1
|
Sasamori R, Sato Y, Nomura K, Wakita A, Nagaki Y, Kemuriyama K, Sasaki Y, Nozaki S, Takahashi T, Terata K, Imai K, Minamiya Y. Lipopolysaccharide induces CCL2 through TLR4 signaling and promotes esophageal squamous cell carcinoma cell proliferation. Am J Cancer Res 2024; 14:3497-3512. [PMID: 39113860 PMCID: PMC11301279 DOI: 10.62347/eike6128] [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: 05/09/2024] [Accepted: 06/12/2024] [Indexed: 08/10/2024] Open
Abstract
Poor oral health is an independent risk factor for upper-aerodigestive tract cancers, including esophageal squamous cell carcinoma (ESCC). Our previous findings suggest that high expression of toll-like receptor (TLR) 4, which recognizes lipopolysaccharide (LPS) released from periodontal pathogens, correlates with a poor prognosis after esophagectomy for ESCC. We therefore hypothesized that LPS influences cancer cell proliferation and disease progression in ESCC. We used 8 ESCC cell lines to investigate how LPS affects ESCC cell proliferation and migration activity. We also assessed mRNA and protein expression to determine how LPS affects cytokine production and whether blocking TLR4 signaling attenuates that effect. We also used a mouse xenograft model to investigate whether LPS upregulates ESCC tumor progression in vivo. We then determined whether C-C motif chemokine ligand 2 (CCL2) expression in clinical samples correlates with 5-year overall survival (OS) and disease-specific survival (DSS) in ESCC patients after esophagectomy. LPS significantly upregulated cell proliferation and migration in all ESCC lines. It also upregulated CCL2 production. In vivo, subcutaneous LPS administration significantly increased ESCC tumor volume in mice. In clinical samples, high CCL2 expression significantly correlated with 5-year OS and DSS. There was also a significant correlation between CCL2 and TLR4 expression status, suggesting the involvement of an LPS-TLR4-CCL2 cascade in clinical settings. LPS significantly upregulates cell proliferation and tumor progression through an LPS-TLR4-CCL2 cascade and influences prognosis after esophagectomy for ESCC. This suggests improving the oral environment has the potential to improve the prognosis of ESCC patients after esophagectomy.
Collapse
Affiliation(s)
- Ryohei Sasamori
- Department of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Yusuke Sato
- Department of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Kyoko Nomura
- Department of Environmental Health Science and Public Health, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Akiyuki Wakita
- Department of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Yushi Nagaki
- Department of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Kohei Kemuriyama
- Department of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Yoshihiro Sasaki
- Department of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Shu Nozaki
- Department of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Tsukasa Takahashi
- Department of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Kaori Terata
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Kazuhiro Imai
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Yoshihiro Minamiya
- Department of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| |
Collapse
|
2
|
Martin A, Gallot YS, Freyssenet D. Molecular mechanisms of cancer cachexia-related loss of skeletal muscle mass: data analysis from preclinical and clinical studies. J Cachexia Sarcopenia Muscle 2023; 14:1150-1167. [PMID: 36864755 PMCID: PMC10235899 DOI: 10.1002/jcsm.13073] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 06/15/2022] [Accepted: 08/14/2022] [Indexed: 03/04/2023] Open
Abstract
Cancer cachexia is a systemic hypoanabolic and catabolic syndrome that diminishes the quality of life of cancer patients, decreases the efficiency of therapeutic strategies and ultimately contributes to decrease their lifespan. The depletion of skeletal muscle compartment, which represents the primary site of protein loss during cancer cachexia, is of very poor prognostic in cancer patients. In this review, we provide an extensive and comparative analysis of the molecular mechanisms involved in the regulation of skeletal muscle mass in human cachectic cancer patients and in animal models of cancer cachexia. We summarize data from preclinical and clinical studies investigating how the protein turnover is regulated in cachectic skeletal muscle and question to what extent the transcriptional and translational capacities, as well as the proteolytic capacity (ubiquitin-proteasome system, autophagy-lysosome system and calpains) of skeletal muscle are involved in the cachectic syndrome in human and animals. We also wonder how regulatory mechanisms such as insulin/IGF1-AKT-mTOR pathway, endoplasmic reticulum stress and unfolded protein response, oxidative stress, inflammation (cytokines and downstream IL1ß/TNFα-NF-κB and IL6-JAK-STAT3 pathways), TGF-ß signalling pathways (myostatin/activin A-SMAD2/3 and BMP-SMAD1/5/8 pathways), as well as glucocorticoid signalling, modulate skeletal muscle proteostasis in cachectic cancer patients and animals. Finally, a brief description of the effects of various therapeutic strategies in preclinical models is also provided. Differences in the molecular and biochemical responses of skeletal muscle to cancer cachexia between human and animals (protein turnover rates, regulation of ubiquitin-proteasome system and myostatin/activin A-SMAD2/3 signalling pathways) are highlighted and discussed. Identifying the various and intertwined mechanisms that are deregulated during cancer cachexia and understanding why they are decontrolled will provide therapeutic targets for the treatment of skeletal muscle wasting in cancer patients.
Collapse
Affiliation(s)
- Agnès Martin
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| | - Yann S. Gallot
- LBEPS, Univ Evry, IRBA, Université Paris SaclayEvryFrance
| | - Damien Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| |
Collapse
|
3
|
Walsh KA, Kastrappis G, Fifis T, Paolini R, Christophi C, Perini MV. SAR131675, a VEGRF3 Inhibitor, Modulates the Immune Response and Reduces the Growth of Colorectal Cancer Liver Metastasis. Cancers (Basel) 2022; 14:2715. [PMID: 35681695 PMCID: PMC9179346 DOI: 10.3390/cancers14112715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 01/27/2023] Open
Abstract
Most patients with colorectal cancer (CRC) develop metastases, predominantly in the liver (CLM). Targeted therapies are being investigated to improve current CLM treatments. This study tested the effectiveness of SAR131675, a selective VEGFR-3 tyrosine kinase inhibitor, to inhibit CLM in a murine model. Following intrasplenic induction of CLM, mice were treated daily with SAR131675. Tumor growth and immune infiltrates into tumor and liver tissues were assessed at 10-, 16- and 22-days post tumor induction by stereology, IHC and flow cytometry. SAR151675 treatment significantly reduced tumor burden and F4/80+ macrophages in the liver tissues. Analysis of immune cell infiltrates in liver showed tissue that at day 22, had the proportion of CD45+ leukocytes significantly reduced, particularly myeloid cells. Analysis of myeloid cells (CD11b+ CD45+) indicated that the proportion of F4/80- Ly6Clow was significantly reduced, including a predominate PD-L1+ subset, while CD3+ T cells increased, particularly CD8+ PD1+, reflected by an increase in the CD8+:CD4+ T cell ratio. In the tumor tissue SAR11675 treatment reduced the predominant population of F4/80+ Ly6Clo and increased CD4+ T cells. These results suggest that SAR131675 alters the immune composition within tumor and the surrounding liver in the later stages of development, resulting in a less immunosuppressive environment. This immunomodulation effect may contribute to the suppression of tumor growth.
Collapse
Affiliation(s)
- Katrina A. Walsh
- Department of Surgery, The University of Melbourne, Austin Health, Lance Townsend Building, Level 8, 145 Studley Road, Heidelberg, VIC 3084, Australia; (K.A.W.); (G.K.); (T.F.); (C.C.)
| | - Georgios Kastrappis
- Department of Surgery, The University of Melbourne, Austin Health, Lance Townsend Building, Level 8, 145 Studley Road, Heidelberg, VIC 3084, Australia; (K.A.W.); (G.K.); (T.F.); (C.C.)
| | - Theodora Fifis
- Department of Surgery, The University of Melbourne, Austin Health, Lance Townsend Building, Level 8, 145 Studley Road, Heidelberg, VIC 3084, Australia; (K.A.W.); (G.K.); (T.F.); (C.C.)
| | - Rita Paolini
- Melbourne Dental School, The University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia;
| | - Christopher Christophi
- Department of Surgery, The University of Melbourne, Austin Health, Lance Townsend Building, Level 8, 145 Studley Road, Heidelberg, VIC 3084, Australia; (K.A.W.); (G.K.); (T.F.); (C.C.)
| | - Marcos V. Perini
- Department of Surgery, The University of Melbourne, Austin Health, Lance Townsend Building, Level 8, 145 Studley Road, Heidelberg, VIC 3084, Australia; (K.A.W.); (G.K.); (T.F.); (C.C.)
| |
Collapse
|
4
|
Martin A, Freyssenet D. Phenotypic features of cancer cachexia-related loss of skeletal muscle mass and function: lessons from human and animal studies. J Cachexia Sarcopenia Muscle 2021; 12:252-273. [PMID: 33783983 PMCID: PMC8061402 DOI: 10.1002/jcsm.12678] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 12/18/2022] Open
Abstract
Cancer cachexia is a complex multi-organ catabolic syndrome that reduces mobility, increases fatigue, decreases the efficiency of therapeutic strategies, diminishes the quality of life, and increases the mortality of cancer patients. This review provides an exhaustive and comprehensive analysis of cancer cachexia-related phenotypic changes in skeletal muscle at both the cellular and subcellular levels in human cancer patients, as well as in animal models of cancer cachexia. Cancer cachexia is characterized by a major decrease in skeletal muscle mass in human and animals that depends on the severity of the disease/model and the localization of the tumour. It affects both type 1 and type 2 muscle fibres, even if some animal studies suggest that type 2 muscle fibres would be more prone to atrophy. Animal studies indicate an impairment in mitochondrial oxidative metabolism resulting from a decrease in mitochondrial content, an alteration in mitochondria morphology, and a reduction in mitochondrial metabolic fluxes. Immuno-histological analyses in human and animal models also suggest that a faulty mechanism of skeletal muscle repair would contribute to muscle mass loss. An increase in collagen deposit, an accumulation of fat depot outside and inside the muscle fibre, and a disrupted contractile machinery structure are also phenotypic features that have been consistently reported in cachectic skeletal muscle. Muscle function is also profoundly altered during cancer cachexia with a strong reduction in skeletal muscle force. Even though the loss of skeletal muscle mass largely contributes to the loss of muscle function, other factors such as muscle-nerve interaction and calcium handling are probably involved in the decrease in muscle force. Longitudinal analyses of skeletal muscle mass by imaging technics and skeletal muscle force in cancer patients, but also in animal models of cancer cachexia, are necessary to determine the respective kinetics and functional involvements of these factors. Our analysis also emphasizes that measuring skeletal muscle force through standardized tests could provide a simple and robust mean to early diagnose cachexia in cancer patients. That would be of great benefit to cancer patient's quality of life and health care systems.
Collapse
Affiliation(s)
- Agnès Martin
- Inter‐university Laboratory of Human Movement BiologyUniversité de Lyon, University Jean Monnet Saint‐EtienneSaint‐ÉtienneFrance
| | - Damien Freyssenet
- Inter‐university Laboratory of Human Movement BiologyUniversité de Lyon, University Jean Monnet Saint‐EtienneSaint‐ÉtienneFrance
| |
Collapse
|
5
|
MC38 Tumors Induce Musculoskeletal Defects in Colorectal Cancer. Int J Mol Sci 2021; 22:ijms22031486. [PMID: 33540821 PMCID: PMC7867345 DOI: 10.3390/ijms22031486] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related death, and the prevalence of CRC in young adults is on the rise, making this a largescale clinical concern. Advanced CRC patients often present with liver metastases (LM) and an increased incidence of cachexia, i.e., musculoskeletal wasting. Despite its high incidence in CRC patients, cachexia remains an unresolved issue, and animal models for the study of CRC cachexia, in particular, metastatic CRC cachexia, remain limited; therefore, we aimed to establish a new model of metastatic CRC cachexia. C57BL/6 male mice (8 weeks old) were subcutaneously (MC38) or intrasplenically injected (mMC38) with MC38 murine CRC cells to disseminate LM, while experimental controls received saline (n = 5-8/group). The growth of subcutaneous MC38 tumors was accompanied by a reduction in skeletal muscle mass (-16%; quadriceps muscle), plantarflexion force (-22%) and extensor digitorum longus (EDL) contractility (-20%) compared to experimental controls. Meanwhile, the formation of MC38 LM (mMC38) led to heighted reductions in skeletal muscle mass (-30%; quadriceps), plantarflexion force (-28%) and EDL contractility (-35%) compared to sham-operated controls, suggesting exacerbated cachexia associated with LM. Moreover, both MC38 and mMC38 tumor hosts demonstrated a marked loss of bone indicated by reductions in trabecular (Tb.BV/TV: -49% in MC38, and -46% in mMC38) and cortical (C.BV/TV: -12% in MC38, and -8% in mMC38) bone. Cell culture experiments revealed that MC38 tumor-derived factors directly promote myotube wasting (-18%) and STAT3 phosphorylation (+5-fold), while the pharmacologic blockade of STAT3 signaling was sufficient to preserve myotube atrophy in the presence of MC38 cells (+21%). Overall, these results reinforce the notion that the formation of LM heightens cachexia in an experimental model of CRC.
Collapse
|
6
|
Huot JR, Pin F, Narasimhan A, Novinger LJ, Keith AS, Zimmers TA, Willis MS, Bonetto A. ACVR2B antagonism as a countermeasure to multi-organ perturbations in metastatic colorectal cancer cachexia. J Cachexia Sarcopenia Muscle 2020; 11:1779-1798. [PMID: 33200567 PMCID: PMC7749603 DOI: 10.1002/jcsm.12642] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/11/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Advanced colorectal cancer (CRC) is often accompanied by the development of liver metastases, as well as cachexia, a multi-organ co-morbidity primarily affecting skeletal (SKM) and cardiac muscles. Activin receptor type 2B (ACVR2B) signalling is known to cause SKM wasting, and its inhibition restores SKM mass and prolongs survival in cancer. Using a recently generated mouse model, here we tested whether ACVR2B blockade could preserve multiple organs, including skeletal and cardiac muscle, in the presence of metastatic CRC. METHODS NSG male mice (8 weeks old) were injected intrasplenically with HCT116 human CRC cells (mHCT116), while sham-operated animals received saline (n = 5-10 per group). Sham and tumour-bearing mice received weekly injections of ACVR2B/Fc, a synthetic peptide inhibitor of ACVR2B. RESULTS mHCT116 hosts displayed losses in fat mass ( - 79%, P < 0.0001), bone mass ( - 39%, P < 0.05), and SKM mass (quadriceps: - 22%, P < 0.001), in line with reduced muscle cross-sectional area ( - 24%, P < 0.01) and plantarflexion force ( - 28%, P < 0.05). Further, despite only moderately affected heart size, cardiac function was significantly impaired (ejection fraction %: - 16%, P < 0.0001; fractional shortening %: - 25%, P < 0.0001) in the mHCT116 hosts. Conversely, ACVR2B/Fc preserved fat mass ( + 238%, P < 0.001), bone mass ( + 124%, P < 0.0001), SKM mass (quadriceps: + 31%, P < 0.0001), size (cross-sectional area: + 43%, P < 0.0001) and plantarflexion force ( + 28%, P < 0.05) in tumour hosts. Cardiac function was also completely preserved in tumour hosts receiving ACVR2B/Fc (ejection fraction %: + 19%, P < 0.0001), despite no effect on heart size. RNA sequencing analysis of heart muscle revealed rescue of genes related to cardiac development and contraction in tumour hosts treated with ACVR2B/Fc. CONCLUSIONS Our metastatic CRC model recapitulates the multi-systemic derangements of cachexia by displaying loss of fat, bone, and SKM along with decreased muscle strength in mHCT116 hosts. Additionally, with evidence of severe cardiac dysfunction, our data support the development of cardiac cachexia in the occurrence of metastatic CRC. Notably, ACVR2B antagonism preserved adipose tissue, bone, and SKM, whereas muscle and cardiac functions were completely maintained upon treatment. Altogether, our observations implicate ACVR2B signalling in the development of multi-organ perturbations in metastatic CRC and further dictate that ACVR2B represents a promising therapeutic target to preserve body composition and functionality in cancer cachexia.
Collapse
Affiliation(s)
- Joshua R Huot
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ashok Narasimhan
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leah J Novinger
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Teresa A Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.,Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.,Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.,Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
7
|
Huot JR, Novinger LJ, Pin F, Narasimhan A, Zimmers TA, O'Connell TM, Bonetto A. Formation of colorectal liver metastases induces musculoskeletal and metabolic abnormalities consistent with exacerbated cachexia. JCI Insight 2020; 5:136687. [PMID: 32298240 DOI: 10.1172/jci.insight.136687] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/08/2020] [Indexed: 12/18/2022] Open
Abstract
Advanced colorectal cancer (CRC) is often accompanied by development of liver metastases (LMs) and skeletal muscle wasting (i.e., cachexia). Despite plaguing the majority of CRC patients, cachexia remains unresolved. By using mice injected with Colon-26 mouse tumors, either subcutaneously (s.c.; C26) or intrasplenically to mimic hepatic dissemination of cancer cells (mC26), here we aimed to further characterize functional, molecular, and metabolic effects on skeletal muscle and examine whether LMs exacerbate CRC-induced cachexia. C26-derived LMs were associated with progressive loss of body weight, as well as with significant reductions in skeletal muscle size and strength, in line with reduced phosphorylation of markers of protein anabolism and enhanced protein catabolism. mC26 hosts showed prevalence of fibers with glycolytic metabolism and enhanced lipid accumulation, consistent with abnormalities of mitochondrial homeostasis and energy metabolism. In a comparison with mice bearing s.c. C26, cachexia appeared exacerbated in the mC26 hosts, as also supported by differentially expressed pathways within skeletal muscle. Overall, our model recapitulates the cachectic phenotype of metastatic CRC and reveals that formation of LMs resulting from CRC exacerbate cancer-induced skeletal muscle wasting by promoting differential gene expression signatures.
Collapse
Affiliation(s)
- Joshua R Huot
- Department of Surgery.,Department of Anatomy, Cell Biology and Physiology
| | | | - Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology
| | | | - Teresa A Zimmers
- Department of Surgery.,Department of Anatomy, Cell Biology and Physiology.,Simon Cancer Center, and.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Thomas M O'Connell
- Department of Anatomy, Cell Biology and Physiology.,Department of Otolaryngology-Head & Neck Surgery.,Simon Cancer Center, and.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrea Bonetto
- Department of Surgery.,Department of Anatomy, Cell Biology and Physiology.,Department of Otolaryngology-Head & Neck Surgery.,Simon Cancer Center, and.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
8
|
Huot JR, Novinger LJ, Pin F, Bonetto A. HCT116 colorectal liver metastases exacerbate muscle wasting in a mouse model for the study of colorectal cancer cachexia. Dis Model Mech 2020; 13:dmm043166. [PMID: 31915140 PMCID: PMC6994937 DOI: 10.1242/dmm.043166] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/16/2019] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is often accompanied by formation of liver metastases (LM) and skeletal muscle wasting, i.e. cachexia. Despite affecting the majority of CRC patients, cachexia remains underserved, understudied and uncured. Animal models for the study of CRC-induced cachexia, in particular models containing LM, are sparse; therefore, we aimed to characterize two new models of CRC cachexia. Male NSG mice were injected subcutaneously (HCT116) or intrasplenically (mHCT116) with human HCT116 CRC tumor cells to disseminate LM, whereas experimental controls received saline (n=5-8/group). Tumor growth was accompanied by loss of skeletal muscle mass (HCT116: -20%; mHCT116: -31%; quadriceps muscle) and strength (HCT116: -20%; mHCT116: -27%), with worsened loss of skeletal muscle mass in mHCT116 compared with HCT116 (gastrocnemius: -19%; tibialis anterior: -22%; quadriceps: -21%). Molecular analyses revealed elevated protein ubiquitination in HCT116, whereas mHCT116 also displayed elevated Murf1 and atrogin-1 expression, along with reduced mitochondrial proteins PGC1α, OPA1, mitofusin 2 and cytochrome C. Further, elevated IL6 levels were found in the blood of mHCT116 hosts, which was associated with higher phosphorylation of STAT3 in skeletal muscle. To clarify whether STAT3 was a main player in muscle wasting in this model, HCT116 cells were co-cultured with C2C12 myotubes. Marked myotube atrophy (-53%) was observed, along with elevated phospho-STAT3 levels (+149%). Conversely, inhibition of STAT3 signaling by means of a JAK/STAT3 inhibitor was sufficient to rescue myotube atrophy induced by HCT116 cells (+55%). Overall, our results indicate that the formation of LM exacerbates cachectic phenotype and associated skeletal muscle molecular alterations in HCT116 tumor hosts.
Collapse
Affiliation(s)
- Joshua R Huot
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Leah J Novinger
- Department of Otolaryngology - Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Otolaryngology - Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- IUPUI Center for Cachexia Research Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
9
|
Tomasin R, Martin ACBM, Cominetti MR. Metastasis and cachexia: alongside in clinics, but not so in animal models. J Cachexia Sarcopenia Muscle 2019; 10:1183-1194. [PMID: 31436396 PMCID: PMC6903449 DOI: 10.1002/jcsm.12475] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 05/06/2019] [Accepted: 06/12/2019] [Indexed: 12/16/2022] Open
Abstract
Cancer cachexia is a paraneoplastic syndrome characterized by lean mass wasting (with or without fat mass decrease), culminating in involuntary weight loss, which is the key clinical observation nowadays. There is a notable lack of studies involving animal models to mimic the clinical reality, which are mostly patients with cachexia and metastatic disease. This mismatch between the clinical reality and animal models could at least partly contribute to the poor translation observed in the field. In this paper, we retrieved and compared animal models used for cachexia research from 2017 and 10 years earlier (2007) and observed that very little has changed. Especially, clinically relevant models where cachexia is studied in an orthotopic or metastatic context were and still are very scarce. Finally, we described and supported the biological rationale behind why, despite technical challenges, these two phenomena-metastasis and cachexia-should be modelled in parallel, highlighting the overlapping pathways between them. To sum up, this review aims to contribute to rethinking and possibly switching the models currently used for cachexia research, to hopefully obtain better and more translational outcomes.
Collapse
Affiliation(s)
- Rebeka Tomasin
- Laboratory of Biology of Aging (LABEN), Department of Gerontology, Federal University of São Carlos, São Carlos, Brazil
| | | | - Márcia Regina Cominetti
- Laboratory of Biology of Aging (LABEN), Department of Gerontology, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|
10
|
Pin F, Novinger LJ, Huot JR, Harris RA, Couch ME, O'Connell TM, Bonetto A. PDK4 drives metabolic alterations and muscle atrophy in cancer cachexia. FASEB J 2019; 33:7778-7790. [PMID: 30894018 DOI: 10.1096/fj.201802799r] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cachexia is frequently accompanied by severe metabolic derangements, although the mechanisms responsible for this debilitating condition remain unclear. Pyruvate dehydrogenase kinase (PDK)4, a critical regulator of cellular energetic metabolism, was found elevated in experimental models of cancer, starvation, diabetes, and sepsis. Here we aimed to investigate the link between PDK4 and the changes in muscle size in cancer cachexia. High PDK4 and abnormal energetic metabolism were found in the skeletal muscle of colon-26 tumor hosts, as well as in mice fed a diet enriched in Pirinixic acid, previously shown to increase PDK4 levels. Viral-mediated PDK4 overexpression in myotube cultures was sufficient to promote myofiber shrinkage, consistent with enhanced protein catabolism and mitochondrial abnormalities. On the contrary, blockade of PDK4 was sufficient to restore myotube size in C2C12 cultures exposed to tumor media. Our data support, for the first time, a direct role for PDK4 in promoting cancer-associated muscle metabolic alterations and skeletal muscle atrophy.-Pin, F., Novinger, L. J., Huot, J. R., Harris, R. A., Couch, M. E., O'Connell, T. M., Bonetto, A. PDK4 drives metabolic alterations and muscle atrophy in cancer cachexia.
Collapse
Affiliation(s)
- Fabrizio Pin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana University-Purdue University Indianapolis Center for Cachexia Research, Innovation, and Therapy, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Leah J Novinger
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Joshua R Huot
- Indiana University-Purdue University Indianapolis Center for Cachexia Research, Innovation, and Therapy, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert A Harris
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Marion E Couch
- Indiana University-Purdue University Indianapolis Center for Cachexia Research, Innovation, and Therapy, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Thomas M O'Connell
- Indiana University-Purdue University Indianapolis Center for Cachexia Research, Innovation, and Therapy, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrea Bonetto
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana University-Purdue University Indianapolis Center for Cachexia Research, Innovation, and Therapy, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
11
|
Hardee JP, Counts BR, Carson JA. Understanding the Role of Exercise in Cancer Cachexia Therapy. Am J Lifestyle Med 2019; 13:46-60. [PMID: 30627079 PMCID: PMC6311610 DOI: 10.1177/1559827617725283] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/12/2017] [Accepted: 07/19/2017] [Indexed: 12/17/2022] Open
Abstract
Cachexia, the unintentional loss of body weight, is prevalent in many cancer types, and the associated skeletal muscle mass depletion increases patient morbidity and mortality. While anorexia can be present, cachexia is not reversible with nutritional therapies alone. Pharmacological agents have been proposed to treat this condition, but there are currently no approved treatments. Nonetheless, the hallmark characteristics associated with cancer cachexia remain viable foundations for future therapies. Regular physical activity holds a promising future as a nonpharmacological alternative to improve patient survival through cachexia prevention. Evidence suggests exercise training is beneficial during cancer treatment and survival. However, the mechanistic examination of cachectic skeletal muscle's response to exercise is both needed and justified. The primary objective of this review is to discuss the role of exercise for the prevention and treatment of cancer-associated muscle wasting. Initially, we provide an overview of systemic alterations induced by cancer and their role in the regulation of wasting processes during cachexia progression. We then discuss how exercise could alter disrupted regulatory pathways related to growth and metabolism during cancer-induced muscle atrophy. Last, we outline current exercise prescription guidelines and how exercise could be a potential behavioral therapy to curtail cachexia development in cancer patients.
Collapse
Affiliation(s)
- Justin P. Hardee
- Department of Exercise Science (JPH, BRC, JAC), University of South Carolina, Columbia, South Carolina
- Center for Colon Cancer Research (JAC), University of South Carolina, Columbia, South Carolina
| | - Brittany R. Counts
- Department of Exercise Science (JPH, BRC, JAC), University of South Carolina, Columbia, South Carolina
- Center for Colon Cancer Research (JAC), University of South Carolina, Columbia, South Carolina
| | - James A. Carson
- James A. Carson, PhD, Department of Exercise Science, University of South Carolina, 921 Assembly Street, Public Health Research Center, Rm 301, Columbia, SC 29208; e-mail:
| |
Collapse
|
12
|
Meyerhardt JA, Kroenke CH, Prado CM, Kwan ML, Castillo A, Weltzien E, Cespedes Feliciano EM, Xiao J, Caan BJ. Association of Weight Change after Colorectal Cancer Diagnosis and Outcomes in the Kaiser Permanente Northern California Population. Cancer Epidemiol Biomarkers Prev 2016; 26:30-37. [PMID: 27986654 DOI: 10.1158/1055-9965.epi-16-0145] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Higher body mass index (BMI) is associated with incident colorectal cancer but not consistently with colorectal cancer survival. Whether weight gain or loss is associated with colorectal cancer survival is largely unknown. METHODS We identified 2,781 patients from Kaiser Permanente Northern California diagnosed with stages I-III colorectal cancer between 2006 and 2011 with weight and height measurements within 3 months of diagnosis and approximately 18 months after diagnosis. We evaluated associations between weight change and colorectal cancer-specific and overall mortality, adjusted for sociodemographics, disease severity, and treatment. RESULTS After completion of treatment and recovery from stage I-III colorectal cancer, loss of at least 10% of baseline weight was associated with significantly worse colorectal cancer-specific mortality (HR 3.20; 95% confidence interval [CI], 2.33-4.39; Ptrend < 0.0001) and overall mortality (HR 3.27; 95% CI, 2.56-4.18; Ptrend < 0.0001). For every 5% loss of baseline weight, there was a 41% increased risk of colorectal cancer-specific mortality (95% CI, 29%-56%). Weight gain was not significantly associated with colorectal cancer-specific mortality (Ptrend = 0.54) or overall mortality (Ptrend = 0.27). The associations were largely unchanged after restricting analyses to exclude patients who died within 6 months and 12 months of the second weight measurement. No significant interactions were demonstrated for weight loss or gain by gender, stage, primary tumor location, or baseline BMI. CONCLUSIONS Weight loss after diagnosis was associated with worse colorectal cancer-specific mortality and overall mortality. Reverse causation does not appear to explain our findings. IMPACT Understanding mechanistic underpinnings for the association of weight to worse mortality is important to improving patient outcomes. Cancer Epidemiol Biomarkers Prev; 26(1); 30-37. ©2016 AACR SEE ALL THE ARTICLES IN THIS CEBP FOCUS SECTION, "THE OBESITY PARADOX IN CANCER EVIDENCE AND NEW DIRECTIONS".
Collapse
Affiliation(s)
| | - Candyce H Kroenke
- Kaiser Permanente Northern California Division of Research, Oakland, California
| | - Carla M Prado
- Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Marilyn L Kwan
- Kaiser Permanente Northern California Division of Research, Oakland, California
| | - Adrienne Castillo
- Kaiser Permanente Northern California Division of Research, Oakland, California
| | - Erin Weltzien
- Kaiser Permanente Northern California Division of Research, Oakland, California
| | | | - Jingjie Xiao
- Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Bette J Caan
- Kaiser Permanente Northern California Division of Research, Oakland, California
| |
Collapse
|
13
|
Lee-Young RS, Hoffman NJ, Murphy KT, Henstridge DC, Samocha-Bonet D, Siebel AL, Iliades P, Zivanovic B, Hong YH, Colgan TD, Kraakman MJ, Bruce CR, Gregorevic P, McConell GK, Lynch GS, Drummond GR, Kingwell BA, Greenfield JR, Febbraio MA. Glucose-6-phosphate dehydrogenase contributes to the regulation of glucose uptake in skeletal muscle. Mol Metab 2016; 5:1083-1091. [PMID: 27818934 PMCID: PMC5081409 DOI: 10.1016/j.molmet.2016.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 08/29/2016] [Accepted: 09/05/2016] [Indexed: 12/25/2022] Open
Abstract
Objective The development of skeletal muscle insulin resistance is an early physiological defect, yet the intracellular mechanisms accounting for this metabolic defect remained unresolved. Here, we have examined the role of glucose-6-phosphate dehydrogenase (G6PDH) activity in the pathogenesis of insulin resistance in skeletal muscle. Methods Multiple mouse disease states exhibiting insulin resistance and glucose intolerance, as well as obese humans defined as insulin-sensitive, insulin-resistant, or pre-diabetic, were examined. Results We identified increased glucose-6-phosphate dehydrogenase (G6PDH) activity as a common intracellular adaptation that occurs in parallel with the induction of insulin resistance in skeletal muscle and is present across animal and human disease states with an underlying pathology of insulin resistance and glucose intolerance. We observed an inverse association between G6PDH activity and nitric oxide synthase (NOS) activity and show that increasing NOS activity via the skeletal muscle specific neuronal (n)NOSμ partially suppresses G6PDH activity in skeletal muscle cells. Furthermore, attenuation of G6PDH activity in skeletal muscle cells via (a) increased nNOSμ/NOS activity, (b) pharmacological G6PDH inhibition, or (c) genetic G6PDH inhibition increases insulin-independent glucose uptake. Conclusions We have identified a novel, previously unrecognized role for G6PDH in the regulation of skeletal muscle glucose metabolism. Defective skeletal muscle G6PDH activity in multiple insulin resistant animal models. Demonstration of defective skeletal muscle G6PDH activity in pre-diabetic individuals. Identification of nNOSμ as a regulator of G6PDH activity in skeletal muscle. G6PDH activity modulates insulin-independent glucose uptake in skeletal muscle.
Collapse
Affiliation(s)
- Robert S Lee-Young
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia.
| | - Nolan J Hoffman
- Diabetes & Metabolism Division, Garvan Institute of Medical Research, NSW, Australia
| | - Kate T Murphy
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - Darren C Henstridge
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Dorit Samocha-Bonet
- Diabetes & Metabolism Division, Garvan Institute of Medical Research, NSW, Australia
| | - Andrew L Siebel
- Metabolic and Vascular Physiology Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Peter Iliades
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Borivoj Zivanovic
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Yet H Hong
- Institute for Sports, Exercise and Active Living, Victoria University, Footscray, VIC, Australia
| | - Timothy D Colgan
- Muscle Research and Therapeutics Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Michael J Kraakman
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Clinton R Bruce
- School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, Australia
| | - Paul Gregorevic
- Muscle Research and Therapeutics Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Glenn K McConell
- Institute for Sports, Exercise and Active Living, Victoria University, Footscray, VIC, Australia
| | - Gordon S Lynch
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - Grant R Drummond
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Bronwyn A Kingwell
- Metabolic and Vascular Physiology Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Jerry R Greenfield
- Diabetes & Metabolism Division, Garvan Institute of Medical Research, NSW, Australia
| | - Mark A Febbraio
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia; Diabetes & Metabolism Division, Garvan Institute of Medical Research, NSW, Australia.
| |
Collapse
|
14
|
Murphy KT. The pathogenesis and treatment of cardiac atrophy in cancer cachexia. Am J Physiol Heart Circ Physiol 2015; 310:H466-77. [PMID: 26718971 DOI: 10.1152/ajpheart.00720.2015] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/29/2015] [Indexed: 02/08/2023]
Abstract
Cancer cachexia is a multifactorial syndrome characterized by a progressive loss of skeletal muscle mass associated with significant functional impairment. In addition to a loss of skeletal muscle mass and function, many patients with cancer cachexia also experience cardiac atrophy, remodeling, and dysfunction, which in the field of cancer cachexia is described as cardiac cachexia. The cardiac alterations may be due to underlying heart disease, the cancer itself, or problems initiated by the cancer treatment and, unfortunately, remains largely underappreciated by clinicians and basic scientists. Despite recent major advances in the treatment of cancer, little progress has been made in the treatment of cardiac cachexia in cancer, and much of this is due to lack of information regarding the mechanisms. This review focuses on the cardiac atrophy associated with cancer cachexia, describing some of the known mechanisms and discussing the current and future therapeutic strategies to treat this condition. Above all else, improved awareness of the condition and an increased focus on identification of mechanisms and therapeutic targets will facilitate the eventual development of an effective treatment for cardiac atrophy in cancer cachexia.
Collapse
Affiliation(s)
- Kate T Murphy
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
15
|
Carson JA, Hardee JP, VanderVeen BN. The emerging role of skeletal muscle oxidative metabolism as a biological target and cellular regulator of cancer-induced muscle wasting. Semin Cell Dev Biol 2015; 54:53-67. [PMID: 26593326 DOI: 10.1016/j.semcdb.2015.11.005] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/12/2015] [Indexed: 12/17/2022]
Abstract
While skeletal muscle mass is an established primary outcome related to understanding cancer cachexia mechanisms, considerable gaps exist in our understanding of muscle biochemical and functional properties that have recognized roles in systemic health. Skeletal muscle quality is a classification beyond mass, and is aligned with muscle's metabolic capacity and substrate utilization flexibility. This supplies an additional role for the mitochondria in cancer-induced muscle wasting. While the historical assessment of mitochondria content and function during cancer-induced muscle loss was closely aligned with energy flux and wasting susceptibility, this understanding has expanded to link mitochondria dysfunction to cellular processes regulating myofiber wasting. The primary objective of this article is to highlight muscle mitochondria and oxidative metabolism as a biological target of cancer cachexia and also as a cellular regulator of cancer-induced muscle wasting. Initially, we examine the role of muscle metabolic phenotype and mitochondria content in cancer-induced wasting susceptibility. We then assess the evidence for cancer-induced regulation of skeletal muscle mitochondrial biogenesis, dynamics, mitophagy, and oxidative stress. In addition, we discuss environments associated with cancer cachexia that can impact the regulation of skeletal muscle oxidative metabolism. The article also examines the role of cytokine-mediated regulation of mitochondria function, followed by the potential role of cancer-induced hypogonadism. Lastly, a role for decreased muscle use in cancer-induced mitochondrial dysfunction is reviewed.
Collapse
Affiliation(s)
- James A Carson
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, 921 Assembly St., Columbia, SC, 29208, USA.
| | - Justin P Hardee
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, 921 Assembly St., Columbia, SC, 29208, USA
| | - Brandon N VanderVeen
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, 921 Assembly St., Columbia, SC, 29208, USA
| |
Collapse
|