Chemotherapy impairs skeletal muscle mitochondrial homeostasis in early breast cancer patients

Joris Mallard, Elyse Hucteau, Anne-Laure Charles, Laura Bender, Claire Baeza, Mathilde Pélissie, Philippe Trensz, Carole Pflumio, Michal Kalish-Weindling, Bernard Gény, Roland Schott, Fabrice Favret, Xavier Pivot, Thomas J Hureau, Allan F Pagano, Joris Mallard, Elyse Hucteau, Anne-Laure Charles, Laura Bender, Claire Baeza, Mathilde Pélissie, Philippe Trensz, Carole Pflumio, Michal Kalish-Weindling, Bernard Gény, Roland Schott, Fabrice Favret, Xavier Pivot, Thomas J Hureau, Allan F Pagano

Abstract

Background: Chemotherapy is extensively used to treat breast cancer and is associated with skeletal muscle deconditioning, which is known to reduce patients' quality of life, treatment efficiency, and overall survival. To date, skeletal muscle mitochondrial alterations represent a major aspect explored in breast cancer patients; nevertheless, the cellular mechanisms remain relatively unknown. This study was dedicated to investigating overall skeletal muscle mitochondrial homeostasis in early breast cancer patients undergoing chemotherapy, including mitochondrial quantity, function, and dynamics.

Methods: Women undergoing (neo)adjuvant anthracycline-cyclophosphamide and taxane-based chemotherapy participated in this study (56 ± 12 years). Two muscle biopsies were collected from the vastus lateralis muscle before the first and after the last chemotherapy administration. Mitochondrial respiratory capacity, reactive oxygen species production, and western blotting analyses were performed.

Results: Among the 11 patients, we found a decrease in key markers of mitochondrial quantity, reaching -52.0% for citrate synthase protein levels (P = 0.02) and -38.2% for VDAC protein levels (P = 0.04). This mitochondrial content loss is likely explained by reduced mitochondrial biogenesis, as evidenced by a decrease in PGC-1α1 protein levels (-29.5%; P = 0.04). Mitochondrial dynamics were altered, as documented by a decrease in MFN2 protein expression (-33.4%; P = 0.01), a key marker of mitochondrial outer membrane fusion. Mitochondrial fission is a prerequisite for mitophagy activation, and no variation was found in either key markers of mitochondrial fission (Fis1 and DRP1) or mitophagy (Parkin, PINK1, and Mul1). Two contradictory hypotheses arise from these results: defective mitophagy, which probably increases the number of damaged and fragmented mitochondria, or a relative increase in mitophagy through elevated mitophagic potential (Parkin/VDAC ratio; +176.4%; P < 0.02). Despite no change in mitochondrial respiratory capacity and COX IV protein levels, we found an elevation in H2 O2 production (P < 0.05 for all substrate additions) without change in antioxidant enzymes. We investigated the apoptosis pathway and found an increase in the protein expression of the apoptosis initiation marker Bax (+72.0%; P = 0.04), without variation in the anti-apoptotic protein Bcl-2.

Conclusions: This study demonstrated major mitochondrial alterations subsequent to chemotherapy in early breast cancer patients: (i) a striking reduction in mitochondrial biogenesis, (ii) altered mitochondrial dynamics and potential mitophagy defects, (iii) exacerbated H2 O2 production, and (iv) increased initiation of apoptosis. All of these alterations likely explain, at least in part, the high prevalence of skeletal muscle and cardiorespiratory deconditioning classically observed in breast cancer patients.

Keywords: Apoptosis; H2O2; Mitochondrial biogenesis; Mitochondrial dynamics; Mitophagy; Skeletal muscle deconditioning.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

© 2022 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.

Figures

Figure 1
Figure 1
Flow chart of the participants.
Figure 2
Figure 2
Changes in key markers of mitochondrial quantity and mitochondrial biogenesis. Citrate synthase (A), VDAC (B), and PGC‐1α1 (C) protein levels from vastus lateralis muscle biopsies taken before (pre‐CT) and after (post‐CT) breast cancer chemotherapy (CT). Above each panel, western blots from two representative subjects are displayed. All values are expressed as the mean ± SD (N = 11). *P < 0.05.
Figure 3
Figure 3
Changes in protein markers of mitochondrial dynamics and mitophagy. MFN2 (A); MFN1 85 and 71 kDa isoforms (B); OPA1 isoforms: L, S1, S2, and S3 (C); Fis1 (D); p‐DRP1 (Ser616)/DRP1 (E); Mul1 (F); Parkin (G); PINK1 (H); and Parkin/VDAC (I) protein levels from vastus lateralis muscle biopsies taken before (pre) and after (post) breast cancer chemotherapy (CT). Above each panel, western blots from two representative subjects are displayed. All values are expressed as the mean ± SD (N = 11). *P < 0.05.
Figure 4
Figure 4
Changes in mitochondrial respiration, reactive oxygen species production, and redox signalling protein markers. Mitochondrial respiratory capacity quantification (A), and associated mean representative curves (B), for CI‐linked substrate state, OXPHOS by CI, OXPHOS by CI&CII, and OXPHOS by CII on permeabilized fibres from vastus lateralis muscle biopsies taken before (pre) and after (post) breast cancer chemotherapy (CT). COX IV (C) protein levels in pre‐CT vs. post‐CT. H2O2 production quantification (D), and associated mean representative curves (E), for CI‐linked substrate state, OXPHOS by CI, OXPHOS by CI&CII, and OXPHOS by CII in pre‐CT vs. post‐CT permeabilized fibres. Superoxide anion measurement (F) through electron paramagnetic resonance in pre‐CT vs. post‐CT minced muscle samples. 4‐HNE (G), total carbonylated (H), SOD2 (I), catalase (J), GPx1 (K), p‐eIF2α (Ser51)/eIF2α (L), p‐p38 MAPK (Thr180/Tyr182)/p38 MAPK (M) and p‐p53 (Ser15)/p53 (N) protein levels in pre‐CT vs. post‐CT. Above each panel with western blot analyses, two representative subjects are displayed. All values are expressed as the mean ± SD (N = 11). *P < 0.05; **P < 0.01; ***P < 0.001; CI: complex I; CII: complex II; OXPHOS: oxidative phosphorylation.
Figure 5
Figure 5
Changes in protein markers of apoptosis induction. Bax (A), Bcl‐2 (B), and active/pro‐caspase 3 (C) protein levels from vastus lateralis muscle biopsies taken before (pre) and after (post) breast cancer chemotherapy (CT). Above each panel, western blots from two representative subjects are displayed. All values are expressed as the mean ± SD (N = 11). *P < 0.05.
Figure 6
Figure 6
Summary of the mitochondrial alterations observed in early breast cancer patients undergoing chemotherapy. Through the analysis of vastus lateralis muscle biopsies taken before (pre) and after (post) breast cancer chemotherapy (CT), we identified an increased mitochondrial loss (grey box) that may be explained by a decrease in PGC‐1α1 protein levels, reflecting a reduced mitochondrial biogenesis process (in blue). Mitochondrial dynamics (in red) were also impaired, as evidenced by a dysregulation in the fusion process, mediated by a decrease in the mitochondrial outer membrane fusion marker MFN2, without any change in MFN1 and OPA1. No change was observed in fission markers (Fis1 and DRP1), indicating a dysregulation in the mitochondrial fusion/fission balance. These alterations are likely leading to the accumulation of fragmented and damaged mitochondria (rose‐coloured mitochondria), corroborated by the accumulation of S3‐OPA1 isoform. As a consequence, fragmented and damaged mitochondria accumulate in skeletal muscle and produce excessive amounts of reactive oxygen species such as H2O2 (in orange). As mentioned above, we found no change in mitochondrial fission, a prerequisite step for the activation of mitophagy. Despite the absence of change in the expression of different mitophagy markers (Parkin, PINK1, and Mul1; in green), two contradictory hypotheses arise from these results: defective mitophagy (Hypothesis 1) or a relative increase in mitophagy (increased mitophagic potential reflected by the Parkin/VDAC ratio; Hypothesis 2). Antioxidant enzyme (in orange), lipid peroxidation (in brown), and carbonylated protein levels (in yellow) were unchanged. However, we found a large increase in Bax protein levels, a proapoptotic protein that initiates apoptosis, and no change was observed for Bcl‐2, its antiapoptotic counterpart (in black). While several pre‐clinical studies found increased levels in the active (cleaved)‐caspase 3/pro‐caspase 3 ratio, the slight increase observed in our study did not reach significance (in black dotted line).

References

    1. Denduluri N, Chavez‐MacGregor M, Telli ML, Eisen A, Graff SL, Hassett MJ, et al. Selection of optimal adjuvant chemotherapy and targeted therapy for early breast cancer: ASCO clinical practice guideline focused update. J Clin Oncol Off J Am Soc Clin Oncol 2018;36:2433–2443.
    1. Mallard J, Hucteau E, Hureau TJ, Pagano AF. Skeletal muscle deconditioning in breast cancer patients undergoing chemotherapy: current knowledge and insights from other cancers. Front Cell Dev Biol 2021;9:2550.
    1. 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.
    1. Brioche T, Pagano AF, Py G, Chopard A. Muscle wasting and aging: experimental models, fatty infiltrations, and prevention. Mol Aspects Med 2016;50:56–87.
    1. Chopard A, Hillock S, Jasmin BJ. Molecular events and signalling pathways involved in skeletal muscle disuse‐induced atrophy and the impact of countermeasures. J Cell Mol Med 2009;13:3032–3050.
    1. Beltrà M, Pin F, Ballarò R, Costelli P, Penna F. Mitochondrial dysfunction in cancer cachexia: impact on muscle health and regeneration. Cell 2021;10:3150.
    1. Hyatt HW, Powers SK. Mitochondrial dysfunction is a common denominator linking skeletal muscle wasting due to disease, aging, and prolonged inactivity. Antioxid Basel Switz 2021;10:588.
    1. Kemp PR, Paul R, Hinken AC, Neil D, Russell A, Griffiths MJ. Metabolic profiling shows pre‐existing mitochondrial dysfunction contributes to muscle loss in a model of ICU‐acquired weakness. J Cachexia Sarcopenia Muscle 2020;11:1321–1335.
    1. Mallard J, Hucteau E, Schott R, Petit T, Demarchi M, Belletier C, et al. Evolution of physical status from diagnosis to the end of first‐line treatment in breast, lung, and colorectal cancer patients: the PROTECT‐01 cohort study protocol. Front Oncol 2020;10:1304, 10.3389/fonc.2020.01304
    1. Caan BJ, Cespedes Feliciano EM, Prado CM, Alexeeff S, Kroenke CH, Bradshaw P, et al. Association of muscle and adiposity measured by computed tomography with survival in patients with nonmetastatic breast cancer. JAMA Oncol 2018;4:798–804.
    1. Baracos VE, Martin L, Korc M, Guttridge DC, Fearon KCH. Cancer‐associated cachexia. Nat Rev Dis Primers 2018;4:17105.
    1. Gouspillou G, Scheede‐Bergdahl C, Spendiff S, Vuda M, Meehan B, Mlynarski H, et al. Anthracycline‐containing chemotherapy causes long‐term impairment of mitochondrial respiration and increased reactive oxygen species release in skeletal muscle. Sci Rep 2015;5:8717.
    1. Nissinen TA, Degerman J, Räsänen M, Poikonen AR, Koskinen S, Mervaala E, et al. Systemic blockade of ACVR2B ligands prevents chemotherapy‐induced muscle wasting by restoring muscle protein synthesis without affecting oxidative capacity or atrogenes. Sci Rep 2016;6:32695.
    1. Hiensch AE, Bolam KA, Mijwel S, Jeneson JAL, Huitema ADR, Kranenburg O, et al. Doxorubicin‐induced skeletal muscle atrophy: elucidating the underlying molecular pathways. Acta Physiol (Oxf) 2020;229:e13400.
    1. Bohlen J, McLaughlin SL, Hazard‐Jenkins H, Infante AM, Montgomery C, Davis M, et al. Dysregulation of metabolic‐associated pathways in muscle of breast cancer patients: preclinical evaluation of interleukin‐15 targeting fatigue. J Cachexia Sarcopenia Muscle 2018;9:701–714.
    1. Guigni BA, Callahan DM, Tourville TW, Miller MS, Fiske B, Voigt T, et al. Skeletal muscle atrophy and dysfunction in breast cancer patients: role for chemotherapy‐derived oxidant stress. Am J Physiol Cell Physiol 2018;315:C744–C756.
    1. Mijwel S, Cardinale DA, Norrbom J, Chapman M, Ivarsson N, Wengström Y, et al. Exercise training during chemotherapy preserves skeletal muscle fiber area, capillarization, and mitochondrial content in patients with breast cancer. FASEB J 2018;32:5495–5505.
    1. Wilson HE, Stanton DA, Rellick S, Geldenhuys W, Pistilli EE. Breast cancer‐associated skeletal muscle mitochondrial dysfunction and lipid accumulation is reversed by PPARG. Am J Physiol Cell Physiol. 2021;320(4):C577–C590.
    1. Larsen S, Nielsen J, Hansen CN, Nielsen LB, Wibrand F, Stride N, et al. Biomarkers of mitochondrial content in skeletal muscle of healthy young human subjects. J Physiol 2012;590:3349–3360.
    1. Gilliam LAA, Fisher‐Wellman KH, Lin C‐T, Maples JM, Cathey BL, Neufer PD. The anticancer agent doxorubicin disrupts mitochondrial energy metabolism and redox balance in skeletal muscle. Free Radic Biol Med 2013;65:988–996.
    1. Brown JL, Rosa‐Caldwell ME, Lee DE, Blackwell TA, Brown LA, Perry RA, et al. Mitochondrial degeneration precedes the development of muscle atrophy in progression of cancer cachexia in tumour‐bearing mice. J Cachexia Sarcopenia Muscle 2017;8:926–938.
    1. Pin F, Barreto R, Kitase Y, Mitra S, Erne CE, Novinger LJ, et al. Growth of ovarian cancer xenografts causes loss of muscle and bone mass: a new model for the study of cancer cachexia. J Cachexia Sarcopenia Muscle 2018;9:685–700.
    1. Marzetti E, Lorenzi M, Landi F, Picca A, Rosa F, Tanganelli F, et al. Altered mitochondrial quality control signaling in muscle of old gastric cancer patients with cachexia. Exp Gerontol 2017;87:92–99.
    1. Bergstrom J. Percutaneous needle biopsy of skeletal muscle in physiological and clinical research. Scand J Clin Lab Invest 1975;35:609–616.
    1. Pagano AF, Brioche T, Arc‐Chagnaud C, Demangel R, Chopard A, Py G. Short‐term disuse promotes fatty acid infiltration into skeletal muscle. J Cachexia Sarcopenia Muscle 2018;9:335–347.
    1. Arc‐Chagnaud C, Py G, Fovet T, Roumanille R, Demangel R, Pagano AF, et al. Evaluation of an antioxidant and anti‐inflammatory cocktail against human hypoactivity‐induced skeletal muscle deconditioning. Front Physiol 2020;11:71.
    1. Jacob KJ, Sonjak V, Spendiff S, Hepple RT, Chevalier S, Perez A, et al. Mitochondrial content, but not function, is altered with a multimodal resistance training protocol and adequate protein intake in leucine‐supplemented pre/frail women. Front Nutr 2021;7:619216.
    1. Huot JR, Pin F, Narasimhan A, Novinger LJ, Keith AS, Zimmers TA, et al. ACVR2B antagonism as a countermeasure to multi‐organ perturbations in metastatic colorectal cancer cachexia. J Cachexia Sarcopenia Muscle 2020;11:1779–1798.
    1. Ju W‐K, Lindsey JD, Angert M, Patel A, Weinreb RN. Glutamate receptor activation triggers OPA1 release and induces apoptotic cell death in ischemic rat retina. Mol Vis 2008;14:2629–2638.
    1. Geisler S, Holmström KM, Skujat D, Fiesel FC, Rothfuss OC, Kahle PJ, et al. PINK1/Parkin‐mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol 2010;12:119–131.
    1. Estaquier J, Vallette F, Vayssiere J‐L, Mignotte B. The mitochondrial pathways of apoptosis. Adv Exp Med Biol 2012;942:157–183.
    1. Shang H, Xia Z, Bai S, Zhang HE, Gu B, Wang R. Downhill running acutely elicits mitophagy in rat soleus muscle. Med Sci Sports Exerc 2019;51:1396–1403.
    1. Barreto R, Waning DL, Gao H, Liu Y, Zimmers TA, Bonetto A. Chemotherapy‐related cachexia is associated with mitochondrial depletion and the activation of ERK1/2 and p38 MAPKs. Oncotarget 2016;7:43442–43460.
    1. Lee H, Yoon Y. Mitochondrial fission and fusion. Biochem Soc Trans 2016;44:1725–1735.
    1. Pernas L, Scorrano L. Mito‐morphosis: mitochondrial fusion, fission, and cristae remodeling as key mediators of cellular function. Annu Rev Physiol 2016;78:505–531.
    1. Wang R, Mishra P, Garbis SD, Moradian A, Sweredoski MJ, Chan DC. Identification of new OPA1 cleavage site reveals that short isoforms regulate mitochondrial fusion. Mol Biol Cell 2021;32:157–168.
    1. Aversa Z, Pin F, Lucia S, Penna F, Verzaro R, Fazi M, et al. Autophagy is induced in the skeletal muscle of cachectic cancer patients. Sci Rep 2016;6:30340.
    1. Gouspillou G, Sgarioto N, Kapchinsky S, Purves‐Smith F, Norris B, Pion CH, et al. Increased sensitivity to mitochondrial permeability transition and myonuclear translocation of endonuclease G in atrophied muscle of physically active older humans. FASEB J 2014;28:1621–1633.
    1. Koleini N, Kardami E. Autophagy and mitophagy in the context of doxorubicin‐induced cardiotoxicity. Oncotarget 2017;8:46663–46680.
    1. Hoshino A, Mita Y, Okawa Y, Ariyoshi M, Iwai‐Kanai E, Ueyama T, et al. Cytosolic p53 inhibits Parkin‐mediated mitophagy and promotes mitochondrial dysfunction in the mouse heart. Nat Commun 2013;4:2308.
    1. Chacon‐Cabrera A, Mateu‐Jimenez M, Langohr K, Fermoselle C, García‐Arumí E, Andreu AL, et al. Role of PARP activity in lung cancer‐induced cachexia: effects on muscle oxidative stress, proteolysis, anabolic markers, and phenotype. J Cell Physiol 2017;232:3744–3761.
    1. Puig‐Vilanova E, Rodriguez DA, Lloreta J, Ausin P, Pascual‐Guardia S, Broquetas J, et al. Oxidative stress, redox signaling pathways, and autophagy in cachectic muscles of male patients with advanced COPD and lung cancer. Free Radic Biol Med 2015;79:91–108.
    1. Hulmi JJ, Nissinen TA, Räsänen M, Degerman J, Lautaoja JH, Hemanthakumar KA, et al. Prevention of chemotherapy‐induced cachexia by ACVR2B ligand blocking has different effects on heart and skeletal muscle. J Cachexia Sarcopenia Muscle 2018;9:417–432.
    1. Smuder AJ, Kavazis AN, Min K, Powers SK. Exercise protects against doxorubicin‐induced oxidative stress and proteolysis in skeletal muscle. J Appl Physiol Bethesda Md 1985 2011;110:935–942.
    1. Min K, Kwon O‐S, Smuder AJ, Wiggs MP, Sollanek KJ, Christou DD, et al. Increased mitochondrial emission of reactive oxygen species and calpain activation are required for doxorubicin‐induced cardiac and skeletal muscle myopathy. J Physiol 2015;593:2017–2036.
    1. von Haehling S, Morley JE, Coats AJS, Anker SD. Ethical guidelines for publishing in the Journal of Cachexia, Sarcopenia and Muscle: update 2021. J Cachexia Sarcopenia Muscle 2021;12:2259–2261.

Source: PubMed

3
Předplatit