The cystine/glutamate antiporter system xc- drives breast tumor cell glutamate release and cancer-induced bone pain

Lauren M Slosky, Neemah M BassiriRad, Ashley M Symons, Michelle Thompson, Timothy Doyle, Brittany L Forte, William D Staatz, Lynn Bui, William L Neumann, Patrick W Mantyh, Daniela Salvemini, Tally M Largent-Milnes, Todd W Vanderah, Lauren M Slosky, Neemah M BassiriRad, Ashley M Symons, Michelle Thompson, Timothy Doyle, Brittany L Forte, William D Staatz, Lynn Bui, William L Neumann, Patrick W Mantyh, Daniela Salvemini, Tally M Largent-Milnes, Todd W Vanderah

Abstract

Bone is one of the leading sites of metastasis for frequently diagnosed malignancies, including those arising in the breast, prostate and lung. Although these cancers develop unnoticed and are painless in their primary sites, bone metastases result in debilitating pain. Deeper investigation of this pain may reveal etiology and lead to early cancer detection. Cancer-induced bone pain (CIBP) is inadequately managed with current standard-of-care analgesics and dramatically diminishes patient quality of life. While CIBP etiology is multifaceted, elevated levels of glutamate, an excitatory neurotransmitter, in the bone-tumor microenvironment may drive maladaptive nociceptive signaling. Here, we establish a relationship between the reactive nitrogen species peroxynitrite, tumor-derived glutamate, and CIBP. In vitro and in a syngeneic in vivo model of breast CIBP, murine mammary adenocarcinoma cells significantly elevated glutamate via the cystine/glutamate antiporter system xc. The well-known system xc inhibitor sulfasalazine significantly reduced levels of glutamate and attenuated CIBP-associated flinching and guarding behaviors. Peroxynitrite, a highly reactive species produced in tumors, significantly increased system xc functional expression and tumor cell glutamate release. Scavenging peroxynitrite with the iron and mangano-based porphyrins, FeTMPyP and SRI10, significantly diminished tumor cell system xc functional expression, reduced femur glutamate levels and mitigated CIBP. In sum, we demonstrate how breast cancer bone metastases upregulate a cystine/glutamate co-transporter to elevate extracellular glutamate. Pharmacological manipulation of peroxynitrite or system xc attenuates CIBP, supporting a role for tumor-derived glutamate in CIBP and validating the targeting of system xc as a novel therapeutic strategy for the management of metastatic bone pain.

Conflict of interest statement

Sponsorships or competing interests that may be relevant to content are disclosed at the end of this article.

Figures

Figure 1.
Figure 1.
The murine mammary adenocarcinoma cell line 66.1 expresses the cystine/glutamate antiporter system xc−. (A) Representative images of 66.1 XCT and DAPI staining, as indicated. 66.1 cell glutamate release was quantified over a 24-hour period in untreated cells (B), and at 18 hours in those exposed to vehicle, sulfasalazine (SSZ) (C) or (S)-4-carboxyphenylglycine (CPG-4) (D). Asterisks denote values statistically different from time 0 or vehicle control group, *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 2.
Figure 2.
Blockade of system xc− with sulfasalazine (SSZ) attenuates cancer-induced bone pain behaviors and reduces femur glutamate. Animal femurs were injected with breast cancer cells (66.1) or cell-free media (sham) after baseline (presurgery) behavioral measurements. SSZ (30 mg/kg, i.p.) or vehicle (10 mL/kg, i.p.) was administered after behavioral measurements on postsurgery day 7 and continued for 3 days (q.d.). Cancer-induced spontaneous flinching (A) and guarding (B) were significantly reduced 60 minutes after SSZ treatment on day 10, as compared with vehicle-treated animals. Femur contents from inoculated femur (ipsilateral) (C) and contralateral femur (D) were collected on day 10 for glutamate analysis; *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 3.
Figure 3.
Peroxynitrite regulates system xc− functional expression. 66.1 cells were treated SIN-1 (50 nm–5 μM) or vehicle. Eighteen hours after treatment, cells were harvested and analyzed by Western blot analysis for XCT (A and B). SIN-1 stimulated 66.1 glutamate release in the absence (C) or presence (D) of sulfasalazine was quantified. 66.1 cells were exposed to SIN-1 in the presence of FeTMPyP or vehicle. Eighteen hours after treatment, cells were harvested and analyzed through Western blot analysis for expression of XCT (E and F). 66.1 glutamate release in the presence of SIN-1 and FeTMPyP, SRI10, or vehicle was quantified (G and H). Asterisks represent data points that are significantly different from those of vehicle control, unless otherwise indicated; *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 4.
Figure 4.
Repeated FeTMPyP treatment modulates spontaneous cancer-induced bone pain behaviors and system xc− functional expression in vivo. Animal femurs were injected with breast cancer cells (66.1) or cell-free media (sham) after baseline (presurgery) behavioral measurements. On day 7 after femoral inoculation, 66.1 animals demonstrated bone cancer–induced flinching (A) and guarding (B). FeTMPyP (10 mg/kg, i.p.) or vehicle (saline, 10 mL/kg, i.p.) was administered after behavioral measurements day 7 and continued to day 14 (q.d.). Spontaneous flinching and guarding were assessed before and after treatment on days 10 and 14 (A and B). Femur marrow from inoculated femur (ipsilateral) (C) and contralateral femur (D) was collected from tumor-bearing animals before or after the final treatment on day 14 for glutamate analysis. Day 14 ipsilateral marrow samples from tumor-bearing animals were analyzed for XCT expression using Western blot analysis (E–H); *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 5.
Figure 5.
Repeated FeTMPyP does not alter tumor burden. Animal femurs were inoculated with cell-free media (sham) or breast cancer cells (66.1). FeTMPyP (10 mg/kg, i.p., q.d.) or vehicle (saline, 10 mL/kg, i.p., q.d.) was administered on postsurgery days 7 to 14. On day 14, femurs were collected, paraffin-embedded, and stained with hematoxylin and eosin (H&E) to visualize normal marrow elements and cancer cells under bright field microscopy. Representative images (A) and quantification of cancer cell marrow occupancy (B) are shown.
Figure 6.
Figure 6.
Repeated FeTMPyP does not alter bone integrity. Animal femurs were inoculated with cell-free media (sham) or breast cancer cells (66.1). FeTMPyP (10 mg/kg, i.p., q.d.) or vehicle (saline, 10 mL/kg, i.p., q.d.) was administered on postsurgery days 7 to 14. On day 14, live radiographs were taken. Bone loss was rated by 3 blinded observers according to an established scale (A). Elevated bone loss scores were found in the tumor-bearing animals as compared with sham controls, but no treatment differences were evident (B); ***P < 0.001.
Figure 7.
Figure 7.
Repeated SRI10 treatment attenuates spontaneous cancer-induced bone pain behaviors and reduces femur glutamate. Animal femurs were injected with breast cancer cells (66.1) or cell-free media (sham) after baseline (presurgery) behavioral measurements. SRI10 (3 mg/kg, i.p.) or vehicle (10 mL/kg, i.p.) was administered after behavioral measurements day 7 and continued to day 14 (q.d.). Spontaneous flinching (A) and guarding (B) were assessed before and after treatment on days 10 and 14. Femur marrow from inoculated femur (ipsilateral) (C) and contralateral femur (D) was collected from tumor-bearing animals before or after the final treatment on day 14 for glutamate analysis; *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 8.
Figure 8.
System xc− can be targeted directly or indirectly to reduce glutamate in the bone-tumor microenvironment and assuage cancer-induced bone pain (CIBP). System xc− can be targeted pharmacologically through direct blockage with sulfasalazine or through elimination of peroxynitrite with the decomposition catalysts FeTMPyP and SRI10. Both therapeutic strategies reduced glutamate in the bone–tumor microenvironment and CIBP-related behaviors.

References

    1. American Cancer Society. Global Cancer Facts & Figures 2nd Edition Atlanta: American Cancer Society; 2011.
    1. Barclay JS, Owens JE, Blackhall LJ. Screening for substance abuse risk in cancer patients using the Opioid Risk Tool and urine drug screen. Support Care Cancer 2014;22:1883–8.
    1. Bloom AP, Jimenez-Andrade JM, Taylor RN, Castaneda-Corral G, Kaczmarska MJ, Freeman KT, Coughlin KA, Ghilardi JR, Kuskowski MA, Mantyh PW. Breast cancer-induced bone remodeling, skeletal pain, and sprouting of sensory nerve fibers. J Pain 2011;12:698–711.
    1. Buckley BJ, Whorton AR. Adaptive responses to peroxynitrite: increased glutathione levels and cystine uptake in vascular cells. Am J Physiol Cell Physiol 2000;279:C1168–76.
    1. Coleman RE. Metastatic bone disease: clinical features, pathophysiology and treatment strategies. Cancer Treat Rev 2001;27:165–76.
    1. Coleman RE. Clinical features of metastatic bone disease and risk of skeletal morbidity. Clin Cancer Res 2006;12(20 pt 2):6243s-9s.
    1. Coleman RE, Rubens RD. The clinical course of bone metastases from breast cancer. Br J Cancer 1987;55:61–6.
    1. Currie GL, Delaney A, Bennett MI, Dickenson AH, Egan KJ, Vesterinen HM, Sena ES, Macleod MR, Colvin LA, Fallon MT. Animal models of bone cancer pain: systematic review and meta-analyses. PAIN 2013;154:917–26.
    1. Delaney A, Fleetwood-Walker SM, Colvin LA, Fallon M. Translational medicine: cancer pain mechanisms and management. Br J Anaesth 2008;101:87–94.
    1. Denicola A, Souza JM, Radi R. Diffusion of peroxynitrite across erythrocyte membranes. Proc Natl Acad Sci U S A 1998;95:3566–71.
    1. Dexter DL, Kowalski HM, Blazar BA, Fligiel Z, Vogel R, Heppner GH. Heterogeneity of tumor cells from a single mouse mammary tumor. Cancer Res 1978;38:3174–81.
    1. Fisch MJ, Lee JW, Weiss M, Wagner LI, Chang VT, Cella D, Manola JB, Minasian LM, McCaskill-Stevens W, Mendoza TR, Cleeland CS. Prospective, observational study of pain and analgesic prescribing in medical oncology outpatients with breast, colorectal, lung, or prostate cancer. J Clin Oncol 2012;30:1980–8.
    1. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, O'Dwyer PJ, Lee RJ, Grippo JF, Nolop K, Chapman PB. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med 2010;363:809–19.
    1. Ischiropoulos H. Biological tyrosine nitration: a pathophysiological function of nitric oxide and reactive oxygen species. Arch Biochem Biophys 1998;356:1–11.
    1. Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, Massie A, Smolders I, Methner A, Pergande M, Smith SB, Ganapathy V, Maher P. The cystine/glutamate antiporter system x(c)(-) in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal 2013;18:522–55.
    1. Lozano-Ondoua AN, Hanlon KE, Symons-Liguori AM, Largent-Milnes TM, Havelin JJ, Ferland HL, III, Chandramouli A, Owusu-Ankomah M, Nikolich-Zugich T, Bloom AP, Jimenez-Andrade JM, King T, Porreca F, Nelson MA, Mantyh PW, Vanderah TW. Disease modification of breast cancer-induced bone remodeling by cannabinoid 2 receptor agonists. J Bone Miner Res 2013;28:92–107.
    1. Lozano-Ondoua AN, Symons-Liguori AM, Vanderah TW. Cancer-induced bone pain: mechanisms and models. Neurosci Lett 2013;(557 pt A):52–9.
    1. Lozano-Ondoua AN, Wright C, Vardanyan A, King T, Largent-Milnes TM, Nelson M, Jimenez-Andrade JM, Mantyh PW, Vanderah TW. A cannabinoid 2 receptor agonist attenuates bone cancer-induced pain and bone loss. Life Sci 2010;86:646–53.
    1. Lu T, Ramakrishnan R, Altiok S, Youn JI, Cheng P, Celis E, Pisarev V, Sherman S, Sporn MB, Gabrilovich D. Tumor-infiltrating myeloid cells induce tumor cell resistance to cytotoxic T cells in mice. J Clin Invest 2011;121:4015–29.
    1. Mach DB, Rogers SD, Sabino MC, Luger NM, Schwei MJ, Pomonis JD, Keyser CP, Clohisy DR, Adams DJ, O'Leary P, Mantyh PW. Origins of skeletal pain: sensory and sympathetic innervation of the mouse femur. Neuroscience 2002;113:155–66.
    1. Mercadante S. Malignant bone pain: pathophysiology and treatment. PAIN 1997;69:1–18.
    1. Nagae M, Hiraga T, Yoneda T. Acidic microenvironment created by osteoclasts causes bone pain associated with tumor colonization. J Bone Miner Metab 2007;25:99–104.
    1. Nakamura Y, Yasuoka H, Tsujimoto M, Yoshidome K, Nakahara M, Nakao K, Nakamura M, Kakudo K. Nitric oxide in breast cancer: induction of vascular endothelial growth factor-C and correlation with metastasis and poor prognosis. Clin Cancer Res 2006;12:1201–7.
    1. O'Connell JX, Nanthakumar SS, Nielsen GP, Rosenberg AE. Osteoid osteoma: the uniquely innervated bone tumor. Mod Pathol 1998;11:175–80.
    1. Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev 2007;87:315–424.
    1. Rausaria S, Ghaffari MM, Kamadulski A, Rodgers K, Bryant L, Chen Z, Doyle T, Shaw MJ, Salvemini D, Neumann WL. Retooling manganese(III) porphyrin-based peroxynitrite decomposition catalysts for selectivity and oral activity: a potential new strategy for treating chronic pain. J Med Chem 2011;54:8658–69.
    1. Sabino MA, Mantyh PW. Pathophysiology of bone cancer pain. J Support Oncol 2005;3:15–24.
    1. Salvemini D, Little JW, Doyle T, Neumann WL. Roles of reactive oxygen and nitrogen species in pain. Free Radic Biol Med 2011;51:951–66.
    1. Salvemini D, Wang ZQ, Stern MK, Currie MG, Misko TP. Peroxynitrite decomposition catalysts: therapeutics for peroxynitrite-mediated pathology. Proc Natl Acad Sci U S A 1998;95:2659–63.
    1. Sasaki H, Sato H, Kuriyama-Matsumura K, Sato K, Maebara K, Wang H, Tamba M, Itoh K, Yamamoto M, Bannai S. Electrophile response element-mediated induction of the cystine/glutamate exchange transporter gene expression. J Biol Chem 2002;277:44765–71.
    1. Seidlitz EP, Sharma MK, Saikali Z, Ghert M, Singh G. Cancer cell lines release glutamate into the extracellular environment. Clin Exp Metastasis 2009;26:781–7.
    1. Sharma MK, Seidlitz EP, Singh G. Cancer cells release glutamate via the cystine/glutamate antiporter. Biochem Biophys Res Commun 2010;391:91–5.
    1. Shukla K, Thomas AG, Ferraris DV, Hin N, Sattler R, Alt J, Rojas C, Slusher BS, Tsukamoto T. Inhibition of xc(-) transporter-mediated cystine uptake by sulfasalazine analogs. Bioorg Med Chem Lett 2011;21:6184–7.
    1. Slosky LM, Largent-Milnes TM, Vanderah TW. Use of animal models in understanding cancer-induced bone pain. Cancer Growth Metastasis 2015;8(suppl 1):47–62.
    1. Slosky LM, Vanderah TW. Therapeutic potential of peroxynitrite decomposition catalysts: a patent review. Expert Opin Ther Pat 2015;25:443–66.
    1. Tas F, Hansel H, Belce A, Ilvan S, Argon A, Camlica H, Topuz E. Oxidative stress in breast cancer. Med Oncology 2005;22:11–15.
    1. Ungard RG, Seidlitz EP, Singh G. Oxidative stress and cancer pain. Can J Physiol Pharmacol 2013;91:31–7.
    1. Ungard RG, Seidlitz EP, Singh G. Inhibition of breast cancer-cell glutamate release with sulfasalazine limits cancer-induced bone pain. PAIN 2014;155:28–36.
    1. Vakkala M, Kahlos K, Lakari E, Paakko P, Kinnula V, Soini Y. Inducible nitric oxide synthase expression, apoptosis, and angiogenesis in in situ and invasive breast carcinomas. Clin Cancer Research 2000;6:2408–16.
    1. van den Beuken-van Everdingen MH, de Rijke JM, Kessels AG, Schouten HC, van Kleef M, Patijn J. Prevalence of pain in patients with cancer: a systematic review of the past 40 years. Ann Oncol 2007;18:1437–49.
    1. Virag L, Szabo E, Gergely P, Szabo C. Peroxynitrite-induced cytotoxicity: mechanism and opportunities for intervention. Toxicol Lett 2003;140:113–24.
    1. Walser TC, Rifat S, Ma X, Kundu N, Ward C, Goloubeva O, Johnson MG, Medina JC, Collins TL, Fulton AM. Antagonism of CXCR3 inhibits lung metastasis in a murine model of metastatic breast cancer. Cancer Res 2006;66:7701–7.
    1. Watkins LR, Wiertelak EP, Goehler LE, Smith KP, Martin D, Maier SF. Characterization of cytokine-induced hyperalgesia. Brain Res 1994;654:15–26.
    1. WHO. Cancer pain relief. Geneva, World Health Organziation, 1998.

Source: PubMed

3
Subscribe