1
|
Zhao N, Michelucci A, Pietrangelo L, Malik S, Groom L, Leigh J, O'Connor TN, Takano T, Kingsley PD, Palis J, Boncompagni S, Protasi F, Dirksen RT. An Orai1 gain-of-function tubular aggregate myopathy mouse model phenocopies key features of the human disease. EMBO J 2024; 43:5941-5971. [PMID: 39420094 PMCID: PMC11612304 DOI: 10.1038/s44318-024-00273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Tubular aggregate myopathy (TAM) is a heritable myopathy primarily characterized by progressive muscle weakness, elevated levels of creatine kinase (CK), hypocalcemia, exercise intolerance, and the presence of tubular aggregates (TAs). Here, we generated a knock-in mouse model based on a human gain-of-function mutation which results in a severe, early-onset form of TAM, by inducing a glycine-to-serine point mutation in the ORAI1 pore (Orai1G100S/+ or GS mice). By 8 months of age, GS mice exhibited significant muscle weakness, exercise intolerance, elevated CK levels, hypocalcemia, and robust TA presence. Unexpectedly, constitutive Ca2+ entry in mutant mice was observed in muscle only during early development and was abolished in adult skeletal muscle, partly due to reduced ORAI1 expression. Consistent with proteomic results, significant mitochondrial damage and dysfunction was observed in skeletal muscle of GS mice. Thus, GS mice represent a powerful model for investigation of the pathophysiological mechanisms that underlie key TAM symptoms, as well as those compensatory responses that limit the damaging effects of uncontrolled ORAI1-mediated Ca2+ influx.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Antonio Michelucci
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Laura Pietrangelo
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University Gabriele d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Linda Groom
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jennifer Leigh
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas N O'Connor
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Takahiro Takano
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Paul D Kingsley
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - James Palis
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology & DNICS, Department of Neuroscience and Clinical Sciences, University Gabriele d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Feliciano Protasi
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University Gabriele d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
2
|
Fu L, Song L, Zhou X, Chen L, Zheng L, Hu D, Zhu S, Hu Y, Gong D, Chen CL, Ye X, Yu S. Serum metabolomics analysis of malnutrition in patients with gastric cancer: a cross sectional study. BMC Cancer 2024; 24:1195. [PMID: 39333934 PMCID: PMC11438121 DOI: 10.1186/s12885-024-12964-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Although malnutrition is common in cancer patients, its molecular mechanisms has not been fully clarified. This study aims to identify significantly differential metabolites, match the corresponding metabolic pathways, and develop a predictive model of malnutrition in patients with gastric cancer. METHODS In this cross-sectional study, we applied non-targeted metabolomics using liquid chromatography-mass spectrometry to explore the serum fingerprinting of malnutrition in patients with gastric cancer. Malnutrition-specific differential metabolites were identified by orthogonal partial least-squares discriminant analysis and t-test and matched with the Human Metabolome Database and the LIPID Metabolites and Pathways Strategy. We matched the corresponding metabolic pathways of malnutrition using pathway analysis at the MetaboAnalyst 5.0. We used random forest analyses to establish the predictive model. RESULTS We recruited 220 malnourished and 198 non-malnourished patients with gastric cancer. The intensities of 25 annotated significantly differential metabolites were lower in patients with malnutrition than those without, while two others were higher in patients with malnutrition than those without, including newly identified significantly differential metabolites such as indoleacrylic acid and lysophosphatidylcholine(18:3/0:0). We matched eight metabolic pathways associated with malnutrition, including aminoacyl-tRNA biosynthesis, tryptophan metabolism, and glycerophospholipid metabolism. We established a predictive model with an area under the curve of 0.702 (95% CI: 0.651-0.768) based on four annotated significantly differential metabolites, namely indoleacrylic acid, lysophosphatidylcholine(18:3/0:0), L-tryptophan, and lysophosphatidylcholine(20:3/0:0). CONCLUSIONS We identified 27 specific differential metabolites of malnutrition in malnourished compared to non-malnourished patients with gastric cancer. We also matched eight corresponding metabolic pathways and developed a predictive model. These findings provide supportive data to better understand molecular mechanisms of malnutrition in patients with gastric cancer and new strategies for the prediction, diagnosis, prevention, and treatment for those malnourished.
Collapse
Affiliation(s)
- Liang Fu
- Department of Nursing, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Lixin Song
- School of Nursing, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, 78229, USA
| | - Xi Zhou
- Institute of Analysis, Guangdong Academy of Sciences (China National Analytical Center), Guangzhou, Guangzhou, 510070, China
| | - Lin Chen
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Lushan Zheng
- Department of Nursing, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Dandan Hu
- Department of Nursing, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Sha Zhu
- Department of Nursing, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Yanting Hu
- Department of Pathology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Daojun Gong
- Department of Gastrointestinal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
| | - Chun-Liang Chen
- School of Nursing, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, 78229, USA
| | - Xianghong Ye
- Department of Nursing, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China.
| | - Shian Yu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China.
| |
Collapse
|
3
|
Jadzic J, Djonic D. Hepatocellular carcinoma and musculoskeletal system: A narrative literature review. World J Gastroenterol 2024; 30:2109-2117. [PMID: 38681992 PMCID: PMC11045483 DOI: 10.3748/wjg.v30.i15.2109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/07/2024] [Accepted: 03/26/2024] [Indexed: 04/19/2024] Open
Abstract
Musculoskeletal alterations in hepatocellular carcinoma (HCC) are less common than liver-related complications. However, they can significantly impact the quality of life and overall prognosis of patients with HCC. The main obstacle in the clinical assessment of HCC-induced musculoskeletal alterations is related to effective and timely diagnosis because these complications are often asymptomatic and unapparent during routine clinical evaluations. This narrative literature review aimed to provide a comprehensive overview of the contemporary literature related to the changes in the musculoskeletal system in patients with HCC, focusing on its clinical implications and underlying etiopathogenetic mechanisms. Osteolytic bone metastases are the most common skeletal alterations associated with HCC, which could be associated with an increased risk of low-trauma bone fracture. Moreover, previous studies reported that osteopenia, sarcopenia, and myosteatosis are associated with poor clinical outcomes in patients with HCC. Even though low bone mineral density and sarcopenia are consistently reported as reliable predictors of pretransplantation and post-transplantation mortality in HCC patients, these complications are frequently overlooked in the clinical management of patients with HCC. Taken together, contemporary literature suggests that a multidisciplinary approach is essential for early recognition and clinical management of HCC-associated musculoskeletal alterations to improve patient prognosis. Further research into the mechanisms and treatment options for musculoskeletal complications is warranted to enhance our understanding and clinical management of this aspect of HCC.
Collapse
Affiliation(s)
- Jelena Jadzic
- Center of Bone Biology, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Danijela Djonic
- Center of Bone Biology, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| |
Collapse
|
4
|
Jadzic J, Djonic D. Hepatocellular carcinoma and musculoskeletal system: A narrative literature review. World J Gastroenterol 2024; 30:2109-2117. [DOI: https:/doi.org/10.3748/wjg.v30.i15.2109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/10/2024] Open
Abstract
Musculoskeletal alterations in hepatocellular carcinoma (HCC) are less common than liver-related complications. However, they can significantly impact the quality of life and overall prognosis of patients with HCC. The main obstacle in the clinical assessment of HCC-induced musculoskeletal alterations is related to effective and timely diagnosis because these complications are often asymptomatic and unapparent during routine clinical evaluations. This narrative literature review aimed to provide a comprehensive overview of the contemporary literature related to the changes in the musculoskeletal system in patients with HCC, focusing on its clinical implications and underlying etiopathogenetic mechanisms. Osteolytic bone metastases are the most common skeletal alterations associated with HCC, which could be associated with an increased risk of low-trauma bone fracture. Moreover, previous studies reported that osteopenia, sarcopenia, and myosteatosis are associated with poor clinical outcomes in patients with HCC. Even though low bone mineral density and sarcopenia are consistently reported as reliable predictors of pretransplantation and post-transplantation mortality in HCC patients, these complications are frequently overlooked in the clinical management of patients with HCC. Taken together, contemporary literature suggests that a multidisciplinary approach is essential for early recognition and clinical management of HCC-associated musculoskeletal alterations to improve patient prognosis. Further research into the mechanisms and treatment options for musculoskeletal complications is warranted to enhance our understanding and clinical management of this aspect of HCC.
Collapse
|
5
|
Le Bacquer O, Sanchez P, Patrac V, Rivoirard C, Saroul N, Giraudet C, Kocer A, Walrand S. Cannabidiol protects C2C12 myotubes against cisplatin-induced atrophy by regulating oxidative stress. Am J Physiol Cell Physiol 2024; 326:C1226-C1236. [PMID: 38406827 DOI: 10.1152/ajpcell.00622.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/09/2024] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
Cancer and chemotherapy induce a severe loss of muscle mass (known as cachexia), which negatively impact cancer treatment and patient survival. The aim of the present study was to investigate whether cannabidiol (CBD) administration may potentially antagonize the effects of cisplatin in inducing muscle atrophy, using a model of myotubes in culture. Cisplatin treatment resulted in a reduction of myotube diameter (15.7 ± 0.3 vs. 22.2 ± 0.5 µm, P < 0.01) that was restored to control level with 5 µM CBD (20.1 ± 0.4 µM, P < 0.01). Protein homeostasis was severely altered with a ≈70% reduction in protein synthesis (P < 0.01) and a twofold increase in proteolysis (P < 0.05) in response to cisplatin. Both parameters were dose dependently restored by CBD cotreatment. Cisplatin treatment was associated with increased thiobarbituric acid reactive substances (TBARS) content (0.21 ± 0.03 to 0.48 ± 0.03 nmol/mg prot, P < 0.05), catalase activity (0.24 ± 0.01 vs. 0.13 ± 0.02 nmol/min/µg prot, P < 0.01), whereas CBD cotreatment normalized TBARS content to control values (0.22 ± 0.01 nmol/mg prot, P < 0.01) and reduced catalase activity (0.17 ± 0.01 nmol/min/µg prot, P < 0.05). These changes were associated with increased mRNA expression of GPX1, SOD1, SOD2, and CAT mRNA expression in response to cisplatin (P < 0.01), which was corrected by CBD cotreatment (P < 0.05). Finally, cisplatin treatment increased the mitochondrial protein content of NDUFB8, UQCRC2, COX4, and VDAC1 (involved in mitochondrial respiration and apoptosis), and CBD cotreatment restored their expression to control values. Altogether, our results demonstrated that CBD antagonize the cisplatin-induced C2C12 myotube atrophy and could be used as an adjuvant in the treatment of cancer cachexia to help maintain muscle mass and improve patient quality of life.NEW & NOTEWORTHY In an in vitro model, cisplatin treatment led to myotube atrophy associated with dysregulation of protein homeostasis and increased oxidative stress, resulting in increased apoptosis. Cotreatment with cannabidiol was able to prevent this phenotype by promoting protein homeostasis and reducing oxidative stress.
Collapse
Affiliation(s)
- Olivier Le Bacquer
- Unité de Nutrition Humaine (UNH), Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
| | - Phelipe Sanchez
- Unité de Nutrition Humaine (UNH), Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
| | - Véronique Patrac
- Unité de Nutrition Humaine (UNH), Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
| | - César Rivoirard
- Unité de Nutrition Humaine (UNH), Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
| | - Nicolas Saroul
- Unité de Nutrition Humaine (UNH), Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
- Department of Head and Neck Surgery, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Christophe Giraudet
- Unité de Nutrition Humaine (UNH), Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
| | - Ayhan Kocer
- Université Clermont Auvergne, iGReD, CNRS UMR 6293, INSERM U1103, Clermont-Ferrand, France
| | - Stéphane Walrand
- Unité de Nutrition Humaine (UNH), Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
- CHU Clermont-Ferrand, Service Nutrition Clinique, Clermont-Ferrand, France
| |
Collapse
|
6
|
Affourtit C, Carré JE. Mitochondrial involvement in sarcopenia. Acta Physiol (Oxf) 2024; 240:e14107. [PMID: 38304924 DOI: 10.1111/apha.14107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
Sarcopenia lowers the quality-of-life for millions of people across the world, as accelerated loss of skeletal muscle mass and function contributes to both age- and disease-related frailty. Physical activity remains the only proven therapy for sarcopenia to date, but alternatives are much sought after to manage this progressive muscle disorder in individuals who are unable to exercise. Mitochondria have been widely implicated in the etiology of sarcopenia and are increasingly suggested as attractive therapeutic targets to help restore the perturbed balance between protein synthesis and breakdown that underpins skeletal muscle atrophy. Reviewing current literature, we note that mitochondrial bioenergetic changes in sarcopenia are generally interpreted as intrinsic dysfunction that renders muscle cells incapable of making sufficient ATP to fuel protein synthesis. Based on the reported mitochondrial effects of therapeutic interventions, however, we argue that the observed bioenergetic changes may instead reflect an adaptation to pathologically decreased energy expenditure in sarcopenic muscle. Discrimination between these mechanistic possibilities will be crucial for improving the management of sarcopenia.
Collapse
Affiliation(s)
| | - Jane E Carré
- School of Biomedical Sciences, University of Plymouth, Plymouth, UK
| |
Collapse
|
7
|
Cardaci TD, VanderVeen BN, Bullard BM, McDonald SJ, Unger CA, Enos RT, Fan D, Velázquez KT, Frizzell N, Spangenburg EE, Murphy EA. Obesity worsens mitochondrial quality control and does not protect against skeletal muscle wasting in murine cancer cachexia. J Cachexia Sarcopenia Muscle 2024; 15:124-137. [PMID: 38062911 PMCID: PMC10834333 DOI: 10.1002/jcsm.13391] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/15/2023] [Accepted: 11/02/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND More than 650 million people are obese (BMI > 30) worldwide, which increases their risk for several metabolic diseases and cancer. While cachexia and obesity are at opposite ends of the weight spectrum, leading many to suggest a protective effect of obesity against cachexia, mechanistic support for obesity's benefit is lacking. Given that obesity and cachexia are both accompanied by metabolic dysregulation, we sought to investigate the impact of obesity on skeletal muscle mass loss and mitochondrial dysfunction in murine cancer cachexia. METHODS Male C57BL/6 mice were given a purified high fat or standard diet for 16 weeks before being implanted with 106 Lewis lung carcinoma (LLC) cells. Mice were monitored for 25 days, and hindlimb muscles were collected for cachexia indices and mitochondrial assessment via western blotting, high-resolution respirometry and transmission electron microscopy (TEM). RESULTS Obese LLC mice experienced significant tumour-free body weight loss similar to lean (-12.8% vs. -11.8%, P = 0.0001) but had reduced survival (33.3% vs. 6.67%, χ2 = 10.04, P = 0.0182). Obese LLC mice had reduced muscle weights (-24%, P < 0.0354) and mCSA (-16%, P = 0.0004) with similar activation of muscle p65 (P = 0.0337), and p38 (P = 0.0008). ADP-dependent coupled respiration was reduced in both Obese and Obese LLC muscle (-30%, P = 0.0072) consistent with reductions in volitional cage activity (-39%, P < 0.0001) and grip strength (-41%, P < 0.0001). TEM revealed stepwise reductions in intermyofibrillar and subsarcolemmal mitochondrial size with Obese (IMF: -37%, P = 0.0009; SS: -21%, P = 0.0101) and LLC (IMF: -40%, P = 0.0019; SS: -27%, P = 0.0383) mice. Obese LLC mice had increased pAMPK (T172; P = 0.0103) and reduced FIS1 (P = 0.0029) and DRP1 (P < 0.0001) mitochondrial fission proteins, which were each unchanged in Lean LLC. Further, mitochondrial TEM analysis revealed that Obese LLC mice had an accumulation of damaged and dysfunctional mitochondria (IMF: 357%, P = 0.0395; SS: 138%, P = 0.0174) in concert with an accumulation of p62 (P = 0.0328) suggesting impaired autophagy and clearance of damaged mitochondria. Moreover, we observed increases in electron lucent vacuoles only in Obese LLC muscle (IMF: 421%, P = 0.0260; SS: 392%, P = 0.0192), further supporting an accumulation of damaged materials that cannot be properly cleared in the obese cachectic muscle. CONCLUSIONS Taken together, these results demonstrate that obesity is not protective against cachexia and suggest exacerbated impairments to mitochondrial function and quality control with a particular disruption in the removal of damaged mitochondria. Our findings highlight the need for consideration of the severity of obesity and pre-existing metabolic conditions when determining the impact of weight status on cancer-induced cachexia and functional mitochondrial deficits.
Collapse
Affiliation(s)
- Thomas D Cardaci
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Brandon N VanderVeen
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Brooke M Bullard
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Sierra J McDonald
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Christian A Unger
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Reilly T Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Kandy T Velázquez
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Norma Frizzell
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Espen E Spangenburg
- Department of Physiology, East Carolina University, Greenville, North Carolina, USA
- Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - E Angela Murphy
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| |
Collapse
|
8
|
Sandek A, Gertler C, Valentova M, Jauert N, Wallbach M, Doehner W, von Haehling S, Anker SD, Fielitz J, Volk HD. Increased Expression of Proinflammatory Genes in Peripheral Blood Cells Is Associated with Cardiac Cachexia in Patients with Heart Failure with Reduced Ejection Fraction. J Clin Med 2024; 13:733. [PMID: 38337428 PMCID: PMC10856330 DOI: 10.3390/jcm13030733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Background: Cardiac cachexia (CC) in chronic heart failure with reduced ejection fraction (HFrEF) is characterized by catabolism and inflammation predicting poor prognosis. Levels of responsible transcription factors like signal transducer and activator of transcription (STAT)1, STAT3, suppressor of cytokine signaling (SOCS)1 and SOCS3 in peripheral blood cells (PBC) are underinvestigated in CC. Expression of mediators was related to patients' functional status, body composition (BC) and metabolic gene expression in skeletal muscle (SM). Methods: Gene expression was quantified by qRT-PCR in three cohorts: non-cachectic patients (ncCHF, n = 19, LVEF 31 ± 7%, BMI 30.2 ± 5.0 kg/m2), cachectic patients (cCHF; n = 18, LVEF 27 ± 7%, BMI 24.3 ± 2.5 kg/m2) and controls (n = 17, LVEF 70 ± 7%, BMI 27.6 ± 4.6 kg/m2). BC was assessed by dual-energy X-ray absorptiometry. Blood inflammatory markers were measured. We quantified solute carrier family 2 member 4 (SLC2A4) and protein degradation by expressions of proteasome 20S subunit beta 2 and calpain-1 catalytic subunit in SM biopsies. Results: TNF and IL-10 expression was higher in cCHF than in ncCHF and controls (all p < 0.004). cCHF had a lower fat mass index (FMI) and lower fat-free mass index (FFMI) compared to ncCHF and controls (p < 0.05). STAT1 and STAT3 expression was higher in cCHF vs. ncCHF or controls (1.1 [1.6] vs. 0.8 [0.9] vs. 0.9 [1.1] RU and 4.6 [5.5] vs. 2.5 [4.8] vs. 3.0 [4.2] RU, all ANOVA-p < 0.05). The same applied for SOCS1 and SOCS3 expression (1.1 [1.5] vs. 0.4 [0.4] vs. 0.4 [0.5] and 0.9 [3.3] vs. 0.4 [1.1] vs. 0.8 [0.9] RU, all ANOVA-p < 0.04). In cCHF, higher TNF and STAT1 expression was associated with lower FMI (r = 0.5, p = 0.053 and p < 0.05) but not with lower FFMI (p > 0.4). In ncCHF, neither cytokine nor STAT/SOCS expression was associated with BC (all p > 0.3). SLC2A4 was upregulated in SM of cCHF vs. ncCHF (p < 0.03). Conclusions: Increased STAT1, STAT3, SOCS1 and SOCS3 expression suggests their involvement in CC. In cCHF, higher TNF and STAT-1 expression in PBC were associated with lower FMI. Increased SLC2A4 in cachectic SM biopsies indicates altered glucose metabolism.
Collapse
Affiliation(s)
- Anja Sandek
- Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Christoph Gertler
- Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Miroslava Valentova
- Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Nadja Jauert
- Centre for Stroke Research Berlin, Charité-University Medicine Berlin, Corporate Member of Free University Berlin and Humboldt-University Berlin, 10117 Berlin, Germany
- Division of Physiology, Department of Human Medicine, MSB Medical School Berlin, Rüdesheimerstr 50, 14197 Berlin, Germany
| | - Manuel Wallbach
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Wolfram Doehner
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, German Heart Center Charité, Charité-University Medicine Berlin, Corporate Member of Free University Berlin and Humboldt-University Berlin, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 13353 Berlin, Germany
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Stefan D Anker
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, German Heart Center Charité, Charité-University Medicine Berlin, Corporate Member of Free University Berlin and Humboldt-University Berlin, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 13353 Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Charité-University Medicine Berlin, Corporate Member of Free University Berlin and Humboldt-University Berlin, 10117 Berlin, Germany
| | - Jens Fielitz
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, 17475 Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, 17475 Greifswald, Germany
| | - Hans-Dieter Volk
- BIH Center for Regenerative Therapies (BCRT), Charité-University Medicine Berlin, Corporate Member of Free University Berlin and Humboldt-University Berlin, 10117 Berlin, Germany
- Department of Medical Immunology, Charité-University Medicine Berlin, Corporate Member of Free University Berlin and Humboldt-University Berlin, 10117 Berlin, Germany
| |
Collapse
|
9
|
Lloyd EM, Pinniger GJ, Murphy RM, Grounds MD. Slow or fast: Implications of myofibre type and associated differences for manifestation of neuromuscular disorders. Acta Physiol (Oxf) 2023; 238:e14012. [PMID: 37306196 DOI: 10.1111/apha.14012] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Many neuromuscular disorders can have a differential impact on a specific myofibre type, forming the central premise of this review. The many different skeletal muscles in mammals contain a spectrum of slow- to fast-twitch myofibres with varying levels of protein isoforms that determine their distinctive contractile, metabolic, and other properties. The variations in functional properties across the range of classic 'slow' to 'fast' myofibres are outlined, combined with exemplars of the predominantly slow-twitch soleus and fast-twitch extensor digitorum longus muscles, species comparisons, and techniques used to study these properties. Other intrinsic and extrinsic differences are discussed in the context of slow and fast myofibres. These include inherent susceptibility to damage, myonecrosis, and regeneration, plus extrinsic nerves, extracellular matrix, and vasculature, examined in the context of growth, ageing, metabolic syndrome, and sexual dimorphism. These many differences emphasise the importance of carefully considering the influence of myofibre-type composition on manifestation of various neuromuscular disorders across the lifespan for both sexes. Equally, understanding the different responses of slow and fast myofibres due to intrinsic and extrinsic factors can provide deep insight into the precise molecular mechanisms that initiate and exacerbate various neuromuscular disorders. This focus on the influence of different myofibre types is of fundamental importance to enhance translation for clinical management and therapies for many skeletal muscle disorders.
Collapse
Affiliation(s)
- Erin M Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Gavin J Pinniger
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria, Australia
| | - Miranda D Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
10
|
Gortan Cappellari G, Guillet C, Poggiogalle E, Ballesteros Pomar MD, Batsis JA, Boirie Y, Breton I, Frara S, Genton L, Gepner Y, Gonzalez MC, Heymsfield SB, Kiesswetter E, Laviano A, Prado CM, Santini F, Serlie MJ, Siervo M, Villareal DT, Volkert D, Voortman T, Weijs PJ, Zamboni M, Bischoff SC, Busetto L, Cederholm T, Barazzoni R, Donini LM. Sarcopenic obesity research perspectives outlined by the sarcopenic obesity global leadership initiative (SOGLI) - Proceedings from the SOGLI consortium meeting in rome November 2022. Clin Nutr 2023; 42:687-699. [PMID: 36947988 DOI: 10.1016/j.clnu.2023.02.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 02/17/2023] [Indexed: 02/26/2023]
Abstract
The European Society for Clinical Nutrition and Metabolism (ESPEN) and the European Association for the Study of Obesity (EASO) launched the Sarcopenic Obesity Global Leadership Initiative (SOGLI) to reach expert consensus on a definition and diagnostic criteria for Sarcopenic Obesity (SO). The present paper describes the proceeding of the Sarcopenic Obesity Global Leadership Initiative (SOGLI) meeting that was held on November 25th and 26th, 2022 in Rome, Italy. This consortium involved the participation of 50 researchers from different geographic regions and countries. The document outlines an agenda advocated by the SOGLI expert panel regarding the pathophysiology, screening, diagnosis, staging and treatment of SO that needs to be prioritized for future research in the field.
Collapse
Affiliation(s)
| | - Christelle Guillet
- University of Clermont Auvergne, INRA, CRNH, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | | | | | - John A Batsis
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yves Boirie
- University of Clermont Auvergne, INRA, CRNH, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Irene Breton
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Stefano Frara
- Università Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | | | | | | | - Eva Kiesswetter
- Institute for Evidence in Medicine, Medical Center & Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | | | | | | | | | | | - Dorothee Volkert
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Nuremberg, Germany
| | - Trudy Voortman
- Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Peter Jm Weijs
- Amsterdam University Medical Centers, Amsterdam, the Netherlands; Amsterdam University of Applied Sciences, Amsterdam, Netherlands
| | | | | | | | - Tommy Cederholm
- Uppsala University and Karolinska University Hospital, Stockholm, Sweden
| | - Rocco Barazzoni
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | | |
Collapse
|
11
|
Miyazaki M, Sawada A, Sawamura D, Yoshida S. Decreased insulin-like growth factor-1 expression in response to mechanical loading is associated with skeletal muscle anabolic resistance in cancer cachexia. Growth Horm IGF Res 2023; 69-70:101536. [PMID: 37229943 DOI: 10.1016/j.ghir.2023.101536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 05/07/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
OBJECTIVE Cachexia is a systemic metabolic syndrome characterized by loss of body weight and skeletal muscle mass during chronic wasting diseases, such as cancer. Skeletal muscle in cancer cachexia is less responsive to anabolic factors including mechanical loading; however, the precise molecular mechanism is largely unknown. In this study, we examined the underlying mechanism of anabolic resistance in skeletal muscle in a cancer cachexia model. METHODS CD2F1 mice (male, 8 weeks old) were subcutaneously transplanted (1 × 106 cells per mouse) with a mouse colon cancer-derived cell line (C26) as a model of cancer cachexia. Mechanical overload of the plantaris muscle by synergist tenotomy was performed during the 2nd week and the plantaris muscle was sampled at the 4th week following C26 transplantation. RESULTS The hypertrophic response of skeletal muscle (increased skeletal muscle weight/protein synthesis efficiency and activation of mechanistic target of rapamycin complex 1 signaling) associated with mechanical overload was significantly suppressed during cancer cachexia. Screening of gene expression profile and pathway analysis using microarray revealed that blunted muscle protein synthesis was associated with cancer cachexia and was likely induced by downregulation of insulin-like growth factor-1 (IGF-1) and impaired activation of IGF-1-dependent signaling. CONCLUSIONS These observations indicate that cancer cachexia induces resistance to muscle protein synthesis, which may be a factor for inhibiting the anabolic adaptation of skeletal muscle to physical exercise in cancer patients.
Collapse
Affiliation(s)
- Mitsunori Miyazaki
- Department of Integrative Physiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan; Department of Physical Therapy, School of Rehabilitation Sciences, Health Sciences University of Hokkaido, Japan.
| | - Atsushi Sawada
- Department of Physical Therapy, School of Rehabilitation Sciences, Health Sciences University of Hokkaido, Japan
| | - Daisuke Sawamura
- Department of Rehabilitation Science, Faculty of Health Sciences, Hokkaido University, Japan
| | - Susumu Yoshida
- Department of Physical Therapy, School of Rehabilitation Sciences, Health Sciences University of Hokkaido, Japan
| |
Collapse
|
12
|
Boran T, Zengin OS, Seker Z, Akyildiz AG, Oztas E, Özhan G. Ripretinib induced skeletal muscle toxicity through mitochondrial impairment in C2C12 myotubes. Toxicology 2023; 489:153489. [PMID: 36933644 DOI: 10.1016/j.tox.2023.153489] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
Ripretinib is a multikinase inhibitor drug approved in 2020 by the FDA and in 2021 by EMA for use in the treatment of advanced gastrointestinal stromal tumors (GIST) which have not adequately responded to previous treatments with kinase inhibitors. The most common side effects of the drug are myalgia and fatigue, which likely causes interruption of the treatment or reduction of the dose. Skeletal muscle cells highly depend on ATP to perform their functions and mitochondrial damage may play a role in skeletal muscle toxicity induced by kinase inhibitors. However, the molecular mechanism has not been clearly identified in the literature yet. In this study, it has been aimed to elucidate the role of mitochondria in the toxic effect of ripretinib on skeletal muscle using the mouse C2C12 myoblast-derived myotubes. The myotubes were exposed to ripretinib at the range of 1-20 μM concentrations for 24 h. To determine the potential role of mitochondrial impairment in ripretinib-induced skeletal muscle toxicity, intracellular ATP level, mitochondrial membrane potential (MMP), mitochondrial ROS production (mtROS), mitochondrial DNA (mtDNA) copy number, and mitochondrial mass were examined after ripretinib treatment. Furthermore, changes in PGC 1α/NRF 1/NRF 2 expression levels that play a role in mitochondrial biogenesis and mitophagy were investigated. Additionally, the mitochondrial electron transport chain (ETC) enzyme activities were evaluated. Lastly, a molecular docking study was done to see ripretinib's possible interaction with DNA polymerase gamma (POLG) which is important for DNA replication in the mitochondria. According to the findings, ripretinib decreases the ATP level and mtDNA copy number, induces loss of MMP, and reduces mitochondrial mass. The activities of the ETC complexes were inhibited with ripretinib exposure which is in line with the observed ATP depletion and MMP loss. The molecular docking study revealed that ripretinib has inhibitory potential against POLG which supports the observed inhibition of mtDNA. The expression of PGC 1α was reduced in the nuclear fraction indicating that PGC-1α was not activated since the NRF 1 expression was reduced and NRF 2 level did not show significant change. Consequently, mtROS production increased in all treatment groups and mitophagy-related gene expressions and Parkin protein expression level were up-regulated at high doses. In conclusion, mitochondrial damage/loss can be one of the underlying causes of ripretinib-induced skeletal muscle toxicity. However, further studies are needed to confirm the results in vivo.
Collapse
Affiliation(s)
- Tugce Boran
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey; Istanbul University-Cerrahpaşa, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34500 Istanbul, Turkey
| | - Ozge Sultan Zengin
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey; Institute of Graduate Studies in Health Sciences, Istanbul University, 34116 Istanbul, Turkey
| | - Zehra Seker
- Institute of Graduate Studies in Health Sciences, Istanbul University, 34116 Istanbul, Turkey; Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34093 Istanbul, Turkey
| | - Aysenur Gunaydin Akyildiz
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34093 Istanbul, Turkey
| | - Ezgi Oztas
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey
| | - Gül Özhan
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey.
| |
Collapse
|
13
|
Meanti R, Licata M, Rizzi L, Bresciani E, Molteni L, Coco S, Locatelli V, Omeljaniuk RJ, Torsello A. Protective Effects of Hexarelin and JMV2894 in a Human Neuroblastoma Cell Line Expressing the SOD1-G93A Mutated Protein. Int J Mol Sci 2023; 24:ijms24020993. [PMID: 36674509 PMCID: PMC9863688 DOI: 10.3390/ijms24020993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 01/07/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable motor neuron disease whose etiology remains unresolved; nonetheless, mutations of superoxide dismutase 1 (SOD1) have been associated with several variants of ALS. Currently available pharmacologic interventions are only symptomatic and palliative in effect; therefore, there is a pressing demand for more effective drugs. This study examined potential therapeutic effects of growth hormone secretagogues (GHSs), a large family of synthetic compounds, as possible candidates for the treatment of ALS. Human neuroblastoma cells expressing the SOD1-G93A mutated protein (SH-SY5Y SOD1G93A cells) were incubated for 24 h with H2O2 (150 µM) in the absence, or presence, of GHS (1 µM), in order to study the protective effect of GHS against increased oxidative stress. The two GHSs examined in this study, hexarelin and JMV2894, protected cells from H2O2-induced cytotoxicity by activating molecules that regulate apoptosis and promote cell survival processes. These findings suggest the possibility of developing new GHS-based anti-oxidant and neuroprotective drugs with improved therapeutic potential. Further investigations are required for the following: (i) to clarify GHS molecular mechanisms of action, and (ii) to envisage the development of new GHSs that may be useful in ALS therapy.
Collapse
Affiliation(s)
- Ramona Meanti
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Monza, Italy
| | - Martina Licata
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Monza, Italy
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Monza, Italy
- Correspondence: ; Tel.: +39-02-6448-8224
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Monza, Italy
| | - Laura Molteni
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Monza, Italy
| | - Silvia Coco
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Monza, Italy
| | - Vittorio Locatelli
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Monza, Italy
| | | | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Monza, Italy
| |
Collapse
|
14
|
VanderVeen BN, Cardaci TD, McDonald SJ, Madero SS, Unger CA, Bullard BM, Enos RT, Velázquez KT, Kubinak JL, Fan D, Murphy EA. Obesity reduced survival with 5-fluorouracil and did not protect against chemotherapy-induced cachexia or immune cell cytotoxicity in mice. Cancer Biol Ther 2022; 23:1-15. [PMID: 35968771 PMCID: PMC9377261 DOI: 10.1080/15384047.2022.2108306] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/28/2022] [Accepted: 07/24/2022] [Indexed: 01/12/2023] Open
Abstract
Fluorouracil/5-flourouracil (5FU) is a first-line chemotherapy drug for many cancer types; however, its associated toxicities contribute to poor quality of life and reduced dose intensities negatively impacting patient prognosis. While obesity remains a critical risk factor for most cancers, our understanding regarding how obesity may impact chemotherapy's toxicities is extremely limited. C56BL/6 mice were given high fat (Obese) or standard diets (Lean) for 4 months and then subjected to three cycles of 5FU (5d-40 mg/kg Lean Mass, 9d rest) or PBS vehicle control. Shockingly, only 60% of Obese survived 3 cycles compared to 100% of Lean, and Obese lost significantly more body weight. Dihydropyrimidine dehydrogenase (DPD), the enzyme responsible for 5FU catabolism, was reduced in obese livers. Total white blood cells, neutrophils, and lymphocytes were reduced in Obese 5FU compared to Lean 5FU and PBS controls. While adipocyte size was not affected by 5FU in Obese, skeletal muscle mass and myofibrillar cross section area were decreased following 5FU in Lean and Obese. Although adipose tissue inflammatory gene expression was not impacted by 5FU, distinct perturbations to skeletal muscle inflammatory gene expression and immune cell populations (CD45+ Immune cells, CD45+CD11b+CD68+ macrophages and CD45+CD11b+Ly6clo/int macrophage/monocytes) were observed in Obese only. Our evidence suggests that obesity induced liver pathologies and reduced DPD exacerbated 5FU toxicities. While obesity has been suggested to protect against cancer/chemotherapy-induced cachexia and other toxicities, our results demonstrate that obese mice are not protected, but rather show evidence of increased susceptibility to 5FU-induced cytotoxicity even when dosed for relative lean mass.
Collapse
Affiliation(s)
- Brandon N. VanderVeen
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Thomas D. Cardaci
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Sierra J. McDonald
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Sarah S. Madero
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Christian A. Unger
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Brooke M. Bullard
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Reilly T. Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Kandy T. Velázquez
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Jason L. Kubinak
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| | - E. Angela Murphy
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine – Columbia, Columbia, SC, USA
| |
Collapse
|
15
|
VanderVeen BN, Cardaci TD, Cunningham P, McDonald SJ, Bullard BM, Fan D, Murphy EA, Velázquez KT. Quercetin Improved Muscle Mass and Mitochondrial Content in a Murine Model of Cancer and Chemotherapy-Induced Cachexia. Nutrients 2022; 15:102. [PMID: 36615760 PMCID: PMC9823918 DOI: 10.3390/nu15010102] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
A cachexia diagnosis is associated with a doubling in hospital stay and increased healthcare cost for cancer patients and most cachectic patients do not survive treatment. Unfortunately, complexity in treating cachexia is amplified by both the underlying malignancy and the anti-cancer therapy which can independently promote cachexia. Quercetin, an organic polyphenolic flavonoid, has demonstrated anti-inflammatory and antioxidant properties with promise in protecting against cancer and chemotherapy-induced dysfunction; however, whether quercetin is efficacious in maintaining muscle mass in tumor-bearing animals receiving chemotherapy has not been investigated. C26 tumor-bearing mice were given 5-fluorouracil (5FU; 30 mg/kg of lean mass i.p.) concomitant with quercetin (Quer; 50 mg/kg of body weight via oral gavage) or vehicle. Both C26 + 5FU and C26 + 5FU + Quer had similar body weight loss; however, muscle mass and cross-sectional area was greater in C26 + 5FU + Quer compared to C26 + 5FU. Additionally, C26 + 5FU + Quer had a greater number and larger intermyofibrillar mitochondria with increased relative protein expression of mitochondrial complexes V, III, and II as well as cytochrome c expression. C26 + 5FU + Quer also had increased MFN1 and reduced FIS1 relative protein expression without apparent benefits to muscle inflammatory signaling. Our data suggest that quercetin protected against cancer and chemotherapy-induced muscle mass loss through improving mitochondrial homeostatic balance.
Collapse
Affiliation(s)
- Brandon N. VanderVeen
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
- AcePre, LLC, Columbia, SC 29209, USA
| | - Thomas D. Cardaci
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Patrice Cunningham
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Sierra J. McDonald
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Brooke M. Bullard
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Daping Fan
- AcePre, LLC, Columbia, SC 29209, USA
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - E. Angela Murphy
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
- AcePre, LLC, Columbia, SC 29209, USA
| | - Kandy T. Velázquez
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| |
Collapse
|
16
|
Di Girolamo D, Tajbakhsh S. Pathological features of tissues and cell populations during cancer cachexia. CELL REGENERATION 2022; 11:15. [PMID: 35441960 PMCID: PMC9021355 DOI: 10.1186/s13619-022-00108-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/28/2021] [Indexed: 11/10/2022]
Abstract
Cancers remain among the most devastating diseases in the human population in spite of considerable advances in limiting their impact on lifespan and healthspan. The multifactorial nature of cancers, as well as the number of tissues and organs that are affected, have exposed a considerable diversity in mechanistic features that are reflected in the wide array of therapeutic strategies that have been adopted. Cachexia is manifested in a number of diseases ranging from cancers to diabetes and ageing. In the context of cancers, a majority of patients experience cachexia and succumb to death due to the indirect effects of tumorigenesis that drain the energy reserves of different organs. Considerable information is available on the pathophysiological features of cancer cachexia, however limited knowledge has been acquired on the resident stem cell populations, and their function in the context of these diseases. Here we review current knowledge on cancer cachexia and focus on how tissues and their resident stem and progenitor cell populations are individually affected.
Collapse
|
17
|
Chen TH, Koh KY, Lin KMC, Chou CK. Mitochondrial Dysfunction as an Underlying Cause of Skeletal Muscle Disorders. Int J Mol Sci 2022; 23:12926. [PMID: 36361713 PMCID: PMC9653750 DOI: 10.3390/ijms232112926] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 09/19/2023] Open
Abstract
Mitochondria are an important energy source in skeletal muscle. A main function of mitochondria is the generation of ATP for energy through oxidative phosphorylation (OXPHOS). Mitochondrial defects or abnormalities can lead to muscle disease or multisystem disease. Mitochondrial dysfunction can be caused by defective mitochondrial OXPHOS, mtDNA mutations, Ca2+ imbalances, mitochondrial-related proteins, mitochondrial chaperone proteins, and ultrastructural defects. In addition, an imbalance between mitochondrial fusion and fission, lysosomal dysfunction due to insufficient biosynthesis, and/or defects in mitophagy can result in mitochondrial damage. In this review, we explore the association between impaired mitochondrial function and skeletal muscle disorders. Furthermore, we emphasize the need for more research to determine the specific clinical benefits of mitochondrial therapy in the treatment of skeletal muscle disorders.
Collapse
Affiliation(s)
- Tsung-Hsien Chen
- Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan
| | - Kok-Yean Koh
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan
| | - Kurt Ming-Chao Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Chu-Kuang Chou
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan
- Obesity Center, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan
| |
Collapse
|
18
|
Gao S, Zhang G, Zhang Z, Zhu JZ, Li L, Zhou Y, Rodney GG, Abo-Zahrah RS, Anderson L, Garcia JM, Kwon YT, Li YP. UBR2 targets myosin heavy chain IIb and IIx for degradation: Molecular mechanism essential for cancer-induced muscle wasting. Proc Natl Acad Sci U S A 2022; 119:e2200215119. [PMID: 36252004 PMCID: PMC9618047 DOI: 10.1073/pnas.2200215119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 08/06/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer cachexia is a lethal metabolic syndrome featuring muscle wasting with preferential loss of fast-twitching muscle mass through an undefined mechanism. Here, we show that cancer induces muscle wasting by selectively degrading myosin heavy chain (MHC) subtypes IIb and IIx through E3 ligase UBR2-mediated ubiquitylation. Induction of MHC loss and atrophy in C2C12 myotubes and mouse tibialis anterior (TA) by murine cancer cells required UBR2 up-regulation by cancer. Genetic gain or loss of UBR2 function inversely altered MHC level and muscle mass in TA of tumor-free mice. UBR2 selectively interacted with and ubiquitylated MHC-IIb and MHC-IIx through its substrate recognition and catalytic domain, respectively, in C2C12 myotubes. Elevation of UBR2 in muscle of tumor-bearing or free mice caused loss of MHC-IIb and MHC-IIx but not MHC-I and MHC-IIa or other myofibrillar proteins, including α-actin, troponin, tropomyosin, and tropomodulin. Muscle-specific knockout of UBR2 spared KPC tumor-bearing mice from losing MHC-IIb and MHC-IIx, fast-twitching muscle mass, cross-sectional area, and contractile force. The rectus abdominis (RA) muscle of patients with cachexia-prone cancers displayed a selective reduction of MHC-IIx in correlation with higher UBR2 levels. These data suggest that UBR2 is a regulator of MHC-IIb/IIx essential for cancer-induced muscle wasting, and that therapeutic interventions can be designed by blocking UBR2 up-regulation by cancer.
Collapse
Affiliation(s)
- Song Gao
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Guohua Zhang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Zicheng Zhang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - James Z. Zhu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Li Li
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Yong Zhou
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - George G. Rodney
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030
| | - Reem S. Abo-Zahrah
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030
| | - Lindsey Anderson
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA98018
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA98108
| | - Jose M. Garcia
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA98018
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA98108
| | - Yong Tae Kwon
- World Class University Program, Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Medicine, Seoul National University, Seoul 110-799, Korea
| | - Yi-Ping Li
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
19
|
Wu HY, Trevino JG, Fang BL, Riner AN, Vudatha V, Zhang GH, Li YP. Patient-Derived Pancreatic Cancer Cells Induce C2C12 Myotube Atrophy by Releasing Hsp70 and Hsp90. Cells 2022; 11:cells11172756. [PMID: 36078164 PMCID: PMC9455268 DOI: 10.3390/cells11172756] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 12/02/2022] Open
Abstract
Pancreatic cancer (PC) patients are highly prone to cachexia, a lethal wasting syndrome featuring muscle wasting with an undefined etiology. Recent data indicate that certain murine cancer cells induce muscle wasting by releasing Hsp70 and Hsp90 through extracellular vesicles (EVs) to activate p38β MAPK-mediated catabolic pathways primarily through Toll-like receptor 4 (TLR4). However, whether human PC induces cachexia through releasing Hsp70 and Hsp90 is undetermined. Here, we investigated whether patient-derived PC cells induce muscle cell atrophy directly through this mechanism. We compared cancer cells isolated from patient-derived xenografts (PDX) from three PC patients who had cachexia (PCC) with those of three early-stage lung cancer patients without cachexia (LCC) and two renal cancer patients who were not prone to cachexia (RCC). We observed small increases of Hsp70 and Hsp90 released by LCC and RCC in comparison to non-cancer control cells (NCC). However, PCC released markedly higher levels of Hsp70 and Hsp90 (~ 6-fold on average) than LCC and RCC. In addition, PCC released similarly increased levels of Hsp70/90-containing EVs. In contrast to RCC and LCC, PCC-conditioned media induced a potent catabolic response in C2C12 myotubes including the activation of p38 MAPK and transcription factor C/EBPβ, upregulation of E3 ligases UBR2 and MAFbx, and increase of autophagy marker LC3-II, resulting in the loss of the myosin heavy chain (MHC ~50%) and myotube diameter (~60%). Importantly, the catabolic response was attenuated by Hsp70- and Hsp90-neutralizing antibodies in a dose-dependent manner. These data suggest that human PC cells release high levels of Hsp70 and Hsp90 that induce muscle atrophy through a direct action on muscle cells.
Collapse
Affiliation(s)
- Hong-Yu Wu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX 77030, USA
| | - Jose G. Trevino
- Department of Surgery, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Bing-Liang Fang
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrea N. Riner
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - Vignesh Vudatha
- Department of Surgery, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Guo-Hua Zhang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX 77030, USA
| | - Yi-Ping Li
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-(713)-500-6498; Fax: +1-(713)-500-0689
| |
Collapse
|
20
|
Zhang B, Bi Q, Huang S, Lv S, Zong X, Wang M, Ji X. Baoyuan Jiedu decoction alleviating cancer cachexia–Induced muscle atrophy by regulating muscle mitochondrial function in ApcMin/+ mice. Front Pharmacol 2022; 13:914597. [PMID: 36060011 PMCID: PMC9437209 DOI: 10.3389/fphar.2022.914597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer cachexia is a complex syndrome that leads to an ongoing loss of skeletal muscle mass in many malignant tumors. Our previous studies have evaluated the effectiveness of Baoyuan Jiedu decoction (BJD) in alleviating cancer-induced muscle atrophy. However, the mechanisms of BJD regulating muscle atrophy could not be fully understood. Therefore, we further investigated the mechanisms of BJD mitigating muscle atrophy both in an ApcMin/+ mouse model and the Lewis-conditioned medium–induced C2C12 myotube atrophy model. We confirmed the quality of BJD extracts by HPLC. In an In vivo study, body weight loss and muscle atrophy were alleviated with BJD treatment. GO analysis suggested that ATP metabolism and mitochondria were involved. The results of the electron microscope show that BJD treatment may have a healing effect on mitochondrial structure. Moreover, ATP content and mitochondrial numbers were improved with BJD treatment. Furthermore, both in vivo and in vitro, we demonstrated that the BJD treatment could improve mitochondrial function owing to the increased number of mitochondria, balanced dynamic, and regulation of the electron transport chain according to the protein and mRNA expressions. In addition, oxidative stress caused by mitochondrial dysfunction was ameliorated by BJD treatment in ApcMin/+ mice. Consequently, our study provides proof for BJD treatment alleviating cancer cachexia–induced muscle atrophy by modulating mitochondrial function in ApcMin/+ mice.
Collapse
Affiliation(s)
- Beiying Zhang
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qianyu Bi
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shengqi Huang
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Siyuan Lv
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Zong
- Weifang Nursing Vocational College, Weifang, China
| | - Mengran Wang
- Department of Pediatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuming Ji
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Xuming Ji,
| |
Collapse
|
21
|
Halle JL, Counts BR, Zhang Q, Carson JA. Short duration treadmill exercise improves physical function and skeletal muscle mitochondria protein expression after recovery from FOLFOX chemotherapy in male mice. FASEB J 2022; 36:e22437. [PMID: 35816153 DOI: 10.1096/fj.202200460r] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/10/2022] [Accepted: 06/21/2022] [Indexed: 01/05/2023]
Abstract
FOLFOX (5-FU, leucovorin, oxaliplatin) is a chemotherapy treatment for colorectal cancer which induces toxic side effects involving fatigue, weakness, and skeletal muscle dysfunction. There is a limited understanding of the recovery from these toxicities after treatment cessation. Exercise training can improve chemotherapy-related toxicities. However, how exercise accelerates recovery and the dose required for these benefits are not well examined. The purpose of this study was to examine the effect of exercise duration on physical function, muscle mass, and mitochondria protein expression during the recovery from FOLFOX chemotherapy. 12-week-old male mice were administered four cycles of either PBS or FOLFOX over 8-weeks. Outcomes were assessed after the fourth cycle and after either 4 (short-term; STR) or 10 weeks (long-term; LTR) recovery. Subsets of mice performed 14 sessions (6 d/wk, 18 m/min, 5% grade) of 60 min/d (long) or 15 min/d (short duration) treadmill exercise during STR. Red and white gastrocnemius mRNA and protein expression were examined. FOLFOX treatment decreased run time (RT) (-53%) and grip strength (GS) (-9%) compared to PBS. FOLFOX also reduced muscle OXPHOS complexes, COXIV, and VDAC protein expression. At LTR, FOLFOX RT (-36%) and GS (-16%) remained reduced. Long- and short-duration treadmill exercise improved RT (+58% and +56%) without restoring GS in FOLFOX mice. Both exercise durations increased muscle VDAC and COXIV expression in FOLFOX mice. These data provide evidence that FOLFOX chemotherapy induces persistent deficits in physical function that can be partially reversed by short-duration aerobic exercise.
Collapse
Affiliation(s)
- Jessica L Halle
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Brittany R Counts
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Quan Zhang
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James A Carson
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
22
|
Wang R, Kumar B, Doud EH, Mosley AL, Alexander MS, Kunkel LM, Nakshatri H. Skeletal muscle-specific overexpression of miR-486 limits mammary tumor-induced skeletal muscle functional limitations. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:231-248. [PMID: 35402076 PMCID: PMC8971682 DOI: 10.1016/j.omtn.2022.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/12/2022] [Indexed: 11/28/2022]
Abstract
miR-486 is a myogenic microRNA, and its reduced skeletal muscle expression is observed in muscular dystrophy. Transgenic overexpression of miR-486 using muscle creatine kinase promoter (MCK-miR-486) partially rescues muscular dystrophy phenotype. We had previously demonstrated reduced circulating and skeletal muscle miR-486 levels with accompanying skeletal muscle defects in mammary tumor models. To determine whether skeletal muscle miR-486 is functionally similar in dystrophies and cancer, we performed functional limitations and biochemical studies of skeletal muscles of MMTV-Neu mice that mimic HER2+ breast cancer and MMTV-PyMT mice that mimic luminal subtype B breast cancer and these mice crossed to MCK-miR-486 mice. miR-486 significantly prevented tumor-induced reduction in muscle contraction force, grip strength, and rotarod performance in MMTV-Neu mice. In this model, miR-486 reversed cancer-induced skeletal muscle changes, including loss of p53, phospho-AKT, and phospho-laminin alpha 2 (LAMA2) and gain of hnRNPA0 and SRSF10 phosphorylation. LAMA2 is a part of the dystrophin-associated glycoprotein complex, and its loss of function causes congenital muscular dystrophy. Complementing these beneficial effects on muscle, miR-486 indirectly reduced tumor growth and improved survival, which is likely due to systemic effects of miR-486 on production of pro-inflammatory cytokines such as IL-6. Thus, similar to dystrophy, miR-486 has the potential to reverse skeletal muscle defects and cancer burden.
Collapse
Affiliation(s)
- Ruizhong Wang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brijesh Kumar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Emma H. Doud
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Amber L. Mosley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Matthew S. Alexander
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham and Children’s of Alabama, Birmingham, AL 35294, USA
| | - Louis M. Kunkel
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
23
|
Zhang S, Gong F, Liu J, Liu T, Yang J, Hu J. A novel PHD2 inhibitor acteoside from Cistanche tubulosa induces skeletal muscle mitophagy to improve cancer-related fatigue. Biomed Pharmacother 2022; 150:113004. [PMID: 35658245 DOI: 10.1016/j.biopha.2022.113004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 11/02/2022] Open
Abstract
OBJECTIVE To study whether ACT exerts anti-fatigue activity against CRF by inducing skeletal muscle mitophagy via suppressing PHD2 to upregulate the HIF-1α/BNIP3 signaling pathway. METHODS In this study, the molecular docking virtual screening technique was used to screen active components in Cistanche tubulosa that act as potential PHD2 inhibitors; the preliminary verification was carried out by Surface plasmon resonance (SPR) technology. BALB/c mice were treated with Paclitaxel (PTX, 10 mg/kg) and ACT (50, 100 mg/kg) alone or in combination for 20 days. Fatigue-related behaviors, energy metabolism and skeletal muscle mitochondria were assessed. Murine C2C12 myoblast was cultured and differentiated; then, a C26 tumor cell-conditioned medium was added to induce cachexia. Intracellular reactive oxygen species (ROS), mitochondrial membrane potential, mitochondrial microstructure and function, autophagy, PHD2/HIF-1 and PINK1/Parkin signal pathway proteins were analyzed. Then, interfering RNA technology was used to silence PHD2 and observe the efficacy of ACT. RESULTS We demonstrated that ACT exerted good binding activity with PHD2; ACT administration ameliorated PTX-induced muscle fatigue-like behavior via improving muscle quality and mitochondria function, increasing mitophagy, upregulating COXIV, CytoC, PINK1, Parkin, HIF-1α and BNIP3 expression and inhibiting p62, LC3B, PHD2 and Beclin-1 expression. The protective effect of ACT disappeared after transfection with the PHD2 gene knockdown plasmid Egln-1-RNAi. CONCLUSIONS These results suggest that ACT can improve CRF by promoting mitophagy via suppression of PHD2 to remove dysfunctional mitochondria, demonstrating that ACT has huge prospects for clinical application in CRF treatment.
Collapse
Affiliation(s)
- Shilei Zhang
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China; Department of Toxicology, School of Public Health, Xinjiang Medical University, Urumqi, China.
| | - Fukai Gong
- Department of Pharmacy, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China.
| | - Jiali Liu
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China.
| | - Tao Liu
- Department of Toxicology, School of Public Health, Xinjiang Medical University, Urumqi, China.
| | - Jianhua Yang
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China; Department of Pharmacy, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China.
| | - Junping Hu
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
24
|
Wang T. Searching for the link between inflammaging and sarcopenia. Ageing Res Rev 2022; 77:101611. [PMID: 35307560 DOI: 10.1016/j.arr.2022.101611] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/14/2022] [Accepted: 03/15/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Tiantian Wang
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
25
|
Li J, Xie Q, Liu L, Cheng Y, Han Y, Chen X, Lin J, Li Z, Liu H, Zhang X, Chen H, Peng J, Shen A. Swimming Attenuates Muscle Wasting and Mediates Multiple Signaling Pathways and Metabolites in CT-26 Bearing Mice. Front Mol Biosci 2022; 8:812681. [PMID: 35127824 PMCID: PMC8811507 DOI: 10.3389/fmolb.2021.812681] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives: To investigate the effects of swimming on cancer induced muscle wasting and explore its underlying mechanism in CT-26 bearing mice.Methods: BALB/c mice (n = 16) injected with CT-26 cells were divided into two groups, including Tumor group (n = 8) and Swimming group (n = 8). Another 8 un-injected mice were set as Control group. Mice in Swimming group were subjected to physical training for swimming twice per day for 30 min intervals and 6 days per week for a total of 4 weeks. The tumor volume was monitored every 3 days and tumor weight was measured at the end of experiment. The changes of muscle function, pathological and cell apoptosis of quadriceps muscles were further assessed, and its underlying mechanisms were further explored using multiple biological technologies.Results: Swimming obviously alleviated tumor volume and weight in CT-26 bearing mice. Moreover, swimming attenuated the decrease of muscle tension, autonomic activities, and increase of muscle atrophy, pathological ultrastructure, as well as cell apoptosis of quadriceps muscles in CT-26 bearing mice. Furthermore, swimming significantly down-regulated the protein expression of NF-κB, p-NF-κB, TNF-α, IL-1β, IL-6 and Bax, while up-regulated the expression of Bcl-2. Further differential expressed metabolites (DEMs) analysis identified a total of 76 (in anion mode) and 330 (in cationic mode) DEMs in quadriceps muscles of CT-26 bearing mice after swimming, including taurochenodeoxycholic acid, taurocholic acid, ascorbic acid and eicosapentaenoic acid.Conclusion: Swimming attenuates tumor growth and muscle wasting, and by suppressing the activation of NF-κB signaling pathway mediated inflammation, reducing the level of Bax medicated cell apoptosis, as well as modulating multiple metabolites might be the importantly underlying mechanisms.
Collapse
Affiliation(s)
- Jiapeng Li
- The Department of Physical Education, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- School of Physical Education and Sport Science, Fujian Normal University, Fuzhou, China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Liya Liu
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Ying Cheng
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Yuying Han
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Xiaoping Chen
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Jia Lin
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zuanfang Li
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Huixin Liu
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Xiuli Zhang
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Haichun Chen
- School of Physical Education and Sport Science, Fujian Normal University, Fuzhou, China
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
| | - Jun Peng
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
- *Correspondence: Jun Peng, ; Aling Shen,
| | - Aling Shen
- Academy of Integrative Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
- *Correspondence: Jun Peng, ; Aling Shen,
| |
Collapse
|
26
|
Watson EL, Baker LA, Wilkinson TJ, Gould DW, Xenophontos S, Graham-Brown M, Major RW, Ashford RU, Viana JL, Smith AC. Inflammation and physical dysfunction: responses to moderate intensity exercise in chronic kidney disease. Nephrol Dial Transplant 2021; 37:860-868. [PMID: 35090033 DOI: 10.1093/ndt/gfab333] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND People with chronic kidney disease (CKD) experience skeletal muscle wasting, reduced levels of physical function and performance, and chronic systemic inflammation. While it is known that a relationship exists between inflammation and muscle wasting, the association between inflammation and physical function or performance in CKD has not been well studied. Exercise has anti-inflammatory effects, but little is known regarding the effect of moderate intensity exercise. This study aimed to (i) compare systemic and intramuscular inflammation between CKD stage G3b-5 and non-CKD controls; (ii) establish whether a relationship exists between physical performance, exercise capacity and inflammation in CKD; (iii) determine changes in systemic and intramuscular inflammation following 12 weeks of exercise; and (iv) investigate whether improving inflammatory status via training contributes to improvements in physical performance and muscle mass. METHODS This is a secondary analysis of previously collected data. CKD patients stages G3b-5 (n = 84, n = 43 males) and non-CKD controls (n = 26, n = 17 males) underwent tests of physical performance, exercise capacity, muscle strength and muscle size. In addition, a subgroup of CKD participants underwent 12 weeks of exercise training, randomized to aerobic (AE, n = 21) or combined (CE, n = 20) training. Plasma and intramuscular inflammation and myostatin were measured at rest and following exercise. RESULTS Tumour necrosis factor-α was negatively associated with lower $^{^{^{.}}}{\rm V}$O2Peak (P = 0.01), Rectus femoris-cross sectional area (P = 0.002) and incremental shuttle walk test performance (P < 0.001). Interleukin-6 was negatively associated with sit-to-stand 60 performances (P = 0.006) and hand grip strength (P = 0.001). Unaccustomed exercise created an intramuscular inflammatory response that was attenuated following 12 weeks of training. Exercise training did not reduce systemic inflammation, but AE training did significantly reduce mature myostatin levels (P = 0.02). Changes in inflammation were not associated with changes in physical performance. CONCLUSIONS Systemic inflammation may contribute to reduced physical function in CKD. Twelve weeks of exercise training was unable to reduce the level of chronic systemic inflammation in these patients, but did reduce plasma myostatin concentrations. Further research is required to further investigate this.
Collapse
Affiliation(s)
- Emma L Watson
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK.,Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Luke A Baker
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK.,Department of Health Sciences, University of Leicester, Leicester, UK
| | - Tom J Wilkinson
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK.,Department of Health Sciences, University of Leicester, Leicester, UK.,Leicester Biomedical Research Centre, Leicester, UK
| | - Doug W Gould
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK
| | - Soteris Xenophontos
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK.,Department of Health Sciences, University of Leicester, Leicester, UK
| | - Matthew Graham-Brown
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,John Walls Renal Unit, Leicester General Hospital, Leicester, UK
| | - Rupert W Major
- Department of Health Sciences, University of Leicester, Leicester, UK.,John Walls Renal Unit, Leicester General Hospital, Leicester, UK
| | - Robert U Ashford
- Leicester Orthopaedics, University Hospitals of Leicester, Leicester, UK.,Department of Cancer Studies, University of Leicester, Leicester, UK
| | - Joao L Viana
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University Institute of Maia, ISMAI, Portugal
| | - Alice C Smith
- Leicester Kidney Lifestyle Team, University of Leicester, Leicester, UK.,Department of Health Sciences, University of Leicester, Leicester, UK.,Leicester Biomedical Research Centre, Leicester, UK
| |
Collapse
|
27
|
Liu H, Zang P, Lee I(I, Anderson B, Christiani A, Strait‐Bodey L, Breckheimer BA, Storie M, Tewnion A, Krumm K, Li T, Irwin B, Garcia JM. Growth hormone secretagogue receptor-1a mediates ghrelin's effects on attenuating tumour-induced loss of muscle strength but not muscle mass. J Cachexia Sarcopenia Muscle 2021; 12:1280-1295. [PMID: 34264027 PMCID: PMC8517358 DOI: 10.1002/jcsm.12743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 05/11/2021] [Accepted: 06/08/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Ghrelin may ameliorate cancer cachexia (CC) by preventing anorexia, muscle, and fat loss. However, the mechanisms mediating these effects are not fully understood. This study characterizes the pathways involved in muscle mass and strength loss in the Lewis lung carcinoma (LLC)-induced cachexia model, and the effects of ghrelin in mice with or without its only known receptor: the growth hormone secretagogue receptor-1a ((GHSR-1a), Ghsr+/+ and Ghsr-/- ). METHODS Five to 7-month-old male C57BL/6J Ghsr+/+ and Ghsr-/- mice were inoculated with 1 × 106 heat-killed (HK) or live LLC cells (tumour implantation, TI). When tumours were palpable (7 days after TI), tumour-bearing mice were injected with vehicle (T + V) or ghrelin twice/day for 14 days (T + G, 0.8 mg/kg), while HK-treated mice were given vehicle (HK + V). Body weight and grip strength were evaluated before TI and at termination (21 days after TI). Hindlimb muscles were collected for analysis. RESULTS Less pronounced body weight (BW) loss (87.70 ± 0.98% vs. 83.92 ± 1.23%, percentage of baseline BW in tumour-bearing Ghsr+/+ vs. Ghsr-/- , P = 0.008), and lower upregulation of ubiquitin-proteasome system (UPS, MuRF1/Trim63, 5.71 ± 1.53-fold vs. 9.22 ± 1.94-fold-change from Ghsr+/+ HK + V in tumour-bearing Ghsr+/+ vs. Ghsr-/- , P = 0.036) and autophagy markers (Becn1, Atg5, Atg7, tumour-bearing Ghsr+/+ < Ghsr-/- , all P < 0.02) were found in T + V Ghsr+/+ vs. Ghsr-/- mice. Ghrelin attenuated LLC-induced UPS marker upregulation in both genotypes, [Trim63 was decreased from 5.71 ± 1.53-fold to 1.96 ± 0.47-fold in Ghsr+/+ (T + V vs. T + G: P = 0.032) and 9.22 ± 1.94-fold to 4.72 ± 1.06-fold in Ghsr-/- (T + V vs. T + G: P = 0.008)]. Only in Ghsr+/+ mice ghrelin ameliorated LLC-induced grip strength loss [improved from 89.24 ± 3.48% to 97.80 ± 2.31% of baseline (T + V vs. T + G: P = 0.042)], mitophagy markers [Bnip3 was decreased from 2.28 ± 0.56 to 1.38 ± 0.14-fold (T + V vs. T + G: P ≤ 0.05)], and impaired mitochondrial respiration [State 3u improved from 698.23 ± 73.96 to 934.37 ± 95.21 pmol/min (T + V vs. T + G: P ≤ 0.05)], whereas these markers were not improved by ghrelin Ghsr-/- . Compared with Ghsr+/+ , Ghsr-/- tumour-bearing mice also showed decreased response to ghrelin in BW [T + G-treated Ghsr+/+ vs. Ghsr -/- : 91.75 ± 1.05% vs. 86.18 ± 1.13% of baseline BW, P < 0.001)], gastrocnemius (T + G-treated Ghsr+/+ vs. Ghsr-/- : 96.9 ± 2.08% vs. 88.15 ± 1.78% of Ghsr+/+ HK + V, P < 0.001) and quadriceps muscle mass (T + G-treated Ghsr+/+ vs. Ghsr-/- : 96.12 ± 2.31% vs. 88.36 ± 1.94% of Ghsr+/+ HK + V, P = 0.01), and gastrocnemius type IIA (T + G-treated Ghsr+/+ vs. Ghsr-/- : 1250.49 ± 31.72 vs. 1017.62 ± 70.99 μm2 , P = 0.027) and IIB fibre cross-sectional area (T + G-treated Ghsr+/+ vs. Ghsr-/- : 2496.48 ± 116.88 vs. 2183.04 ± 103.43 μm2 , P = 0.024). CONCLUSIONS Growth hormone secretagogue receptor-1a mediates ghrelin's effects on attenuating LLC-induced weakness but not muscle mass loss by modulating the autophagy-lysosome pathway, mitophagy, and mitochondrial respiration.
Collapse
Affiliation(s)
- Haiming Liu
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Pu Zang
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
- Department of EndocrinologyNanjing Jinling HospitalNanjingChina
| | - Ian (In‐gi) Lee
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Barbara Anderson
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Anthony Christiani
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Lena Strait‐Bodey
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Beatrice A. Breckheimer
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Mackenzie Storie
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Alison Tewnion
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Kora Krumm
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Theresa Li
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Brynn Irwin
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Jose M. Garcia
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| |
Collapse
|
28
|
Abulizi A, Hu L, Ma A, Shao FY, Zhu HZ, Lin SM, Shao GY, Xu Y, Ran JH, Li J, Zhou H, Lin DM, Wang LF, Li M, Yang BX. Ganoderic acid alleviates chemotherapy-induced fatigue in mice bearing colon tumor. Acta Pharmacol Sin 2021; 42:1703-1713. [PMID: 33927358 PMCID: PMC8463583 DOI: 10.1038/s41401-021-00669-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/23/2021] [Indexed: 02/02/2023] Open
Abstract
Chemotherapy-related fatigue (CRF) is increasingly being recognized as one of the severe symptoms in patients undergoing chemotherapy, which not only largely reduces the quality of life in patients, but also diminishes their physical and social function. At present, there is no effective drug for preventing and treating CRF. Ganoderic acid (GA), isolated from traditional Chinese medicine Ganoderma lucidum, has shown a variety of pharmacological activities such as anti-tumor, anti-inflammation, immunoregulation, etc. In this study, we investigated whether GA possessed anti-fatigue activity against CRF. CT26 tumor-bearing mice were treated with 5-fluorouracil (5-FU, 30 mg/kg) and GA (50 mg/kg) alone or in combination for 18 days. Peripheral and central fatigue-related behaviors, energy metabolism and inflammatory factors were assessed. We demonstrated that co-administration of GA ameliorated 5-FU-induced peripheral muscle fatigue-like behavior via improving muscle quality and mitochondria function, increasing glycogen content and ATP production, reducing lactic acid content and LDH activity, and inhibiting p-AMPK, IL-6 and TNF-α expression in skeletal muscle. Co-administration of GA also retarded the 5-FU-induced central fatigue-like behavior accompanied by down-regulating the expression of IL-6, iNOS and COX2 in the hippocampus through inhibiting TLR4/Myd88/NF-κB pathway. These results suggest that GA could attenuate 5-FU-induced peripheral and central fatigue in tumor-bearing mice, which provides evidence for GA as a potential drug for treatment of CRF in clinic.
Collapse
Affiliation(s)
- Abudumijiti Abulizi
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Ling Hu
- Department of Anatomy and Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Ang Ma
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Fang-Yu Shao
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Hui-Ze Zhu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Si-Mei Lin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Guang-Ying Shao
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yue Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Jian-Hua Ran
- Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, China
| | - Jing Li
- Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, China
| | - Hong Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Dong-Mei Lin
- JUNCAO Technology Research Institute, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Lian-Fu Wang
- JUNCAO Technology Research Institute, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Min Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Bao-Xue Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China.
| |
Collapse
|
29
|
Renzini A, Riera CS, Minic I, D’Ercole C, Lozanoska-Ochser B, Cedola A, Gigli G, Moresi V, Madaro L. Metabolic Remodeling in Skeletal Muscle Atrophy as a Therapeutic Target. Metabolites 2021; 11:517. [PMID: 34436458 PMCID: PMC8398298 DOI: 10.3390/metabo11080517] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle is a highly responsive tissue, able to remodel its size and metabolism in response to external demand. Muscle fibers can vary from fast glycolytic to slow oxidative, and their frequency in a specific muscle is tightly regulated by fiber maturation, innervation, or external causes. Atrophic conditions, including aging, amyotrophic lateral sclerosis, and cancer-induced cachexia, differ in the causative factors and molecular signaling leading to muscle wasting; nevertheless, all of these conditions are characterized by metabolic remodeling, which contributes to the pathological progression of muscle atrophy. Here, we discuss how changes in muscle metabolism can be used as a therapeutic target and review the evidence in support of nutritional interventions and/or physical exercise as tools for counteracting muscle wasting in atrophic conditions.
Collapse
Affiliation(s)
- Alessandra Renzini
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| | - Carles Sánchez Riera
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| | - Isidora Minic
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| | - Chiara D’Ercole
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| | - Biliana Lozanoska-Ochser
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| | - Alessia Cedola
- Institute of Nanotechnology, c/o Dipartimento di Fisica, National Research Council (CNR-NANOTEC), Sapienza University of Rome, 00185 Rome, Italy;
| | - Giuseppe Gigli
- Institute of Nanotechnology, c/o Campus Ecotekne, National Research Council (CNR-NANOTEC), Monteroni, 73100 Lecce, Italy;
| | - Viviana Moresi
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
- Institute of Nanotechnology, c/o Dipartimento di Fisica, National Research Council (CNR-NANOTEC), Sapienza University of Rome, 00185 Rome, Italy;
| | - Luca Madaro
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, 00185 Rome, Italy; (A.R.); (C.S.R.); (I.M.); (C.D.); (B.L.-O.); (L.M.)
| |
Collapse
|
30
|
Kutz LC, Cui X, Xie JX, Mukherji ST, Terrell KC, Huang M, Wang X, Wang J, Martin AJ, Pessoa MT, Cai L, Zhu H, Heiny JA, Shapiro JI, Blanco G, Xie Z, Pierre SV. The Na/K-ATPase α1/Src interaction regulates metabolic reserve and Western diet intolerance. Acta Physiol (Oxf) 2021; 232:e13652. [PMID: 33752256 PMCID: PMC8570534 DOI: 10.1111/apha.13652] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023]
Abstract
AIM Highly prevalent diseases such as insulin resistance and heart failure are characterized by reduced metabolic flexibility and reserve. We tested whether Na/K-ATPase (NKA)-mediated regulation of Src kinase, which requires two NKA sequences specific to the α1 isoform, is a regulator of metabolic capacity that can be targeted pharmacologically. METHODS Metabolic capacity was challenged functionally by Seahorse metabolic flux analyses and glucose deprivation in LLC-PK1-derived cells expressing Src binding rat NKA α1, non-Src-binding rat NKA α2 (the most abundant NKA isoform in the skeletal muscle), and Src binding gain-of-function mutant rat NKA α2. Mice with skeletal muscle-specific ablation of NKA α1 (skα1-/-) were generated using a MyoD:Cre-Lox approach and were subjected to treadmill testing and Western diet. C57/Bl6 mice were subjected to Western diet with or without pharmacological inhibition of NKA α1/Src modulation by treatment with pNaKtide, a cell-permeable peptide designed by mapping one of the sites of NKA α1/Src interaction. RESULTS Metabolic studies in mutant cell lines revealed that the Src binding regions of NKA α1 are required to maintain metabolic reserve and flexibility. Skα1-/- mice had decreased exercise endurance and mitochondrial Complex I dysfunction. However, skα1-/- mice were resistant to Western diet-induced insulin resistance and glucose intolerance, a protection phenocopied by pharmacological inhibition of NKA α1-mediated Src regulation with pNaKtide. CONCLUSIONS These results suggest that NKA α1/Src regulatory function may be targeted in metabolic diseases. Because Src regulatory capability by NKA α1 is exclusive to endotherms, it may link the aerobic scope hypothesis of endothermy evolution to metabolic dysfunction.
Collapse
Affiliation(s)
- Laura C Kutz
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Jeffrey X. Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Shreya T Mukherji
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Kayleigh C Terrell
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Minqi Huang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Jiayan Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Adam J Martin
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Marco T Pessoa
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Liquan Cai
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Hua Zhu
- Department of Surgery, Wexner Medical Center, Ohio State University, Columbus, OH
| | - Judith A Heiny
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH
| | - Joseph I Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Gustavo Blanco
- Department of Molecular and Integrative Physiology, and The Kidney Institute, University of Kansas Medical Center, Kansas City, KS
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| |
Collapse
|
31
|
Chang SW, Tsai YH, Hsu CM, Huang EI, Chang GH, Tsai MS, Tsai YT. Masticatory muscle index for indicating skeletal muscle mass in patients with head and neck cancer. PLoS One 2021; 16:e0251455. [PMID: 33970954 PMCID: PMC8109770 DOI: 10.1371/journal.pone.0251455] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/27/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND A typical assessment for sarcopenia involves the use of abdominal computed tomography (CT) for calculating the skeletal muscle index (SMI) at the level of the third lumbar vertebra (L3). However, abdominal CT is not regularly performed on patients with head and neck cancer (HNC). We investigated whether masticatory SMI (M-SMI) measurements based on head and neck CT scans can be used to conduct sarcopenia assessments by evaluating whether M-SMI is correlated with L3-SMI. METHODS Abdominal and head and neck CT images of patients with trauma (n = 50) and HNC (n = 52) were analyzed retrospectively. Both manual delineation and threshold selection methods were used to measure cross-sectional areas of masticatory muscles and those of muscles at the L3 level on CT images. Muscle cross-sectional areas were normalized to height squared to calculate SMI, and a multivariate linear regression model was established to evaluate the correlation between the M-SMI and L3-SMI. Receiver operating characteristic curve analysis was used to assess the ability of the M-SMI to identify sarcopenia, and Cox logistic regression was used to identify predictors of sarcopenia. RESULTS Patients with HNC had significantly lower M-SMI and L3-SMI than did patients with trauma (p = 0.011 and 0.03, respectively). M-SMI and L3-SMI were strongly correlated (r = 0.901, p < 0.001); in the multivariate model that included sex, the correlation was stronger (r = 0.913, p < 0.001). The associations of sarcopenia with a lower M-SMI (p < 0.001), male sex (p = 0.028), and advanced age (p = 0.011) were significant, and multivariate logistic analysis demonstrated that an M-SMI of <5.5 was an independent predictor of sarcopenia (hazard ratio = 5.37, p < 0.001). CONCLUSIONS M-SMI assessment in routine head and neck CT scans is feasible and can be an alternative for detecting sarcopenia in patients with HNC.
Collapse
Affiliation(s)
- Sheng-Wei Chang
- Department of Radiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yuan-Hsiung Tsai
- Department of Radiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Cheng-Ming Hsu
- Department of Otorhinolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ethan I. Huang
- Department of Otorhinolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Geng-He Chang
- Department of Otorhinolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ming-Shao Tsai
- Department of Otorhinolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yao-Te Tsai
- Department of Otorhinolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| |
Collapse
|
32
|
Hexarelin Modulation of MAPK and PI3K/Akt Pathways in Neuro-2A Cells Inhibits Hydrogen Peroxide-Induced Apoptotic Toxicity. Pharmaceuticals (Basel) 2021; 14:ph14050444. [PMID: 34066741 PMCID: PMC8150489 DOI: 10.3390/ph14050444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 11/17/2022] Open
Abstract
Hexarelin, a synthetic hexapeptide, exerts cyto-protective effects at the mitochondrial level in cardiac and skeletal muscles, both in vitro and in vivo, may also have important neuroprotective bioactivities. This study examined the inhibitory effects of hexarelin on hydrogen peroxide (H2O2)-induced apoptosis in Neuro-2A cells. Neuro-2A cells were treated for 24 h with various concentrations of H2O2 or with the combination of H2O2 and hexarelin following which cell viability and nitrite (NO2−) release were measured. Cell morphology was also documented throughout and changes arising were quantified using Image J skeleton and fractal analysis procedures. Apoptotic responses were evaluated by Real-Time PCR (caspase-3, caspase-7, Bax, and Bcl-2 mRNA levels) and Western Blot (cleaved caspase-3, cleaved caspase-7, MAPK, and Akt). Our results indicate that hexarelin effectively antagonized H2O2-induced damage to Neuro-2A cells thereby (i) improving cell viability, (ii) reducing NO2− release and (iii) restoring normal morphologies. Hexarelin treatment also reduced mRNA levels of caspase-3 and its activation, and modulated mRNA levels of the BCL-2 family. Moreover, hexarelin inhibited MAPKs phosphorylation and increased p-Akt protein expression. In conclusion, our results demonstrate neuroprotective and anti-apoptotic effects of hexarelin, suggesting that new analogues could be developed for their neuroprotective effects.
Collapse
|
33
|
Vanderveen BN, Fix DK, Counts BR, Carson JA. The Effect of Wheel Exercise on Functional Indices of Cachexia in Tumor-bearing Mice. Med Sci Sports Exerc 2021; 52:2320-2330. [PMID: 33064407 DOI: 10.1249/mss.0000000000002393] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Cancer-related fatigue and muscle wasting have received significant attention over the last few decades with the goal of establishing interventions that can improve cancer patient life quality and survival. Increased physical activity has shown to reduce cancer-associated fatigue and has been proposed as a promising therapeutic to attenuate cancer-induced wasting. However, significant gaps remain in our understanding of how physical activity affects the compositional and functional changes that initiate muscle wasting. The purpose of the current study was to determine the effect of wheel exercise on body composition and functional indices of cancer cachexia before the development of significant wasting. METHODS Thirteen-week-old male Apc (MIN) and C57BL/6 (B6) mice were given free wheel access (W) or a locked wheel (Sed) for 5 wk. RESULTS Wheel activity was reduced in the MIN compared with B6; however, wheel access increased complex II expression in isolated skeletal muscle mitochondria regardless of genotype. Wheel access had no effect on tumor burden or plasma interleukin-6 in the MIN. MIN-W increased body weight and lean mass compared with MIN-Sed, and there was a direct correlation between wheel distance and lean mass change. MIN-W increased grip strength and treadmill time to fatigue compared with MIN-Sed. Within MIN-W mice, skeletal muscle fatigability was only improved in high runners (>60 min·d). CONCLUSIONS Our results suggest that there were therapeutic benefits of increased activity related to body composition, behavior, and whole-body function that were not dependent on exercise duration; however, there was an exercise threshold needed to improve skeletal muscle fatigability in tumor-bearing mice. Interestingly, wheel access was able to improve compositional and functional outcomes without mitigating tumor number or size.
Collapse
Affiliation(s)
| | - Dennis K Fix
- Department of Exercise Science, University of South Carolina, Columbia, SC
| | - Brittany R Counts
- Department of Physical Therapy, College of Health Professions, The University of Tennessee Health Sciences Center, Memphis, TN
| | - James A Carson
- Department of Physical Therapy, College of Health Professions, The University of Tennessee Health Sciences Center, Memphis, TN
| |
Collapse
|
34
|
Kunz HE, Dorschner JM, Berent TE, Meyer T, Wang X, Jatoi A, Kumar R, Lanza IR. Methylarginine metabolites are associated with attenuated muscle protein synthesis in cancer-associated muscle wasting. J Biol Chem 2021; 295:17441-17459. [PMID: 33453990 DOI: 10.1074/jbc.ra120.014884] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/22/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer cachexia is characterized by reductions in peripheral lean muscle mass. Prior studies have primarily focused on increased protein breakdown as the driver of cancer-associated muscle wasting. Therapeutic interventions targeting catabolic pathways have, however, largely failed to preserve muscle mass in cachexia, suggesting that other mechanisms might be involved. In pursuit of novel pathways, we used untargeted metabolomics to search for metabolite signatures that may be linked with muscle atrophy. We injected 7-week-old C57/BL6 mice with LLC1 tumor cells or vehicle. After 21 days, tumor-bearing mice exhibited reduced body and muscle mass and impaired grip strength compared with controls, which was accompanied by lower synthesis rates of mixed muscle protein and the myofibrillar and sarcoplasmic muscle fractions. Reductions in protein synthesis were accompanied by mitochondrial enlargement and reduced coupling efficiency in tumor-bearing mice. To generate mechanistic insights into impaired protein synthesis, we performed untargeted metabolomic analyses of plasma and muscle and found increased concentrations of two methylarginines, asymmetric dimethylarginine (ADMA) and NG-monomethyl-l-arginine, in tumor-bearing mice compared with control mice. Compared with healthy controls, human cancer patients were also found to have higher levels of ADMA in the skeletal muscle. Treatment of C2C12 myotubes with ADMA impaired protein synthesis and reduced mitochondrial protein quality. These results suggest that increased levels of ADMA and mitochondrial changes may contribute to impaired muscle protein synthesis in cancer cachexia and could point to novel therapeutic targets by which to mitigate cancer cachexia.
Collapse
Affiliation(s)
- Hawley E Kunz
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jessica M Dorschner
- Nephrology and Hypertension Research Unit, Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Taylor E Berent
- Nephrology and Hypertension Research Unit, Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas Meyer
- Nephrology and Hypertension Research Unit, Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Xuewei Wang
- Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Aminah Jatoi
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Rajiv Kumar
- Nephrology and Hypertension Research Unit, Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA.
| | - Ian R Lanza
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
35
|
A transition to degeneration triggered by oxidative stress in degenerative disorders. Mol Psychiatry 2021; 26:736-746. [PMID: 33159186 PMCID: PMC7914161 DOI: 10.1038/s41380-020-00943-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/15/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022]
Abstract
Although the activities of many signaling pathways are dysregulated during the progression of neurodegenerative and muscle degeneration disorders, the precise sequence of cellular events leading to degeneration has not been fully elucidated. Two kinases of particular interest, the growth-promoting Tor kinase and the energy sensor AMPK, appear to show reciprocal changes in activity during degeneration, with increased Tor activity and decreased AMPK activity reported. These changes in activity have been predicted to cause degeneration by attenuating autophagy, leading to the accumulation of unfolded protein aggregates and dysfunctional mitochondria, the consequent increased production of reactive oxygen species (ROS), and ultimately oxidative damage. Here we propose that this increased ROS production not only causes oxidative damage but also ultimately induces an oxidative stress response that reactivates the redox-sensitive AMPK and activates the redox-sensitive stress kinase JNK. Activation of these kinases reactivates autophagy. Because at this late stage, cells have become filled with dysfunctional mitochondria and protein aggregates, which are autophagy targets, this autophagy reactivation induces degeneration. The mechanism proposed here emphasizes that the process of degeneration is dynamic, that dysregulated signaling pathways change over time and can transition from deleterious to beneficial and vice versa as degeneration progresses.
Collapse
|
36
|
Qualls AE, Southern WM, Call JA. Mitochondria-cytokine crosstalk following skeletal muscle injury and disuse: a mini-review. Am J Physiol Cell Physiol 2021; 320:C681-C688. [PMID: 33566726 DOI: 10.1152/ajpcell.00462.2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Skeletal muscle mitochondria are highly adaptable, highly dynamic organelles that maintain the functional integrity of the muscle fiber by providing ATP for contraction and cellular homeostasis (e.g., Na+/K+ ATPase). Emerging as early modulators of inflammation, mitochondria sense and respond to cellular stress. Mitochondria communicate with the environment, in part, by release of physical signals called mitochondrial-derived damage-associated molecular patterns (mito-DAMPs) and deviation from routine function (e.g., reduced ATP production, Ca2+ overload). When skeletal muscle is compromised, mitochondria contribute to an acute inflammatory response necessary for myofibril regeneration; however, exhaustive signaling associated with altered or reduced mitochondrial function can be detrimental to muscle outcomes. Here, we describe changes in mitochondrial content, structure, and function following skeletal muscle injury and disuse and highlight the influence of mitochondria-cytokine crosstalk on muscle regeneration and recovery. Although the appropriate therapeutic modulation following muscle stressors remains unknown, retrospective gene expression analysis reveals that interleukin-6 (IL-6), interleukin-1β (IL-1β), chemokine C-X-C motif ligand 1 (CXCL1), and monocyte chemoattractant protein 1 (MCP-1) are significantly upregulated following three unique muscle injuries. These cytokines modulate mitochondrial function and execute bona fide pleiotropic roles that can aid functional recovery of muscle, however, when aberrant, chronically disrupt healing partly by exacerbating mitochondrial dysfunction. Multidisciplinary efforts to delineate the opposing regulatory roles of inflammatory cytokines in the muscle mitochondrial environment are required to modulate regenerative behavior following skeletal muscle injury or disuse. Future therapeutic directions to consider include quenching or limited release of mito-DAMPs and cytokines present in cytosol or circulation.
Collapse
Affiliation(s)
- Anita E Qualls
- Clinical School, University of Cambridge, Cambridge, United Kingdom
| | - W Michael Southern
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Jarrod A Call
- Department of Kinesiology, University of Georgia, Athens, Georgia.,Regenerative Bioscience Center, University of Georgia, Athens, Georgia
| |
Collapse
|
37
|
De Mario A, Gherardi G, Rizzuto R, Mammucari C. Skeletal muscle mitochondria in health and disease. Cell Calcium 2021; 94:102357. [PMID: 33550207 DOI: 10.1016/j.ceca.2021.102357] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 12/28/2022]
Abstract
Mitochondrial activity warrants energy supply to oxidative myofibres to sustain endurance workload. The maintenance of mitochondrial homeostasis is ensured by the control of fission and fusion processes and by the mitophagic removal of aberrant organelles. Many diseases are due to or characterized by dysfunctional mitochondria, and altered mitochondrial dynamics or turnover trigger myopathy per se. In this review, we will tackle the role of mitochondrial dynamics, turnover and metabolism in skeletal muscle, both in health and disease.
Collapse
Affiliation(s)
- Agnese De Mario
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Gaia Gherardi
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | | |
Collapse
|
38
|
Morigny P, Zuber J, Haid M, Kaltenecker D, Riols F, Lima JDC, Simoes E, Otoch JP, Schmidt SF, Herzig S, Adamski J, Seelaender M, Berriel Diaz M, Rohm M. High levels of modified ceramides are a defining feature of murine and human cancer cachexia. J Cachexia Sarcopenia Muscle 2020; 11:1459-1475. [PMID: 33090732 PMCID: PMC7749558 DOI: 10.1002/jcsm.12626] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cancer cachexia (CCx) is a multifactorial energy-wasting syndrome reducing the efficiency of anti-cancer therapies, quality of life, and survival of cancer patients. In the past years, most studies focused on the identification of tumour and host-derived proteins contributing to CCx. However, there is still a lack of studies addressing the changes in bioactive lipids. The aim of this study was to identify specific lipid species as a hallmark of CCx by performing a broad range lipid analysis of plasma from well-established CCx mouse models as well as cachectic and weight stable cancer patients. METHODS Plasma from non-cachectic (PBS-injected mice, NC26 tumour-bearing mice), pre-cachectic and cachectic mice (C26 and LLC tumour-bearing mice, ApcMin/+ mutant mice), and plasma from weight stable and cachectic patients with gastrointestinal cancer, were analysed using the Lipidyzer™ platform. In total, 13 lipid classes and more than 1100 lipid species, including sphingolipids, neutral and polar glycerolipids, were covered by the analysis. Correlation analysis between specific lipid species and readouts of CCx were performed. Lipidomics data were confirmed by gene expression analysis of metabolic organs to analyse enzymes involved in sphingolipid synthesis and degradation. RESULTS A decrease in several lysophosphatidylcholine (LPC) species and an increase in numerous sphingolipids including sphingomyelins (SMs), ceramides (CERs), hexosyl-ceramides (HCERs) and lactosyl-ceramides (LCERs), were mutual features of CCx in both mice and cancer patients. Notably, sphingolipid levels gradually increased during cachexia development. Key enzymes involved in ceramide synthesis were elevated in liver but not in adipose, muscle, or tumour tissues, suggesting that ceramide turnover in the liver is a major contributor to elevated sphingolipid levels in CCx. LPC(16:1), LPC(20:3), SM(16:0), SM(24:1), CER(16:0), CER(24:1), HCER(16:0), and HCER(24:1) were the most consistently affected lipid species between mice and humans and correlated negatively (LPCs) or positively (SMs, CERs and HCERs) with the severity of body weight loss. CONCLUSIONS High levels of sphingolipids, specifically ceramides and modified ceramides, are a defining feature of murine and human CCx and may contribute to tissue wasting and skeletal muscle atrophy through the inhibition of anabolic signals. The progressive increase in sphingolipids during cachexia development supports their potential as early biomarkers for CCx.
Collapse
Affiliation(s)
- Pauline Morigny
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Julia Zuber
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Mark Haid
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Doris Kaltenecker
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Fabien Riols
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Joanna D C Lima
- Cancer Metabolism Research Group, LIM 26 HC, Medical School, University of São Paulo, São Paulo, Brazil
| | - Estefania Simoes
- Cancer Metabolism Research Group, LIM 26 HC, Medical School, University of São Paulo, São Paulo, Brazil
| | - José Pinhata Otoch
- Cancer Metabolism Research Group, LIM 26 HC, Medical School, University of São Paulo, São Paulo, Brazil
| | - Sören Fisker Schmidt
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Molecular Metabolic Control, Technical University of Munich, Munich, Germany
| | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Experimentelle Genetik, Technical University of Munich, Freising-Weihenstephan, Neuherberg, Germany.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Marilia Seelaender
- Cancer Metabolism Research Group, LIM 26 HC, Medical School, University of São Paulo, São Paulo, Brazil
| | - Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Cachexia, a feature of cancer and other chronic diseases, is marked by progressive weight loss and skeletal muscle wasting. This review aims to highlight the sex differences in manifestations of cancer cachexia in patients, rodent models, and our current understanding of the potential mechanisms accounting for these differences. RECENT FINDINGS Male cancer patients generally have higher prevalence of cachexia, greater weight loss or muscle wasting, and worse outcomes compared with female cancer patients. Knowledge is increasing about sex differences in muscle fiber type and function, mitochondrial metabolism, global gene expression and signaling pathways, and regulatory mechanisms at the levels of sex chromosomes vs. sex hormones; however, it is largely undetermined how such sex differences directly affect the susceptibility to stressors leading to muscle wasting in cancer cachexia. Few studies have investigated basic mechanisms underlying sex differences in cancer cachexia. A better understanding of sex differences would improve cachexia treatment in both sexes.
Collapse
Affiliation(s)
- Xiaoling Zhong
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA
- Research Service, Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Teresa A Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA.
- Research Service, Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
- IU Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
- Department of Otolaryngology-Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
40
|
Brown JL, Lawrence MM, Ahn B, Kneis P, Piekarz KM, Qaisar R, Ranjit R, Bian J, Pharaoh G, Brown C, Peelor FF, Kinter MT, Miller BF, Richardson A, Van Remmen H. Cancer cachexia in a mouse model of oxidative stress. J Cachexia Sarcopenia Muscle 2020; 11:1688-1704. [PMID: 32918528 PMCID: PMC7749559 DOI: 10.1002/jcsm.12615] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/03/2020] [Accepted: 07/07/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Cancer is associated with muscle atrophy (cancer cachexia) that is linked to up to 40% of cancer-related deaths. Oxidative stress is a critical player in the induction and progression of age-related loss of muscle mass and weakness (sarcopenia); however, the role of oxidative stress in cancer cachexia has not been defined. The purpose of this study was to examine if elevated oxidative stress exacerbates cancer cachexia. METHODS Cu/Zn superoxide dismutase knockout (Sod1KO) mice were used as an established mouse model of elevated oxidative stress. Cancer cachexia was induced by injection of one million Lewis lung carcinoma (LLC) cells or phosphate-buffered saline (saline) into the hind flank of female wild-type mice or Sod1KO mice at approximately 4 months of age. The tumour developed for 3 weeks. Muscle mass, contractile function, neuromuscular junction (NMJ) fragmentation, metabolic proteins, mitochondrial function, and motor neuron function were measured in wild-type and Sod1KO saline and tumour-bearing mice. Data were analysed by two-way ANOVA with Tukey-Kramer post hoc test when significant F ratios were determined and α was set at 0.05. Unless otherwise noted, results in abstract are mean ±SEM. RESULTS Muscle mass and cross-sectional area were significantly reduced, in tumour-bearing mice. Metabolic enzymes were dysregulated in Sod1KO mice and cancer exacerbated this phenotype. NMJ fragmentation was exacerbated in tumour-bearing Sod1KO mice. Myofibrillar protein degradation increased in tumour-bearing wild-type mice (wild-type saline, 0.00847 ± 0.00205; wildtype LLC, 0.0211 ± 0.00184) and tumour-bearing Sod1KO mice (Sod1KO saline, 0.0180 ± 0.00118; Sod1KO LLC, 0.0490 ± 0.00132). Muscle mitochondrial oxygen consumption was reduced in tumour-bearing mice compared with saline-injected wild-type mice. Mitochondrial protein degradation increased in tumour-bearing wild-type mice (wild-type saline, 0.0204 ± 0.00159; wild-type LLC, 0.167 ± 0.00157) and tumour-bearing Sod1KO mice (Sod1KO saline, 0.0231 ± 0.00108; Sod1 KO LLC, 0.0645 ± 0.000631). Sciatic nerve conduction velocity was decreased in tumour-bearing wild-type mice (wild-type saline, 38.2 ± 0.861; wild-type LLC, 28.8 ± 0.772). Three out of eleven of the tumour-bearing Sod1KO mice did not survive the 3-week period following tumour implantation. CONCLUSIONS Oxidative stress does not exacerbate cancer-induced muscle loss; however, cancer cachexia may accelerate NMJ disruption.
Collapse
Affiliation(s)
- Jacob L Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Marcus M Lawrence
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Bumsoo Ahn
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Parker Kneis
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Katarzyna M Piekarz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rizwan Qaisar
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Rojina Ranjit
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jan Bian
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Gavin Pharaoh
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Chase Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Fredrick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael T Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Arlan Richardson
- Oklahoma City VA Medical Center, Oklahoma City, OK, USA.,Reynolds Center for Aging Research, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.,Oklahoma City VA Medical Center, Oklahoma City, OK, USA.,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
41
|
Exercise as a therapy for cancer-induced muscle wasting. SPORTS MEDICINE AND HEALTH SCIENCE 2020; 2:186-194. [PMID: 35782998 PMCID: PMC9219331 DOI: 10.1016/j.smhs.2020.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/12/2020] [Accepted: 11/17/2020] [Indexed: 12/17/2022] Open
Abstract
Cancer cachexia is a progressive disorder characterized by body weight, fat, and muscle loss. Cachexia induces metabolic disruptions that can be analogous and distinct from those observed in cancer, obscuring both diagnosis and treatment options. Inflammation, hypogonadism, and physical inactivity are widely investigated as systemic mediators of cancer-induced muscle wasting. At the cellular level, dysregulation of protein turnover and energy metabolism can negatively impact muscle mass and function. Exercise is well known for its anti-inflammatory effects and potent stimulation of anabolic signaling. Emerging evidence suggests the potential for exercise to rescue muscle's sensitivity to anabolic stimuli, reduce wasting through protein synthesis modulation, myokine release, and subsequent downregulation of proteolytic factors. To date, there is no recommendation for exercise in the management of cachexia. Given its complex nature, a multimodal approach incorporating exercise offers promising potential for cancer cachexia treatment. This review's primary objective is to summarize the growing body of research examining exercise regulation of cancer cachexia. Furthermore, we will provide evidence for exercise interactions with established systemic and cellular regulators of cancer-induced muscle wasting.
Collapse
|
42
|
Yang Q, Han B, Xue J, Lv Y, Li S, Liu Y, Wu P, Wang X, Zhang Z. Hexavalent chromium induces mitochondrial dynamics disorder in rat liver by inhibiting AMPK/PGC-1α signaling pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 265:114855. [PMID: 32474337 DOI: 10.1016/j.envpol.2020.114855] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 06/11/2023]
Abstract
Occupational exposure to hexavalent chromium (Cr(VI)) can cause cytotoxicity and carcinogenicity. In this study, we established a liver injury model in rats via intraperitoneal injection of potassium dichromate (0, 2, 4, and 6 mg/kg body weight) for 35 d to investigate the mechanism of Cr(VI)-induced liver injury. We found that Cr(VI) induced hepatic histopathological lesions, oxidative stress, and apoptosis and reduced the expression of mitochondrial-related regulatory factors such as adenosine 5'-monophosphate-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) in a dose-dependent manner. Furthermore, Cr(VI) promoted mitochondrial division and inhibited fusion, leading to increased expression of caspase-3 and production of mitochondrial reactive oxygen species. Our study demonstrates that long-term exposure to Cr(VI) induces mitochondrial dynamics disorder by inhibiting AMPK/PGC-1α signaling pathway in rat liver.
Collapse
Affiliation(s)
- Qingyue Yang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin, 150030, China
| | - Bing Han
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Jiangdong Xue
- College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, 028000, China
| | - Yueying Lv
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Yan Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Pengfei Wu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Xiaoqiao Wang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin, 150030, China.
| |
Collapse
|
43
|
VanderVeen BN, Murphy EA, Carson JA. The Impact of Immune Cells on the Skeletal Muscle Microenvironment During Cancer Cachexia. Front Physiol 2020; 11:1037. [PMID: 32982782 PMCID: PMC7489038 DOI: 10.3389/fphys.2020.01037] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/28/2020] [Indexed: 12/22/2022] Open
Abstract
Progressive weight loss combined with skeletal muscle atrophy, termed cachexia, is a common comorbidity associated with cancer that results in adverse consequences for the patient related to decreased chemotherapy responsiveness and increased mortality. Cachexia's complexity has provided a barrier for developing successful therapies to prevent or treat the condition, since a large number of systemic disruptions that can regulate muscle mass are often present. Furthermore, considerable effort has focused on investigating how tumor derived factors and inflammatory mediators directly signal skeletal muscle to disrupt protein turnover regulation. Currently, there is developing appreciation for understanding how cancer alters skeletal muscle's complex microenvironment and the tightly regulated interactions between multiple cell types. Skeletal muscle microenvironment interactions have established functions in muscle response to regeneration from injury, growth, aging, overload-induced hypertrophy, and exercise. This review explores the growing body of evidence for immune cell modulation of the skeletal muscle microenvironment during cancer-induced muscle wasting. Emphasis is placed on the regulatory network that integrates physiological responses between immune cells with other muscle cell types including satellite cells, fibroblast cells, and endothelial cells to regulate myofiber size and plasticity. The overall goal of this review is to provide an understanding of how different cell types that constitute the muscle microenvironment and their signaling mediators contribute to cancer and chemotherapy-induced muscle wasting.
Collapse
Affiliation(s)
- Brandon N. VanderVeen
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
- AcePre, LLC, Columbia, SC, United States
| | - E. Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
- AcePre, LLC, Columbia, SC, United States
| | - James A. Carson
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
44
|
Shen Y, Zhang Q, Huang Z, Zhu J, Qiu J, Ma W, Yang X, Ding F, Sun H. Isoquercitrin Delays Denervated Soleus Muscle Atrophy by Inhibiting Oxidative Stress and Inflammation. Front Physiol 2020; 11:988. [PMID: 32903465 PMCID: PMC7435639 DOI: 10.3389/fphys.2020.00988] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022] Open
Abstract
Although denervated muscle atrophy is common, the underlying molecular mechanism remains unelucidated. We have previously found that oxidative stress and inflammatory response may be early events that trigger denervated muscle atrophy. Isoquercitrin is a biologically active flavonoid with antioxidative and anti-inflammatory properties. The present study investigated the effect of isoquercitrin on denervated soleus muscle atrophy and its possible molecular mechanisms. We found that isoquercitrin was effective in alleviating soleus muscle mass loss following denervation in a dose-dependent manner. Isoquercitrin demonstrated the optimal protective effect at 20 mg/kg/d, which was the dose used in subsequent experiments. To further explore the protective effect of isoquercitrin on denervated soleus muscle atrophy, we analyzed muscle proteolysis via the ubiquitin-proteasome pathway, mitophagy, and muscle fiber type conversion. Isoquercitrin significantly inhibited the denervation-induced overexpression of two muscle-specific ubiquitin ligases—muscle RING finger 1 (MuRF1) and muscle atrophy F-box (MAFbx), and reduced the degradation of myosin heavy chains (MyHCs) in the target muscle. Following isoquercitrin treatment, mitochondrial vacuolation and autophagy were inhibited, as evidenced by reduced level of autophagy-related proteins (ATG7, BNIP3, LC3B, and PINK1); slow-to-fast fiber type conversion in the target muscle was delayed via triggering expression of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α); and the production of reactive oxygen species (ROS) in the target muscle was reduced, which might be associated with the upregulation of antioxidant factors (SOD1, SOD2, NRF2, NQO1, and HO1) and the downregulation of ROS production-related factors (Nox2, Nox4, and DUOX1). Furthermore, isoquercitrin treatment reduced the levels of inflammatory factors—interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α)—in the target muscle and inactivated the JAK/STAT3 signaling pathway. Overall, isoquercitrin may alleviate soleus muscle atrophy and mitophagy and reverse the slow-to-fast fiber type conversion following denervation via inhibition of oxidative stress and inflammatory response. Our study findings enrich the knowledge regarding the molecular regulatory mechanisms of denervated muscle atrophy and provide a scientific basis for isoquercitrin as a protective drug for the prevention and treatment of denervated muscle atrophy.
Collapse
Affiliation(s)
- Yuntian Shen
- 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
| | - Qiuyu Zhang
- 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
| | - Ziwei Huang
- 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
| | - Jianwei Zhu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiayi Qiu
- School of Nursing, 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
| | - Fei Ding
- 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
|
45
|
Dave DT, Patel BM. Mitochondrial Metabolism in Cancer Cachexia: Novel Drug Target. Curr Drug Metab 2020; 20:1141-1153. [PMID: 31418657 DOI: 10.2174/1389200220666190816162658] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/23/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Cancer cachexia is a metabolic syndrome prevalent in the majority of the advanced cancers and is associated with complications such as anorexia, early satiety, weakness, anaemia, and edema, thereby reducing performance and impairing quality of life. Skeletal muscle wasting is a characteristic feature of cancer-cachexia and mitochondria is responsible for regulating total protein turnover in skeletal muscle tissue. METHODS We carried out exhaustive search for cancer cachexia and role of mitochondria in the same in various databases. All the relevant articles were gathered and the pertinent information was extracted out and compiled which was further structured into different sub-sections. RESULTS Various findings on the mitochondrial alterations in connection to its disturbed normal physiology in various models of cancer-cachexia have been recently reported, suggesting a significant role of the organelle in the pathogenesis of the complications involved in the disorder. It has also been reported that reduced mitochondrial oxidative capacity is due to reduced mitochondrial biogenesis as well as altered balance between fusion and fission protein activities. Moreover, autophagy in mitochondria (termed as mitophagy) is reported to play an important role in cancer cachexia. CONCLUSION The present review aims to put forth the changes occurring in mitochondria and hence explore possible targets which can be exploited in cancer-induced cachexia for treatment of such a debilitating condition.
Collapse
Affiliation(s)
- Dhwani T Dave
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad 382481, Gujarat, India
| | - Bhoomika M Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad 382481, Gujarat, India
| |
Collapse
|
46
|
Leucine-Rich Diet Modulates the Metabolomic and Proteomic Profile of Skeletal Muscle during Cancer Cachexia. Cancers (Basel) 2020; 12:cancers12071880. [PMID: 32668598 PMCID: PMC7408981 DOI: 10.3390/cancers12071880] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Cancer-cachexia induces a variety of metabolic disorders, including skeletal muscle imbalance. Alternative therapy, as nutritional supplementation with leucine, shows a modulatory effect over tumour damage in vivo and in vitro. Method: Adult rats distributed into Control (C), Walker tumour-bearing (W), control fed a leucine-rich diet (L), and tumour-bearing fed a leucine-rich diet (WL) groups had the gastrocnemius muscle metabolomic and proteomic assays performed in parallel to in vitro assays. Results: W group presented an affected muscle metabolomic and proteomic profile mainly related to energy generation and carbohydrates catabolic processes, but leucine-supplemented group (WL) recovered the energy production. In vitro assay showed that cell proliferation, mitochondria number and oxygen consumption were higher under leucine effect than the tumour influence. Muscle proteomics results showed that the main affected cell component was mitochondria, leading to an impacted energy generation, including impairment in proteins of the tricarboxylic cycle and carbohydrates catabolic processes, which were modulated and improved by leucine treatment. Conclusion: In summary, we showed a beneficial effect of leucine upon mitochondria, providing information about the muscle glycolytic pathways used by this amino acid, where it can be associated with the preservation of morphometric parameters and consequent protection against the effects of cachexia.
Collapse
|
47
|
Arneson-Wissink PC, Hogan KA, Ducharme AM, Samani A, Jatoi A, Doles JD. The wasting-associated metabolite succinate disrupts myogenesis and impairs skeletal muscle regeneration. JCSM RAPID COMMUNICATIONS 2020; 3:56-69. [PMID: 32905522 PMCID: PMC7470228 DOI: 10.1002/rco2.14] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
BACKGROUND Muscle wasting is a debilitating co-morbidity affecting most advanced cancer patients. Alongside enhanced muscle catabolism, defects in muscle repair/regeneration contribute to cancer-associated wasting. Among the factors implicated in suppression of muscle regeneration are cytokines that interfere with myogenic signal transduction pathways. Less understood is how other cancer/wasting-associated cues, such as metabolites, contribute to muscle dysfunction. This study investigates how the metabolite succinate affects myogenesis and muscle regeneration. METHODS We leveraged an established ectopic metabolite treatment (cell permeable dimethyl-succinate) strategy to evaluate the ability of intracellular succinate elevation to 1) affect myoblast homeostasis (proliferation, apoptosis), 2) disrupt protein dynamics and induce wasting-associated atrophy, and 3) modulate in vitro myogenesis. In vivo succinate supplementation experiments (2% succinate, 1% sucrose vehicle) were used to corroborate and extend in vitro observations. Metabolic profiling and functional metabolic studies were then performed to investigate the impact of succinate elevation on mitochondria function. RESULTS We found that in vitro succinate supplementation elevated intracellular succinate about 2-fold, and did not have an impact on proliferation or apoptosis of C2C12 myoblasts. Elevated succinate had minor effects on protein homeostasis (~25% decrease in protein synthesis assessed by OPP staining), and no significant effect on myotube atrophy. Succinate elevation interfered with in vitro myoblast differentiation, characterized by significant decreases in late markers of myogenesis and fewer nuclei per myosin heavy chain positive structure (assessed by immunofluorescence staining). While mice orally administered succinate did not exhibit changes in overall body composition or whole muscle weights, these mice displayed smaller muscle myofiber diameters (~6% decrease in the mean of non-linear regression curves fit to the histograms of minimum feret diameter distribution), which was exacerbated when muscle regeneration was induced with barium chloride injury. Significant decreases in the mean of non-linear regression curves fit to the histograms of minimum feret diameter distributions were observed 7 days and 28 days post injury. Elevated numbers of myogenin positive cells (3-fold increase) supportive of the differentiation defects observed in vitro were observed 28 days post injury. Metabolic profiling and functional metabolic assessment of myoblasts revealed that succinate elevation caused both widespread metabolic changes and significantly lowered maximal cellular respiration (~35% decrease). CONCLUSIONS This study broadens the repertoire of wasting-associated factors that can directly modulate muscle progenitor cell function and strengthens the hypothesis that metabolic derangements are significant contributors to impaired muscle regeneration, an important aspect of cancer-associated muscle wasting.
Collapse
Affiliation(s)
- Paige C Arneson-Wissink
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Kelly A Hogan
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Alexandra M Ducharme
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Adrienne Samani
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Aminah Jatoi
- Department of Oncology, Mayo Clinic, Rochester,
Minnesota
| | - Jason D Doles
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
- Corresponding Author: Jason D Doles, Department of
Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Guggenheim
16-11A1, Rochester, MN 55905, Tel: (507) 284-9372, Fax: (507) 284-3383,
| |
Collapse
|
48
|
Hulmi JJ, Penna F, Pöllänen N, Nissinen TA, Hentilä J, Euro L, Lautaoja JH, Ballarò R, Soliymani R, Baumann M, Ritvos O, Pirinen E, Lalowski M. Muscle NAD + depletion and Serpina3n as molecular determinants of murine cancer cachexia-the effects of blocking myostatin and activins. Mol Metab 2020; 41:101046. [PMID: 32599075 PMCID: PMC7364159 DOI: 10.1016/j.molmet.2020.101046] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/16/2020] [Accepted: 06/23/2020] [Indexed: 12/26/2022] Open
Abstract
Objective Cancer cachexia and muscle loss are associated with increased morbidity and mortality. In preclinical animal models, blocking activin receptor (ACVR) ligands has improved survival and prevented muscle wasting in cancer cachexia without an effect on tumour growth. However, the underlying mechanisms are poorly understood. This study aimed to identify cancer cachexia and soluble ACVR (sACVR) administration-evoked changes in muscle proteome. Methods Healthy and C26 tumour-bearing (TB) mice were treated with recombinant sACVR. The sACVR or PBS control were administered either prior to the tumour formation or by continued administration before and after tumour formation. Muscles were analysed by quantitative proteomics with further examination of mitochondria and nicotinamide adenine dinucleotide (NAD+) metabolism. To complement the first prophylactic experiment, sACVR (or PBS) was injected as a treatment after tumour cell inoculation. Results Muscle proteomics in TB cachectic mice revealed downregulated signatures for mitochondrial oxidative phosphorylation (OXPHOS) and increased acute phase response (APR). These were accompanied by muscle NAD+ deficiency, alterations in NAD+ biosynthesis including downregulation of nicotinamide riboside kinase 2 (Nrk2), and decreased muscle protein synthesis. The disturbances in NAD+ metabolism and protein synthesis were rescued by treatment with sACVR. Across the whole proteome and APR, in particular, Serpina3n represented the most upregulated protein and the strongest predictor of cachexia. However, the increase in Serpina3n expression was associated with increased inflammation rather than decreased muscle mass and/or protein synthesis. Conclusions We present evidence implicating disturbed muscle mitochondrial OXPHOS proteome and NAD+ homeostasis in experimental cancer cachexia. Treatment of TB mice with a blocker of activin receptor ligands restores depleted muscle NAD+ and Nrk2, as well as decreased muscle protein synthesis. These results indicate putative new treatment therapies for cachexia and that although acute phase protein Serpina3n may serve as a predictor of cachexia, it more likely reflects a condition of elevated inflammation. Cachectic muscle proteome shows decreased OXPHOS and increased acute phase response. Cancer cachexia is characterized by lowered muscle Nrk2 expression and NAD+ levels. Blocking activin receptor 2B ligands rescues muscle NAD+ homeostasis in cachexia. Blocking activin receptor 2B ligands prevents affected protein synthesis in cachexia. Serpina3n predicts cachexia and cancer-induced APR independently from muscle atrophy.
Collapse
Affiliation(s)
- J J Hulmi
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland; Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - F Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - N Pöllänen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - T A Nissinen
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - J Hentilä
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - L Euro
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J H Lautaoja
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - R Ballarò
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - R Soliymani
- Meilahti Clinical Proteomics Core Facility, HiLIFE, Faculty of Medicine, Biochemistry and Developmental biology, University of Helsinki, Helsinki, Finland
| | - M Baumann
- Meilahti Clinical Proteomics Core Facility, HiLIFE, Faculty of Medicine, Biochemistry and Developmental biology, University of Helsinki, Helsinki, Finland
| | - O Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - E Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - M Lalowski
- Meilahti Clinical Proteomics Core Facility, HiLIFE, Faculty of Medicine, Biochemistry and Developmental biology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
49
|
Hardee JP, Fix DK, Koh HJ, Wang X, Goldsmith EC, Carson JA. Repeated eccentric contractions positively regulate muscle oxidative metabolism and protein synthesis during cancer cachexia in mice. J Appl Physiol (1985) 2020; 128:1666-1676. [PMID: 32407241 DOI: 10.1152/japplphysiol.00908.2019] [Citation(s) in RCA: 15] [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
Cancer-induced wasting is accompanied by disruptions to muscle oxidative metabolism and protein turnover that have been associated with systemic inflammation, whereas exercise and stimulated muscle contractions can positively regulate muscle protein synthesis and mitochondrial homeostasis. In preclinical cancer cachexia models, a single bout of eccentric contractions (ECCs) can induce protein synthesis and repeated ECC bouts prevent myofiber atrophy. The cellular mechanisms providing this protection from atrophy have not been resolved. Therefore, the purpose of this study was to determine whether repeated stimulated ECC bouts affect basal muscle oxidative metabolism and protein synthesis during cancer cachexia, and if these changes were associated with plasma IL-6 levels. Male ApcMin/+ (MIN; n = 10) mice initiating cachexia and healthy C57BL/6 (B6; n = 11) control mice performed repeated ECC bouts over 2 wk. MIN mice exhibited body weight loss and elevated plasma IL-6 before and during repeated ECC bouts. Control MIN muscle demonstrated disrupted signaling related to inflammation, oxidative capacity, and protein synthesis regulation, which were all improved by repeated ECC bouts. With cachexia, plasma IL-6 levels were negatively correlated with myofiber cross-sectional area, oxidative capacity, and protein synthesis. Interestingly, ECC improvements in these outcomes were positively correlated with plasma IL-6 levels in MIN mice. There was also a positive relationship between muscle oxidative capacity and protein synthesis after repeated ECC bouts in MIN mice. Collectively, repeated ECC bouts altered the cachectic muscle phenotype independent of systemic wasting, and there was a strong association between muscle oxidative capacity and protein synthesis in this adaptive response.NEW & NOTEWORTHY Cancer-induced muscle wasting is accompanied by disruptions to muscle oxidative metabolism and protein turnover regulation, whereas exercise is a potent stimulator of muscle protein synthesis and mitochondrial homeostasis. In a preclinical model of cancer cachexia, we report that cachectic muscle retains anabolic and metabolic plasticity to repeated eccentric contraction bouts despite an overall systemic wasting environment. The attenuation of muscle atrophy is linked to improved oxidative capacity and protein synthesis during cancer cachexia progression.
Collapse
Affiliation(s)
- Justin P Hardee
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Dennis K Fix
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Ho-Jin Koh
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Xuewen Wang
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Edie C Goldsmith
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - James A Carson
- Center for Muscle Metabolism and Neuropathology, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
50
|
de Lima FD, Battaglini CL, Chaves SN, Ugliara L, Sarandy J, Lima RM, Bottaro M. Effect of strength training and antioxidant supplementation on perceived and performance fatigability in breast cancer survivors: a randomized, double-blinded, placebo-controlled study. Appl Physiol Nutr Metab 2020; 45:1165-1173. [PMID: 32348688 DOI: 10.1139/apnm-2020-0166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This randomized, double-blinded, placebo-controlled study aimed to investigate the effect of strength training (ST) combined with vitamin C and E supplementation on perceived and performance fatigability in breast cancer survivors (BCS). Twenty-five BCS were randomly assigned to 1 of 2 groups: vitamins (VIT; n = 12; 51.0 ± 9.0 years) or placebo (PLA; n = 13; 48.2 ± 8.3 years). Both groups performed a 10-week ST protocol, twice a week. The VIT group was supplemented with vitamins C (500 mg/day) and E (180 mg/day) and the PLA group with polydextrose (1 g/day), once a day after breakfast. At the beginning and at the end of the training period, perceived fatigability was assessed using Multidimensional Fatigue Inventory (MFI)-20 (general fatigue and physical fatigue). Performance fatigability was assessed during 30 maximal isokinetic knee extensions at 120°/s. General fatigue decreased similarly in the VIT (p = 0.004) and PLA (p = 0.011) groups. Physical fatigue decreased similarly in the VIT (p = 0.011) and PLA (p = 0.001) groups. Performance fatigability also decreased similarly in the VIT (p = 0.026) and PLA (p < 0.001) groups. There was no difference between groups at any moment (p > 0.05). In summary, antioxidant supplementation does not add any positive synergistic effect to ST in terms of improving perceived or performance fatigability in BCS. This clinical trial is registered in the Brazilian Clinical Trials Registry, number RBR-843pth (UTN no.: U1111-1222-6511). Novelty ST with maximal repetitions reduces perceived and performance fatigability of BCS. Vitamins C and E supplementation does not add any positive synergistic effect to ST in terms of reducing fatigability in BCS.
Collapse
Affiliation(s)
- Filipe Dinato de Lima
- College of Health Sciences, University of Brasília, Brasília, DF 70910-900, Brazil.,College of Health and Education Sciences, University Center of Brasília, Brasília, DF 70790-075, Brazil
| | - Cláudio L Battaglini
- Department of Exercise and Sport Science and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-8700, USA
| | - Sandro Nobre Chaves
- College of Physical Education, University of Brasília, Brasília, DF 70910-900, Brazil
| | - Lucas Ugliara
- College of Physical Education, University of Brasília, Brasília, DF 70910-900, Brazil
| | - Jonathan Sarandy
- College of Physical Education, University of Brasília, Brasília, DF 70910-900, Brazil
| | - Ricardo Moreno Lima
- College of Physical Education, University of Brasília, Brasília, DF 70910-900, Brazil
| | - Martim Bottaro
- College of Physical Education, University of Brasília, Brasília, DF 70910-900, Brazil
| |
Collapse
|