiliation(s)
- Manpreet S Mundi
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN, USA,
| | - Jayshil Patel
- Division of Pulmonary, Critical Care & Sleep Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Stephen A McClave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY, USA
| | - Ryan T Hurt
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY, USA.,Division of General Internal Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
184
|
van Niekerk G, Engelbrecht AM. Inflammation-induced metabolic derangements or adaptation: An immunometabolic perspective. Cytokine Growth Factor Rev 2018; 43:47-53. [PMID: 29970338 DOI: 10.1016/j.cytogfr.2018.06.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 02/08/2023]
Abstract
Inflammatory mediators have a well-established role in mediating metabolic disturbances. Chronic low-grade inflammation is implicated in the pathogenesis of obesity and the development of metabolic syndrome. This phenomenon is even more pronounced in severe inflammatory states such as in critically ill patients where hyperglycaemia invariably manifests. Similarly, though inflammatory mediators have a well-established role in promoting bone resorption, the adaptive function of this process remains unknown. Here we review emerging evidence from the field of immunometabolism suggesting that these two processes serve a common goal, namely, to sustain the rapid proliferation of immune cells during an infection. Activated immune cells exhibit an increased demand for glucose which not only provides energy, but also glycolytic intermediates which are fluxed into biosynthetic processes. Similarly, phosphate liberated from bone is consumed during the phosphorylation of glycolytic intermediates, which plays a critical role in the synthesis of nucleotides and phospholipids. Taken together, these considerations suggest that metabolic alterations induced by inflammatory mediators do not manifest as an inability to maintain homeostatic levels of metabolites but represent an adaptive shift in the homeostatic set point during an infection.
Collapse
Affiliation(s)
- Gustav van Niekerk
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa.
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
185
|
Pedler A, McMahon K, Galloway G, Durbridge G, Sterling M. Intramuscular fat is present in cervical multifidus but not soleus in patients with chronic whiplash associated disorders. PLoS One 2018; 13:e0197438. [PMID: 29795590 PMCID: PMC5967697 DOI: 10.1371/journal.pone.0197438] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 05/02/2018] [Indexed: 12/17/2022] Open
Abstract
The presence of intramuscular fat (IMF) in the cervical spine muscles of patients with whiplash associated disorders (WAD) has been consistently found. The mechanisms underlying IMF are not clear but preliminary evidence implicates a relationship with stress system responses. We hypothesised that if systemic stress system responses do play a role then IMF would be present in muscles remote to the cervical spine. We aimed to investigate if IMF are present in muscle tissue remote (soleus) to the cervical spine in people with chronic WAD. A secondary aim was to investigate associations between IMF and posttraumatic stress symptom levels. Forty-three people with chronic WAD (25 female) and 16 asymptomatic control participants (11 female) participated. Measures of pain, disability and posttraumatic stress symptoms were collected from the WAD participants. Both groups underwent MRI measures of IMF in cervical multifidus and the right soleus muscle. There was significantly greater IMF in cervical multifidus in patients with WAD and moderate/severe disability compared to controls (p = 0.009). There was no difference in multifidus IMF between the mild and moderate/severe disability WAD groups (p = 0.64), or the control and mild WAD groups (p = 0.21). IMF in the right soleus was not different between the groups (p = 0.47). In the WAD group, we found no correlation between PDS symptoms and cervical multifidus IMF or between PDS symptoms and soleus IMF. Global differences in IMF are not a feature of chronic WAD, with differences in IMF limited to the cervical spine musculature. While the mechanisms for the development of IMF in the cervical spine following whiplash injury remain unclear, our data indicate that local factors more likely contribute to these differences.
Collapse
Affiliation(s)
- Ashley Pedler
- Recover Injury Research Centre, NHMRC Centre of Research Excellence in Recovery Following Road Traffic Injury, The University of Queensland, Brisbane, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Katie McMahon
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | - Graham Galloway
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Translational Research Institute, Brisbane, Queensland, Australia
| | - Gail Durbridge
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | - Michele Sterling
- Recover Injury Research Centre, NHMRC Centre of Research Excellence in Recovery Following Road Traffic Injury, The University of Queensland, Brisbane, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
- * E-mail:
| |
Collapse
|
186
|
Connolly M, Paul R, Farre-Garros R, Natanek SA, Bloch S, Lee J, Lorenzo JP, Patel H, Cooper C, Sayer AA, Wort SJ, Griffiths M, Polkey MI, Kemp PR. miR-424-5p reduces ribosomal RNA and protein synthesis in muscle wasting. J Cachexia Sarcopenia Muscle 2018; 9:400-416. [PMID: 29215200 PMCID: PMC5879973 DOI: 10.1002/jcsm.12266] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/27/2017] [Accepted: 10/12/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A loss of muscle mass occurs as a consequence of a range of chronic and acute diseases as well as in older age. This wasting results from an imbalance of protein synthesis and degradation with a reduction in synthesis and resistance to anabolic stimulation often reported features. Ribosomes are required for protein synthesis, so changes in the control of ribosome synthesis are potential contributors to muscle wasting. MicroRNAs (miRNAs) are known regulators of muscle phenotype and have been shown to modulate components of the protein synthetic pathway. One miRNA that is predicted to target a number of components of protein synthetic pathway is miR-424-5p, which is elevated in the quadriceps of patients with chronic obstructive pulmonary disease (COPD). METHODS Targets of miR-424-5p were identified by Argonaute2 pull down, and the effects of the miRNA on RNA and protein expression were determined by quantitative polymerase chain reaction and western blotting in muscle cells in vitro. Protein synthesis was determined by puromycin incorporation in vitro. The miRNA was over-expressed in the tibialis anterior muscle of mice by electroporation and the effects quantified. Finally, quadriceps expression of the miRNA was determined by quantitative polymerase chain reaction in patients with COPD and intensive care unit (ICU)-acquired weakness and in patients undergoing aortic surgery as well as in individuals from the Hertfordshire Sarcopenia Study. RESULTS Pull-down assays showed that miR-424-5p bound to messenger RNAs encoding proteins associated with muscle protein synthesis. The most highly enriched messenger RNAs encoded proteins required for the Pol I RNA pre-initiation complex required for ribosomal RNA (rRNA) transcription, (PolR1A and upstream binding transcription factor). In vitro, miR-424-5p reduced the expression of these RNAs, reduced rRNA levels, and inhibited protein synthesis. In mice, over-expression of miR-322 (rodent miR-424 orthologue) caused fibre atrophy and reduced upstream binding transcription factor expression and rRNA levels. In humans, elevated miR-424-5p associated with markers of disease severity in COPD (FEV1 %), in patients undergoing aortic surgery (LVEF%), and in patients with ICU-acquired weakness (days in ICU). In patients undergoing aortic surgery, preoperative miR-424-5p expression in skeletal muscle was associated with muscle loss over the following 7 days. CONCLUSIONS These data suggest that miR-424-5p regulates rRNA synthesis by inhibiting Pol I pre-initiation complex formation. Increased miR-424-5p expression in patients with conditions associated with muscle wasting is likely to contribute to the inhibition of protein synthesis and loss of muscle mass.
Collapse
Affiliation(s)
- Martin Connolly
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Richard Paul
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.,National Institute for Health Research Respiratory Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, SW3 6NP, UK
| | - Roser Farre-Garros
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Samantha A Natanek
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Susannah Bloch
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Jen Lee
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Jose P Lorenzo
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Harnish Patel
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK.,NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Avan A Sayer
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK.,AGE Research Group, Institute of Neuroscience and Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Stephen J Wort
- National Institute for Health Research Respiratory Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, SW3 6NP, UK
| | - Mark Griffiths
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Michael I Polkey
- National Institute for Health Research Respiratory Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, SW3 6NP, UK
| | - Paul R Kemp
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| |
Collapse
|
187
|
Léger T, Charrier A, Moreau C, Hininger-Favier I, Mourmoura E, Rigaudière JP, Pitois E, Bouvier D, Sapin V, Pereira B, Azarnoush K, Demaison L. Early sepsis does not stimulate reactive oxygen species production and does not reduce cardiac function despite an increased inflammation status. Physiol Rep 2018; 5:5/13/e13231. [PMID: 28684640 PMCID: PMC5506518 DOI: 10.14814/phy2.13231] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 12/14/2022] Open
Abstract
If it is sustained for several days, sepsis can trigger severe abnormalities of cardiac function which leads to death in 50% of cases. This probably occurs through activation of toll-like receptor-9 by bacterial lipopolysaccharides and overproduction of proinflammatory cytokines such as TNF-α and IL-1β In contrast, early sepsis is characterized by the development of tachycardia. This study aimed at determining the early changes in the cardiac function during sepsis and at finding the mechanism responsible for the observed changes. Sixty male Wistar rats were randomly assigned to two groups, the first one being made septic by cecal ligation and puncture (sepsis group) and the second one being subjected to the same surgery without cecal ligation and puncture (sham-operated group). The cardiac function was assessed in vivo and ex vivo in standard conditions. Several parameters involved in the oxidative stress and inflammation were determined in the plasma and heart. As evidenced by the plasma level of TNF-α and gene expression of IL-1β and TNF-α in the heart, inflammation was developed in the sepsis group. The cardiac function was also slightly stimulated by sepsis in the in vivo and ex vivo situations. This was associated with unchanged levels of oxidative stress, but several parameters indicated a lower cardiac production of reactive oxygen species in the septic group. In conclusion, despite the development of inflammation, early sepsis did not increase reactive oxygen species production and did not reduce myocardial function. The depressant effect of TNF-α and IL-1β on the cardiac function is known to occur at very high concentrations. The influence of low- to moderate-grade inflammation on the myocardial mechanical behavior must thus be revisited.
Collapse
Affiliation(s)
- Thibault Léger
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France
| | - Alice Charrier
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France
| | - Clarisse Moreau
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France
| | - Isabelle Hininger-Favier
- Laboratoire de Bioénergétique Fondamentale et Appliquée, INSERM U1055, Université Joseph Fourier, Grenoble, France
| | - Evangelia Mourmoura
- Laboratoire de Bioénergétique Fondamentale et Appliquée, INSERM U1055, Université Joseph Fourier, Grenoble, France
| | | | - Elodie Pitois
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France
| | - Damien Bouvier
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Vincent Sapin
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Bruno Pereira
- Department of Clinical Research and Innovation, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Kasra Azarnoush
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France.,Heart Surgery Department, G. Montpied Hospital, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Luc Demaison
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France
| |
Collapse
|
188
|
Chhetri JK, de Souto Barreto P, Fougère B, Rolland Y, Vellas B, Cesari M. Chronic inflammation and sarcopenia: A regenerative cell therapy perspective. Exp Gerontol 2018; 103:115-123. [DOI: 10.1016/j.exger.2017.12.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/20/2017] [Accepted: 12/27/2017] [Indexed: 01/06/2023]
|
189
|
Wu CJ, Kao TW, Chang YW, Peng TC, Wu LW, Yang HF, Chen WL. Does the Additional Component of Calf Circumference Refine Metabolic Syndrome in Correlating With Cardiovascular Risk? J Clin Endocrinol Metab 2018; 103:1151-1160. [PMID: 29346655 DOI: 10.1210/jc.2017-02320] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/10/2018] [Indexed: 01/10/2023]
Abstract
CONTEXT Calf circumference (CC) was a useful anthropometric tool, but there was limited study on the effect of CC on metabolic syndrome (MetS) for cardiovascular risk. OBJECTIVE The objective of our study was to determine whether adding CC as a component of MetS refined correlating MetS with cardiovascular, all-cause, and cancer mortality risks. DESIGN, SETTING, PATIENTS, AND INTERVENTIONS From the National Health and Nutrition Examination Survey data set for 1999 through 2002, we analyzed four types of MetS: (1) increased waist circumference and two or more of four MetS components (WaistMetS); (2) decreased CC and two or more of four MetS components (CalfMetS); (3) increased waist-to-calf ratio and two or more of four MetS components (WCRMetS); and (4) decreased CC and three or more of five MetS components (CC+MetS). PRIMARY OUTCOME MEASURE The cause-specific hazard ratios were measured as categorized by the four types of MetS. RESULTS For cardiovascular mortality, the adjusted hazard ratios for WaistMetS, CalfMetS, WCRMetS, and CC+MetS were 1.867, 1.871, 1.949, and 2.306, respectively (all P < 0.001). Notably, CalfMetS showed the strongest positive correlation with serum C-reactive protein levels, and WCRMetS had the strongest positive relationship with homeostasis model assessment of insulin resistance. CONCLUSIONS Adding CC to the components of MetS correlated with higher cardiovascular and all-cause mortality risk than the traditional definition of MetS.
Collapse
Affiliation(s)
- Chen-Jung Wu
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Division of Family Medicine, Department of Community Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan, Republic of China
| | - Tung-Wei Kao
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Graduate Institute of Clinical Medical, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Yaw-Wen Chang
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Tao-Chun Peng
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Li-Wei Wu
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Hui-Fang Yang
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Wei-Liang Chen
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| |
Collapse
|
190
|
Duchesne E, Dufresne SS, Dumont NA. Impact of Inflammation and Anti-inflammatory Modalities on Skeletal Muscle Healing: From Fundamental Research to the Clinic. Phys Ther 2017; 97:807-817. [PMID: 28789470 DOI: 10.1093/ptj/pzx056] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 05/12/2017] [Indexed: 02/07/2023]
Abstract
Anti-inflammatory modalities are commonly used for the treatment of various musculoskeletal injuries. Although inflammation was originally believed to interfere with skeletal muscle regeneration, several recent studies have highlighted the beneficial effects of inflammatory cells on muscle healing. This discrepancy is attributable to an evolving understanding of the complex inflammatory process. To better appreciate the paradoxical roles of inflammation, clinicians must have a better comprehension of the fundamental mechanisms regulating the inflammatory response. In this perspective article, cellular, animal, and human studies were analyzed to summarize recent knowledge regarding the impact of inflammation on muscle regeneration in acute or chronic conditions. The effect of anti-inflammatory drugs on the treatment of various muscle injuries was also considered. Overall, this work aims to summarize the current state of the literature on the inflammatory process associated with muscle healing in order to give clinicians the necessary tools to have a more efficient and evidence-based approach to the treatment of muscle injuries and disorders.
Collapse
Affiliation(s)
- Elise Duchesne
- Département des Sciences de la Santé, Université du Québec à Chicoutimi, Saguenay, Quebec, Canada; and Groupe de Recherche Interdisciplinaire sur les Maladies Neuromusculaires, Centre Intégré Universitaire de Santé et de Services Sociaux du Saguenay-Lac-St-Jean, Saguenay, Quebec, Canada
| | - Sébastien S Dufresne
- Département des Sciences de la Santé, Université du Québec à Chicoutimi, Saguenay, Quebec, Canada; and CHU de Québec Research Center, Quebec City, Quebec, Canada; and Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Nicolas A Dumont
- Musculoskeletal Diseases and Rehabilitation Department, Ste-Justine Hospital Research Center, Montreal, Quebec, Canada; and Department of Rehabilitation, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
191
|
Yamada T, Steinz MM, Kenne E, Lanner JT. Muscle Weakness in Rheumatoid Arthritis: The Role of Ca 2+ and Free Radical Signaling. EBioMedicine 2017; 23:12-19. [PMID: 28781131 PMCID: PMC5605300 DOI: 10.1016/j.ebiom.2017.07.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/13/2017] [Accepted: 07/24/2017] [Indexed: 02/07/2023] Open
Abstract
In addition to the primary symptoms arising from inflammatory processes in the joints, muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Muscle weakness not only reduces the quality of life for the affected patients, but also dramatically increases the burden on society since patients' work ability decreases. A 25–70% reduction in muscular strength has been observed in pateints with RA when compared with age-matched healthy controls. The reduction in muscle strength is often larger than what could be explained by the reduction in muscle size in patients with RA, which indicates that intracellular (intrinsic) muscle dysfunction plays an important role in the underlying mechanism of muscle weakness associated with RA. In this review, we highlight the present understanding of RA-associated muscle weakness with special focus on how enhanced Ca2 + release from the ryanodine receptor and free radicals (reactive oxygen/nitrogen species) contributes to muscle weakness, and recent developments of novel therapeutic interventions. Muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Intrinsic muscle weakness is important in the underlying mechanisms of muscle weakness associated with rheumatoid arthritis. Enhanced Ca2 + release and peroxynitrite-induced stress contributes to RA-induced muscle weakness.
Collapse
Affiliation(s)
- Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Maarten M Steinz
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ellinor Kenne
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
192
|
Davies MR, Lee L, Feeley BT, Kim HT, Liu X. Lysophosphatidic acid-induced RhoA signaling and prolonged macrophage infiltration worsens fibrosis and fatty infiltration following rotator cuff tears. J Orthop Res 2017; 35:1539-1547. [PMID: 27505847 PMCID: PMC5502767 DOI: 10.1002/jor.23384] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/05/2016] [Indexed: 02/04/2023]
Abstract
Previous studies have suggested that macrophage-mediated chronic inflammation is involved in the development of rotator cuff muscle atrophy and degeneration following massive tendon tears. Increased RhoA signaling has been reported in chronic muscle degeneration, such as muscular dystrophy. However, the role of RhoA signaling in macrophage infiltration and rotator muscle degeneration remains unknown. Using a previously established rat model of massive rotator cuff tears, we found RhoA signaling is upregulated in rotator cuff muscle following a massive tendon-nerve injury. This increase in RhoA expression is greatly potentiated by the administration of a potent RhoA activator, lysophosphatidic acid (LPA), and is accompanied by increased TNFα and TGF-β1 expression in rotator cuff muscle. Boosting RhoA signaling with LPA significantly worsened rotator cuff muscle atrophy, fibrosis, and fatty infiltration, accompanied with massive monocytic infiltration of rotator cuff muscles. Co-staining of RhoA and the tissue macrophage marker CD68 showed that CD68+ tissue macrophages are the dominant cell source of increased RhoA signaling in rotator cuff muscles after tendon tears. Taken together, our findings suggest that LPA-mediated RhoA signaling in injured muscle worsens the outcomes of atrophy, fibrosis, and fatty infiltration by increasing macrophage infiltraion in rotator cuff muscle. Clinically, inhibiting RhoA signaling may represent a future direction for developing new treatments to improve muscle quality following massive rotator cuff tears. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1539-1547, 2017.
Collapse
Affiliation(s)
- Michael R. Davies
- San Francisco Veterans Affairs Health Care System, Department of Veterans Affairs, 1700 Owens Street, Room 364 San Francisco, California 94153
- Department of Orthopaedic Surgery, University of California, San Francisco, California
| | - Lawrence Lee
- San Francisco Veterans Affairs Health Care System, Department of Veterans Affairs, 1700 Owens Street, Room 364 San Francisco, California 94153
| | - Brian T. Feeley
- San Francisco Veterans Affairs Health Care System, Department of Veterans Affairs, 1700 Owens Street, Room 364 San Francisco, California 94153
- Department of Orthopaedic Surgery, University of California, San Francisco, California
| | - Hubert T. Kim
- San Francisco Veterans Affairs Health Care System, Department of Veterans Affairs, 1700 Owens Street, Room 364 San Francisco, California 94153
- Department of Orthopaedic Surgery, University of California, San Francisco, California
| | - Xuhui Liu
- San Francisco Veterans Affairs Health Care System, Department of Veterans Affairs, 1700 Owens Street, Room 364 San Francisco, California 94153
- Department of Orthopaedic Surgery, University of California, San Francisco, California
| |
Collapse
|
193
|
Gomez JP, Gonçalves C, Pichon C, Midoux P. Effect of IL-1β, TNF-α and IGF-1 on trans-endothelial passage of synthetic vectors through an in vitro vascular endothelial barrier of striated muscle. Gene Ther 2017; 24:416-424. [DOI: 10.1038/gt.2017.40] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022]
|
194
|
Haddad M. Adenosine A2B Receptors - Mediated Induction of Interleukin-6 in Skeletal Muscle Cells. Turk J Pharm Sci 2017; 14:19-28. [PMID: 32454590 DOI: 10.4274/tjps.08108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/28/2016] [Indexed: 01/10/2023]
Abstract
Objectives Inflammatory response and cytokine activation are markedly stimulated in skeletal muscle during various conditions. Interleukin-6 (IL-6), a pro-inflammatory cytokine, has pleiotropic effects on skeletal muscle. Adenosine, released by all cell types, binds to a class of G protein-coupled receptors to induce various skeletal muscle effects. The aim of this work was to investigate whether activation of adenosine receptors, particularly adenosine A2B receptors, could stimulate IL-6 gene expression in rat L6 skeletal muscle cells. Materials and Methods The rat L6 skeletal muscle cells were cultured in 25 cm2 flasks. These differentiated cells were treated and then quantitative reverse transcription-polymerase chain reaction (Probe-based) was used to analyze IL-6 gene expression level among different treatment conditions. Results Adenosine-5'-N-ethyluronamide (NECA), a stable adenosine analogue, concentration- and time-dependently stimulates IL-6 gene expression in skeletal muscle cells. The effect of NECA is inhibited by a selective adenosine A2B receptor antagonist, PSB 603. By using cyclic adenosine monophosphate (cAMP)-arising reagent forskolin, cAMP is found to be involved in the up-regulation of IL-6 induction. Conclusion Here, a novel relationship between adenosine and IL-6 up-regulation has been demonstrated for the first time; IL-6 up-regulation induced by NECA is mediated by adenosine A2B receptor activation in skeletal muscle and is dependent on mainly a cAMP pathway. Adenosine A2B receptors are, thus, potentially important pharmacological targets in treating inflammation and related diseases in skeletal muscle tissues.
Collapse
Affiliation(s)
- Mansour Haddad
- Department Of Clinical Sciences, Faculty Of Pharmacy, Philadelphia University, Amman, Jordan
| |
Collapse
|
195
|
Phillips SM, Dickerson RN, Moore FA, Paddon-Jones D, Weijs PJM. Protein Turnover and Metabolism in the Elderly Intensive Care Unit Patient. Nutr Clin Pract 2017; 32:112S-120S. [PMID: 28388378 DOI: 10.1177/0884533616686719] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Many intensive care unit (ICU) patients do not achieve target protein intakes particularly in the early days following admittance. This period of iatrogenic protein undernutrition contributes to a rapid loss of lean, in particular muscle, mass in the ICU. The loss of muscle in older (aged >60 years) patients in the ICU may be particularly rapid due to a perfect storm of increased catabolic factors, including systemic inflammation, disuse, protein malnutrition, and reduced anabolic stimuli. This loss of muscle mass has marked consequences. It is likely that the older patient is already experiencing muscle loss due to sarcopenia; however, the period of stay in the ICU represents a greatly accelerated period of muscle loss. Thus, on discharge, the older ICU patient is now on a steeper downward trajectory of muscle loss, more likely to have ICU-acquired muscle weakness, and at risk of becoming sarcopenic and/or frail. One practice that has been shown to have benefit during ICU stays is early ambulation and physical therapy (PT), and it is likely that both are potent stimuli to induce a sensitivity of protein anabolism. Thus, recommendations for the older ICU patient would be provision of at least 1.2-1.5 g protein/kg usual body weight/d, regular and early utilization of ambulation (if possible) and/or PT, and follow-up rehabilitation for the older discharged ICU patient that includes rehabilitation, physical activity, and higher habitual dietary protein to change the trajectory of ICU-mediated muscle mass loss and weakness.
Collapse
Affiliation(s)
- Stuart M Phillips
- 1 McMaster University, Department of Kinesiology, Hamilton, Ontario, Canada
| | - Roland N Dickerson
- 2 Department of Clinical Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Frederick A Moore
- 3 Department of Surgery, Division of Acute Care Surgery, and Center for Sepsis and Critical Illness Research, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Douglas Paddon-Jones
- 4 Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas, USA
| | - Peter J M Weijs
- 5 Nutrition and Dietetics, Department of Internal Medicine, Department of Intensive Care Medicine, and Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam, The Netherlands.,6 Nutrition and Dietetics, Faculty of Sports and Nutrition, Amsterdam University of Applied Sciences, Amsterdam, The Netherlands
| |
Collapse
|
196
|
Au ED, Desai AP, Koniaris LG, Zimmers TA. The MEK-Inhibitor Selumetinib Attenuates Tumor Growth and Reduces IL-6 Expression but Does Not Protect against Muscle Wasting in Lewis Lung Cancer Cachexia. Front Physiol 2017; 7:682. [PMID: 28149280 PMCID: PMC5241300 DOI: 10.3389/fphys.2016.00682] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/22/2016] [Indexed: 01/06/2023] Open
Abstract
Cachexia, or wasting of skeletal muscle and fat, afflicts many patients with chronic diseases including cancer, organ failure, and AIDS. Muscle wasting reduces quality of life and decreases response to therapy. Cachexia is caused partly by elevated inflammatory cytokines, including interleukin-6 (IL-6). Others and we have shown that IL-6 alone is sufficient to induce cachexia both in vitro and in vivo. The mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK) inhibitor Selumetinib has been tested in clinical trials for various cancers. Moreover, Selumetinib has also been shown to inhibit the production of IL-6. In a retrospective analysis of a phase II clinical trial in advanced cholangiocarcinoma, patients treated with Selumetinib experienced significant gains in skeletal muscle vs. patients receiving standard therapy. However, the use of Selumetinib as a treatment for cachexia has yet to be investigated mechanistically. We sought to determine whether MEK inhibition could protect against cancer-induced cachexia in mice. In vitro, Selumetinib induced C2C12 myotube hypertrophy and nuclear accretion. Next we tested Selumetinib in the Lewis lung carcinoma (LLC) model of cancer cachexia. Treatment with Selumetinib reduced tumor mass and reduced circulating and tumor IL-6; however MEK inhibition did not preserve muscle mass. Similar wasting was seen in limb muscles of Selumetinib and vehicle-treated LLC mice, while greater fat and carcass weight loss was observed with Selumetinib treatment. As well, Selumetinib did not block wasting in C2C12 myotubes treated with LLC serum. Taken together, out results suggest that this MEK inhibitor is not protective in LLC cancer cachexia despite lowering IL-6 levels, and further that it might exacerbate tumor-induced weight loss. Differences from other studies might be disease, species or model-specific.
Collapse
Affiliation(s)
- Ernie D Au
- Department of Surgery, Indiana University School of MedicineIndianapolis, IN, USA; Department of Biochemistry and Molecular Biology, Indiana University School of MedicineIndianapolis, IN, USA; Indiana University Simon Cancer CenterIndianapolis, IN, USA
| | - Aditya P Desai
- Department of Surgery, Indiana University School of MedicineIndianapolis, IN, USA; Indiana University Simon Cancer CenterIndianapolis, IN, USA
| | - Leonidas G Koniaris
- Department of Surgery, Indiana University School of MedicineIndianapolis, IN, USA; Indiana University Simon Cancer CenterIndianapolis, IN, USA; IUPUI Center for Cachexia Research, Innovation and TherapyIndianapolis, IN, USA
| | - Teresa A Zimmers
- Department of Surgery, Indiana University School of MedicineIndianapolis, IN, USA; Department of Biochemistry and Molecular Biology, Indiana University School of MedicineIndianapolis, IN, USA; Indiana University Simon Cancer CenterIndianapolis, IN, USA; IUPUI Center for Cachexia Research, Innovation and TherapyIndianapolis, IN, USA; Department of Otolaryngology, Head and Neck Surgery, Indiana University School of MedicineIndianapolis, IN, USA; Department of Anatomy and Cell Biology, Indiana University School of MedicineIndianapolis, IN, USA
| |
Collapse
|
197
|
Physiological and functional failure in chronic obstructive pulmonary disease, congestive heart failure and cancer: a debilitating intersection of sarcopenia, cachexia and breathlessness. Curr Opin Support Palliat Care 2016; 10:236-41. [DOI: 10.1097/spc.0000000000000222] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
198
|
Coelho Junior HJ, Gambassi BB, Diniz TA, Fernandes IMDC, Caperuto ÉC, Uchida MC, Lira FS, Rodrigues B. Inflammatory Mechanisms Associated with Skeletal Muscle Sequelae after Stroke: Role of Physical Exercise. Mediators Inflamm 2016; 2016:3957958. [PMID: 27647951 PMCID: PMC5018330 DOI: 10.1155/2016/3957958] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/26/2016] [Indexed: 12/25/2022] Open
Abstract
Inflammatory markers are increased systematically and locally (e.g., skeletal muscle) in stroke patients. Besides being associated with cardiovascular risk factors, proinflammatory cytokines seem to play a key role in muscle atrophy by regulating the pathways involved in this condition. As such, they may cause severe decrease in muscle strength and power, as well as impairment in cardiorespiratory fitness. On the other hand, physical exercise (PE) has been widely suggested as a powerful tool for treating stroke patients, since PE is able to regenerate, even if partially, physical and cognitive functions. However, the mechanisms underlying the beneficial effects of physical exercise in poststroke patients remain poorly understood. Thus, in this study we analyze the candidate mechanisms associated with muscle atrophy in stroke patients, as well as the modulatory effect of inflammation in this condition. Later, we suggest the two strongest anti-inflammatory candidate mechanisms, myokines and the cholinergic anti-inflammatory pathway, which may be activated by physical exercise and may contribute to a decrease in proinflammatory markers of poststroke patients.
Collapse
Affiliation(s)
| | | | - Tiego Aparecido Diniz
- Exercise and Immunometabolism Research Group, Department of Physical Education, São Paulo State University (UNESP), 19060-900 Presidente Prudente, SP, Brazil
| | - Isabela Maia da Cruz Fernandes
- Exercise and Immunometabolism Research Group, Department of Physical Education, São Paulo State University (UNESP), 19060-900 Presidente Prudente, SP, Brazil
| | - Érico Chagas Caperuto
- Human Movement Laboratory, São Judas Tadeu University (USJT), 03166-000 São Paulo, SP, Brazil
| | - Marco Carlos Uchida
- Faculty of Physical Education, University of Campinas (UNICAMP), 13083-851 Campinas, SP, Brazil
| | - Fabio Santos Lira
- Exercise and Immunometabolism Research Group, Department of Physical Education, São Paulo State University (UNESP), 19060-900 Presidente Prudente, SP, Brazil
| | - Bruno Rodrigues
- Faculty of Physical Education, University of Campinas (UNICAMP), 13083-851 Campinas, SP, Brazil
| |
Collapse
|
199
|
Zimmers TA, Fishel ML, Bonetto A. STAT3 in the systemic inflammation of cancer cachexia. Semin Cell Dev Biol 2016; 54:28-41. [PMID: 26860754 PMCID: PMC4867234 DOI: 10.1016/j.semcdb.2016.02.009] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/04/2016] [Indexed: 02/07/2023]
Abstract
Weight loss is diagnostic of cachexia, a debilitating syndrome contributing mightily to morbidity and mortality in cancer. Most research has probed mechanisms leading to muscle atrophy and adipose wasting in cachexia; however cachexia is a truly systemic phenomenon. Presence of the tumor elicits an inflammatory response and profound metabolic derangements involving not only muscle and fat, but also the hypothalamus, liver, heart, blood, spleen and likely other organs. This global response is orchestrated in part through circulating cytokines that rise in conditions of cachexia. Exogenous Interleukin-6 (IL6) and related cytokines can induce most cachexia symptomatology, including muscle and fat wasting, the acute phase response and anemia, while IL-6 inhibition reduces muscle loss in cancer. Although mechanistic studies are ongoing, certain of these cachexia phenotypes have been causally linked to the cytokine-activated transcription factor, STAT3, including skeletal muscle wasting, cardiac dysfunction and hypothalamic inflammation. Correlative studies implicate STAT3 in fat wasting and the acute phase response in cancer cachexia. Parallel data in non-cancer models and disease states suggest both pathological and protective functions for STAT3 in other organs during cachexia. STAT3 also contributes to cancer cachexia through enhancing tumorigenesis, metastasis and immune suppression, particularly in tumors associated with high prevalence of cachexia. This review examines the evidence linking STAT3 to multi-organ manifestations of cachexia and the potential and perils for targeting STAT3 to reduce cachexia and prolong survival in cancer patients.
Collapse
Affiliation(s)
- Teresa A Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IUPUI Center for Cachexia Research Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| | - Melissa L Fishel
- IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IUPUI Center for Cachexia Research Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| |
Collapse
|
200
|
Chen T, Moore TM, Ebbert MTW, McVey NL, Madsen SR, Hallowell DM, Harris AM, Char RE, Mackay RP, Hancock CR, Hansen JM, Kauwe JS, Thomson DM. Liver kinase B1 inhibits the expression of inflammation-related genes postcontraction in skeletal muscle. J Appl Physiol (1985) 2016; 120:876-88. [PMID: 26796753 DOI: 10.1152/japplphysiol.00727.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/20/2016] [Indexed: 01/06/2023] Open
Abstract
Skeletal muscle-specific liver kinase B1 (LKB1) knockout mice (skmLKB1-KO) exhibit elevated mitogen-activated protein kinase (MAPK) signaling after treadmill running. MAPK activation is also associated with inflammation-related signaling in skeletal muscle. Since exercise can induce muscle damage, and inflammation is a response triggered by damaged tissue, we therefore hypothesized that LKB1 plays an important role in dampening the inflammatory response to muscle contraction, and that this may be due in part to increased susceptibility to muscle damage with contractions in LKB1-deficient muscle. Here we studied the inflammatory response and muscle damage with in situ muscle contraction or downhill running. After in situ muscle contractions, the phosphorylation of both NF-κB and STAT3 was increased more in skmLKB1-KO vs. wild-type (WT) muscles. Analysis of gene expression via microarray and RT-PCR shows that expression of many inflammation-related genes increased after contraction only in skmLKB1-KO muscles. This was associated with mild skeletal muscle fiber membrane damage in skmLKB1-KO muscles. Gene markers of oxidative stress were also elevated in skmLKB1-KO muscles after contraction. Using the downhill running model, we observed significantly more muscle damage after running in skmLKB1-KO mice, and this was associated with greater phosphorylation of both Jnk and STAT3 and increased expression of SOCS3 and Fos. In conclusion, we have shown that the lack of LKB1 in skeletal muscle leads to an increased inflammatory state in skeletal muscle that is exacerbated by muscle contraction. Increased susceptibility of the muscle to damage may underlie part of this response.
Collapse
Affiliation(s)
- Ting Chen
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Timothy M Moore
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Mark T W Ebbert
- Department of Biology, Brigham Young University, Provo, Utah
| | - Natalie L McVey
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Steven R Madsen
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - David M Hallowell
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Alexander M Harris
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Robin E Char
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Ryan P Mackay
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Chad R Hancock
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, Utah; and
| | - Jason M Hansen
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - John S Kauwe
- Department of Biology, Brigham Young University, Provo, Utah
| | - David M Thomson
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah;
| |
Collapse
|