Doxorubicin acts through tumor necrosis factor receptor subtype 1 to cause dysfunction of murine skeletal muscle.

Cancer patients receiving doxorubicin chemotherapy experience both muscle weakness and fatigue. One postulated mediator of the muscle dysfunction is an increase in tumor necrosis factor-alpha (TNF), a proinflammatory cytokine that mediates limb muscle contractile dysfunction through the TNF receptor subtype 1 (TNFR1). Our main hypothesis was that systemic doxorubicin administration would cause muscle weakness and fatigue. Systemic doxorubicin administration (20 mg/kg) depressed maximal force of the extensor digitorum longus (EDL; P < 0.01), accelerated EDL fatigue (P < 0.01), and elevated serum TNF levels (P < 0.05) 72 h postinjection. Genetic TNFR1 deficiency prevented the fall in specific force caused by systemic doxorubicin, without protecting against fatigue (P < 0.01). These results demonstrate that clinical doxorubicin concentrations disrupt limb muscle function in a TNFR1-dependent manner.

[1]  J. Argilés,et al.  Direct effects of doxorubicin on skeletal muscle contribute to fatigue , 2009, British Journal of Cancer.

[2]  G. Schuler,et al.  Induction of MuRF1 is essential for TNF-alpha-induced loss of muscle function in mice. , 2008, Journal of molecular biology.

[3]  K. Campbell,et al.  TNF-alpha acts via TNFR1 and muscle-derived oxidants to depress myofibrillar force in murine skeletal muscle. , 2008, Journal of applied physiology.

[4]  W. Dröge,et al.  Cysteine-rich protein reverses weight loss in lung cancer patients receiving chemotherapy or radiotherapy. , 2008, Antioxidants & redox signaling.

[5]  N. Mariappan,et al.  TNF-alpha-induced mitochondrial oxidative stress and cardiac dysfunction: restoration by superoxide dismutase mimetic Tempol. , 2007, American journal of physiology. Heart and circulatory physiology.

[6]  D. Graham,et al.  Effects of buthionine sulfoximine treatment on diaphragm contractility and SR Ca2+ pump function in rats. , 2007, Journal of applied physiology.

[7]  A. Nudelman,et al.  Anticancer prodrugs of butyric acid and formaldehyde protect against doxorubicin-induced cardiotoxicity , 2007, British Journal of Cancer.

[8]  N. Kearney,et al.  Patterns of fatigue during a course of chemotherapy: results from a multi-centre study. , 2007, European journal of oncology nursing : the official journal of European Oncology Nursing Society.

[9]  D. Butterfield,et al.  Adriamycin‐mediated nitration of manganese superoxide dismutase in the central nervous system: insight into the mechanism of chemobrain , 2007, Journal of neurochemistry.

[10]  D. Butterfield,et al.  Adriamycin-induced, TNF-α-mediated central nervous system toxicity , 2006, Neurobiology of Disease.

[11]  Francisco H Andrade,et al.  Contractile dysfunction and altered metabolic profile of the aging rat thyroarytenoid muscle. , 2006, Journal of applied physiology.

[12]  D. S. St. Clair,et al.  Tumor necrosis factor receptor deficiency exacerbated Adriamycin-induced cardiomyocytes apoptosis: an insight into the Fas connection , 2006, Molecular Cancer Therapeutics.

[13]  I. Tomášková,et al.  Cardiopulmonary exercise testing in the evaluation of functional capacity after treatment of lymphomas in adults , 2006, Leukemia & lymphoma.

[14]  Jian Huang,et al.  Myosin light chain kinase and myosin phosphorylation effect frequency-dependent potentiation of skeletal muscle contraction. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[15]  P. Jacobsen,et al.  Course of fatigue in women receiving chemotherapy and/or radiotherapy for early stage breast cancer. , 2004, Journal of pain and symptom management.

[16]  A. Bakkaloğlu,et al.  Effects of pentoxifylline in Adriamycin-induced renal disease in rats , 2004, Pediatric Nephrology.

[17]  A. Dinda,et al.  Protection against acute adriamycin-induced cardiotoxicity by garlic: Role of endogenous antioxidants and inhibition of TNF-α expression , 2003, BMC pharmacology.

[18]  G. Schuler,et al.  Anti-inflammatory effects of exercise training in the skeletal muscle of patients with chronic heart failure. , 2003, Journal of the American College of Cardiology.

[19]  V. Shirinsky,et al.  Alterations in Myocardial Cytoskeletal and Regulatory Protein Expression Following a Single Doxorubicin Injection , 2003, Journal of cardiovascular pharmacology.

[20]  G. Lamb,et al.  Effects of oxidation and reduction on contractile function in skeletal muscle fibres of the rat , 2003, The Journal of physiology.

[21]  H. Westerblad,et al.  Respiratory and limb muscle weakness induced by tumor necrosis factor-alpha: involvement of muscle myofilaments. , 2002, American journal of respiratory and critical care medicine.

[22]  Patricia A. Ganz,et al.  Fatigue and Proinflammatory Cytokine Activity in Breast Cancer Survivors , 2002, Psychosomatic medicine.

[23]  B. Decallonne,et al.  An overview of real-time quantitative PCR: applications to quantify cytokine gene expression. , 2001, Methods.

[24]  Thomas D. Schmittgen,et al.  Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. , 2001, Methods.

[25]  Yi-Ping Li,et al.  Tumor necrosis factor-α and muscle wasting: a cellular perspective , 2001, Respiratory research.

[26]  H. Westerblad,et al.  Contractile response of skeletal muscle to low peroxide concentrations: myofibrillar calcium sensitivity as a likely target for redox-modulation. , 2001, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[27]  F. Clubb,et al.  Cardiac-Specific Overexpression of Tumor Necrosis Factor-&agr; Causes Oxidative Stress and Contractile Dysfunction in Mouse Diaphragm , 2000, Circulation.

[28]  C. Schöneich,et al.  Peroxynitrite modification of protein thiols: oxidation, nitrosylation, and S-glutathiolation of functionally important cysteine residue(s) in the sarcoplasmic reticulum Ca-ATPase. , 1999, Biochemistry.

[29]  P. Stone,et al.  Fatigue in advanced cancer: a prospective controlled cross-sectional study , 1999, British Journal of Cancer.

[30]  P. Iaizzo,et al.  Doxorubicin chemomyectomy as a treatment for cervical dystonia: Histological assessment after direct injection into the sternocleidomastoid muscle , 1998, Muscle & nerve.

[31]  N. Agell,et al.  Role of TNF receptor 1 in protein turnover during cancer cachexia using gene knockout mice , 1998, Molecular and Cellular Endocrinology.

[32]  D. Allen,et al.  Effect of hydrogen peroxide and dithiothreitol on contractile function of single skeletal muscle fibres from the mouse , 1998, The Journal of physiology.

[33]  B. Coiffier,et al.  ELEVATED CIRCULATING LEVELS OF TNFα AND ITS p55 SOLUBLE RECEPTOR ARE ASSOCIATED WITH AN ADVERSE PROGNOSIS IN LYMPHOMA PATIENTS , 1996, British journal of haematology.

[34]  C. Garcı́a-Martı́nez,et al.  Acute treatment with tumour necrosis factor-α induces changes in protein metabolism in rat skeletal muscle , 1993, Molecular and Cellular Biochemistry.

[35]  S. Piscitelli,et al.  Pharmacokinetics and pharmacodynamics of doxorubicin in patients with small cell lung cancer , 1993, Clinical pharmacology and therapeutics.

[36]  E. Voest,et al.  Doxorubicin interacts directly with skinned single skeletal muscle fibres. , 1992, European journal of pharmacology.

[37]  M. Siimes,et al.  Skeletal muscle wasting and protein–energy malnutrition in children with newly diagnosed acute leukemia , 1990, Cancer.

[38]  Aquilur Rahman,et al.  A phase I/II study of intraperitoneally administered doxorubicin entrapped in cardiolipin liposomes in patients with ovarian cancer. , 1989, American journal of obstetrics and gynecology.

[39]  H. Westerblad,et al.  The temperature dependence of isometric contractions of single, intact fibres dissected from a mouse foot muscle. , 1987, The Journal of physiology.

[40]  F. Zorzato,et al.  Doxorubicin induces calcium release from terminal cisternae of skeletal muscle. A study on isolated sarcoplasmic reticulum and chemically skinned fibers. , 1985, The Journal of biological chemistry.

[41]  R. Tsien,et al.  A new generation of Ca2+ indicators with greatly improved fluorescence properties. , 1985, The Journal of biological chemistry.

[42]  J. Doroshow,et al.  Ultrastructural features of Adriamycin-induced skeletal and cardiac muscle toxicity. , 1985, The American journal of pathology.

[43]  E J Freireich,et al.  Quantitative comparison of toxicity of anticancer agents in mouse, rat, hamster, dog, monkey, and man. , 1966, Cancer chemotherapy reports.

[44]  L. Goodman,et al.  THE PHARMACOLOGICAL BASIS OF THERAPEUTICS , 1966 .

[45]  D. Allen,et al.  Skeletal muscle fatigue: cellular mechanisms. , 2008, Physiological reviews.

[46]  D. Butterfield,et al.  Adriamycin-induced, TNF-alpha-mediated central nervous system toxicity. , 2006, Neurobiology of disease.

[47]  P. Johansen Doxorubicin pharmacokinetics after intravenous and intraperitoneal administration in the nude mouse , 2004, Cancer Chemotherapy and Pharmacology.

[48]  Y. J. Kang,et al.  Inhibition of doxorubicin chronic toxicity in catalase-overexpressing transgenic mouse hearts. , 2002, Chemical research in toxicology.

[49]  C. Angelini,et al.  Neuromuscular damage after hyperthermic isolated limb perfusion in patients with melanoma or sarcoma treated with chemotherapeutic agents , 2000, Cancer Chemotherapy and Pharmacology.

[50]  A. Scialli Goodman and Gilman's the pharmacologic basis of therapeutics , 1991 .

[51]  S. Ellenberg,et al.  A randomized study of oral nutritional support versus ad lib nutritional intake during chemotherapy for advanced colorectal and non-small-cell lung cancer. , 1987, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[52]  R. Close Dynamic properties of mammalian skeletal muscles. , 1972, Physiological reviews.