Oxaliplatin neurotoxicity involves peroxisome alterations. PPARγ agonism as preventive pharmacological approach

Matteo Zanardelli, Laura Micheli, Lorenzo Cinci, Paola Failli, Carla Ghelardini, Lorenzo Di Cesare Mannelli, Matteo Zanardelli, Laura Micheli, Lorenzo Cinci, Paola Failli, Carla Ghelardini, Lorenzo Di Cesare Mannelli

Abstract

The development of neuropathic syndromes is an important, dose limiting side effect of anticancer agents like platinum derivates, taxanes and vinca alkaloids. The causes of neurotoxicity are still unclear but the impairment of the oxidative equilibrium is strictly related to pain. Two intracellular organelles, mitochondria and peroxisomes cooperate to the maintaining of the redox cellular state. Whereas a relationship between chemotherapy-dependent mitochondrial alteration and neuropathy has been established, the role of peroxisome is poor explored. In order to study the mechanisms of oxaliplatin-induced neurotoxicity, peroxisomal involvement was evaluated in vitro and in vivo. In primary rat astrocyte cell culture, oxaliplatin (10 µM for 48 h or 1 µM for 5 days) increased the number of peroxisomes, nevertheless expression and functionality of catalase, the most important antioxidant defense enzyme in mammalian peroxisomes, were significantly reduced. Five day incubation with the selective Peroxisome Proliferator Activated Receptor-γ (PPAR-γ) antagonist G3335 (30 µM) induced a similar peroxisomal impairment suggesting a relationship between PPARγ signaling and oxaliplatin neurotoxicity. The PPARγ agonist rosiglitazone (10 µM) reduced the harmful effects induced both by G3335 and oxaliplatin. In vivo, in a rat model of oxaliplatin induced neuropathy, a repeated treatment with rosiglitazone (3 and 10 mg kg(-1) per os) significantly reduced neuropathic pain evoked by noxious (Paw pressure test) and non-noxious (Cold plate test) stimuli. The behavioral effect paralleled with the prevention of catalase impairment induced by oxaliplatin in dorsal root ganglia. In the spinal cord, catalase protection was showed by the lower rosiglitazone dosage without effect on the astrocyte density increase induced by oxaliplatin. Rosiglitazone did not alter the oxaliplatin-induced mortality of the human colon cancer cell line HT-29. These results highlight the role of peroxisomes in oxaliplatin-dependent nervous damage and suggest PPARγ stimulation as a candidate to counteract oxaliplatin neurotoxicity.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Catalase immunostaining in primary astrocytes.
Figure 1. Catalase immunostaining in primary astrocytes.
Cells (5·104 cells/well) were incubated for 5 days with 10 µM rosiglitazone (B), 1 µM oxaliplatin (C), 1 µM oxaliplatin+10 µM rosiglitazone (D), 30 µM G3335 (E), negative control (F) in comparison to control condition (A). Representative images are shown in the left panel. Scale bar 50 µm. The measurements of the number of peroxisomes/µm2 and the catalase optical density per number of peroxisomes are shown in the upper and lower graphs, respectively. *P<0.01 vs control; ∧P<0.01 vs 1 µM oxaliplatin.
Figure 2. Expression and activity of catalase…
Figure 2. Expression and activity of catalase in astrocyte cell culture.
Astrocytes (5·105 cells/well) were treated with the PPARγ antagonist G3335 (30 µM) or with oxaliplatin (1 µM) in the absence or in the presence of the PPARγ agonist rosiglitazone (10 µM). Expression and activity were measured after 48 h- (A and B, respectively) or 5 day-treatment (C and D, respectively). GAPDH normalization was performed for each sample. Values are expressed as the mean ± S.E.M. percent of control of three experiments. Control condition was arbitrarily set as 100%. *P<0.05 vs control; ∧P<0.05 vs 1 µM oxaliplatin.
Figure 3. Pain threshold measurements.
Figure 3. Pain threshold measurements.
A) Noxious stimulus, Paw-pressure test. Rats were daily intraperitoneally treated with 2.4 mg kg−1 oxaliplatin (dissolved in 5% glucose). Rosiglitazone (3 and 10 mg kg−1, suspended in CMC) was per os daily administered starting from the first day of oxaliplatin administration. B) Non-noxious stimulus, Cold plate test. The response to a thermal stimulus was evaluated by cold plate test measuring the latency (seconds) to pain-related behaviors (lifting or licking of the paw). Control animals were treated with vehicles. Behavioral measures were performed on day 7, 14 and 21, 24 h after the last treatment. Each value represents the mean of 10 rats per group, performed in 2 different experimental set. *P<0.01 vs vehicle + vehicle (control); ∧P<0.01 vs oxaliplatin + vehicle.
Figure 4. Motor coordination in oxaliplatin-treated rats.
Figure 4. Motor coordination in oxaliplatin-treated rats.
The integrity of the animals’ motor coordination was assessed using a rota-rod apparatus. Rats were placed on a rotating rod (10 rpm) for a maximum of 10 minutes (600 seconds). The number of falls (A) and the time spent in the balance (B) during 10 minutes were counted. Treatments (oxaliplatin 2.4 mg kg−1 i.p. and rosiglitazone 3 and 10 mg kg−1 p.o.) were performed daily. Motor coordination was evaluated on day 21, 24 h after the last treatment. Each value represents the mean of 10 rats per group, performed in 2 different experimental set. *P<0.01 vs vehicle + vehicle (control); ∧P<0.05 vs oxaliplatin + vehicle.
Figure 5. Expression and activity of catalase…
Figure 5. Expression and activity of catalase in the nervous tissue of oxaliplatin-treated animals.
On day 21, dorsal root ganglia (DRG) and spinal cord were analyzed to measure both expression and activity of catalase. Densitometric analysis and representative Western blot of catalase expression in DRG (A) and spinal cord (C) are shown. GAPDH normalization was performed for each sample. Catalase enzymatic activity measurements in DRG (B) and spinal cord (D). Values are expressed as the mean ± S.E.M. percent of control of 10 rats per group, performed in 2 different experimental set. Each value represents the mean of *P

Figure 6. Levels of carbonylated proteins in…

Figure 6. Levels of carbonylated proteins in the spinal cord of oxaliplatin-treated rats.

At 21…

Figure 6. Levels of carbonylated proteins in the spinal cord of oxaliplatin-treated rats.
At 21th day, the lumbar tract of the spinal cord was explanted and analyzed to measure protein oxidation. Densitometric analysis (top panel) and representative Western blot (lower panel) are shown. β-actin normalization was performed for each sample. Values are expressed as the mean ± S.E.M. percent of control of 10 rats per group, performed in 2 different experimental set. Each value represents the mean of *P<0.05 vs vehicle + vehicle; ∧P<0.05 vs oxaliplatin + vehicle.

Figure 7. Glial profile in spinal cord…

Figure 7. Glial profile in spinal cord scored with GFAP-positive cells in the dorsal horn…

Figure 7. Glial profile in spinal cord scored with GFAP-positive cells in the dorsal horn of the lumbar tract.
Transverse sections of spinal cord imaged with 20X objective of A) vehicle + vehicle, B) oxaliplatin + vehicle, C) and D) oxaliplatin + rosiglitazone 3 and 10 mg kg−1, respectively. Scale bar 50 µm. In the lower panel quantitative analysis of cellular density is shown. Each value represents the mean ± S.E.M. of 10 rats per group, performed in 2 different experimental sets. *P<0.05 vs vehicle + vehicle; ∧P<0.05 vs oxaliplatin + vehicle.
All figures (7)
Figure 6. Levels of carbonylated proteins in…
Figure 6. Levels of carbonylated proteins in the spinal cord of oxaliplatin-treated rats.
At 21th day, the lumbar tract of the spinal cord was explanted and analyzed to measure protein oxidation. Densitometric analysis (top panel) and representative Western blot (lower panel) are shown. β-actin normalization was performed for each sample. Values are expressed as the mean ± S.E.M. percent of control of 10 rats per group, performed in 2 different experimental set. Each value represents the mean of *P<0.05 vs vehicle + vehicle; ∧P<0.05 vs oxaliplatin + vehicle.
Figure 7. Glial profile in spinal cord…
Figure 7. Glial profile in spinal cord scored with GFAP-positive cells in the dorsal horn of the lumbar tract.
Transverse sections of spinal cord imaged with 20X objective of A) vehicle + vehicle, B) oxaliplatin + vehicle, C) and D) oxaliplatin + rosiglitazone 3 and 10 mg kg−1, respectively. Scale bar 50 µm. In the lower panel quantitative analysis of cellular density is shown. Each value represents the mean ± S.E.M. of 10 rats per group, performed in 2 different experimental sets. *P<0.05 vs vehicle + vehicle; ∧P<0.05 vs oxaliplatin + vehicle.

References

    1. André T, Boni C, Mounedji-Boudiaf L, Navarro M, Tabernero J, et al. (2004) Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N Engl J Med 350: 2343–2351.
    1. Kannarkat G, Lasher EE, Schiff D (2007) Neurologic complications of chemotherapy agents. Curr Opin Neurol 20: 719–725.
    1. Wolf S, Barton D, Kottschade L, Grothey A, Loprinzi C (2008) Chemotherapy induced peripheral neuropathy: prevention and treatment strategies. Eur J Cancer 44: 1507–1515.
    1. De Gramont A, Figer A, Seymour M, Homerin M, Hmissi A, et al. (2000) Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol 18: 2938–2947.
    1. Souglakos J, Mavroudis D, Kakolyris S, Kourousis CH, Vardakis N, et al. (2002) Triplet combination with irinotecan plus oxaliplatin plus continuous-infusion fluorouracil and leucovorin as first-line treatment in metastatic colorectal cancer: a multicenter phase II trial. J Clin Oncol 20: 2651–2657.
    1. Cavaletti G, Marmiroli P (2010) Chemotherapy-induced peripheral neurotoxicity. Nat Rev Neurol 6: 657–666.
    1. Di Cesare Mannelli L, Zanardelli M, Failli P, Ghelardini C (2012) Oxaliplatin-induced neuropathy: oxidative stress as pathological mechanism. Protective effect of silibinin. J Pain 13: 276–284.
    1. Di Cesare Mannelli L, Zanardelli M, Failli P, Ghelardini C (2013a) Oxaliplatin-induced oxidative stress in nervous system-derived cellular models: Could it correlate with in vivo neuropathy?. Free Radic Biol Med 61C: 143–150.
    1. Di Cesare Mannelli L, Pacini A, Matera C, Zanardelli M, Mello T, et al. (2013b) Involvement of α7 nAChR subtype in rat oxaliplatin-induced neuropathy: Effects of selective activation. Neuropharmacology 79C: 37–48.
    1. Zheng H, Xiao WH, Bennett GJ (2011) Functional deficits in peripheral nerve mitochondria in rats with paclitaxel- and oxaliplatin-evoked painful peripheral neuropathy. Exp Neurol 232: 154–161.
    1. De Duve C, Baudhuin P (1966) Peroxisomes (microbodies and related particles). Physiol Rev 46: 323–357.
    1. Elliott BM, Dodd NJ, Elcombe CR (1986) Increased hydroxyl radical production in liver peroxisomal fractions from rats treated with peroxisome proliferators. Carcinogenesis 7: 795–799.
    1. Zwacka RM, Reuter A, Pfaff E, Moll J, Gorgas K, et al. (1994) The glomerulosclerosis gene Mpv17 encodes a peroxisomal protein producing reactive oxygen species EMBO J. 13: 5129–5134.
    1. Stolz DB, Zamora R, Vodovotz Y, Loughran PA, Billiar TR, et al. (2002) Peroxisomal localization of inducible nitric oxide synthase in hepatocytes. Hepatology 36: 81–93.
    1. Schrader M, Fahimi HD (2006) Peroxisomes and oxidative stress. Biochim Biophys Acta 1763: 1755–1766.
    1. Powes JM, Mosers HW (1998) Peroxisomal disorders: genotype, phenotype, major neuropathologic lesions, and pathogenesis. Brain Pathol 8: 101–120.
    1. Bottelbergs A, Verheijden S, Van Veldhoven PP, Just W, Devos R, et al. (2012) Peroxisome deficiency but not the defect in ether lipid synthesis causes activation of the innate immune system and axonal loss in the central nervous system. J Neuroinflammation 9: 61.
    1. Trompier D, Vejux A, Zarrouk A, Gondcaille C, Geillon F, et al. (2014) Brain peroxisomes. Biochimie 98: 102–110.
    1. D’amico A, Bertini E (2013) Metabolic neuropathies and myopathies. Handb Clin Neurol 113: 1437–1455.
    1. Cimini A, Moreno S, D’Amelio M, Cristiano L, D’Angelo B, et al. (2009) Early biochemical and morphological modifications in the brain of a transgenic mouse model of Alzheimer’s disease: a role for peroxisomes. J Alzheimers Dis 18: 935–952.
    1. Kou J, Kovacs GG, Höftberger R, Kulik W, Brodde A, et al. (2011) Peroxisomal alterations in Alzheimer’s disease. Acta Neuropathol 122: 271–283.
    1. Fransen M, Nordgren M, Wang B, Apanasets O (2012) Role of peroxisomes in ROS/RNS-metabolism: implications for human disease. Biochim Biophys Acta. 1822: 1363–1373.
    1. Issemann I, Green S (1990) Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 347: 645–650.
    1. Crosby MB, Zhang J, Nowling TM, Svenson JL, Nicol CJ, et al. (2006) Inflammatory modulation of PPAR gamma expression and activity. Clin Immunol 118: 276–283.
    1. Girnun GD, Domann FE, Moore SA, Robbins ME (2002) Identification of a functional peroxisome proliferator-activated receptor response element in the rat catalase promoter. Mol Endocrinol 16: 2793–2801.
    1. Putnam CD, Arvai AS, Bourne Y, Tailner JA (2000) Active and inhibited catalase structures: ligand and NAPH binding and catalytic mechanism J Mol Biol. 296: 295–309.
    1. Powers SK, Jackson MJ (2008) Exercise-induced oxidative stress: cellular mechanisms and impact on muscle force production. Physiol. Rev 88: 1243–1276.
    1. Diab A, Deng C, Smith JD, Hussain RZ, Phanavanh B, et al. (2002) Peroxisome proliferator-activated receptor-gamma agonist 15-deoxy-Delta(12,14)-prostaglandin J(2) ameliorates experimental autoimmune encephalomyelitis. J Immunol 168: 2508–2515.
    1. Feinstein DL, Galea E, Gavrilyuk V, Brosnan CF, Whitacre CC, et al. (2002) Peroxisome proliferator-activated receptor-gamma agonists prevent experimental autoimmune encephalomyelitis. Ann Neurol 51: 694–702.
    1. Raikwar HP, Muthian G, Rajasingh J, Johnson CN, Bright JJ (2006) PPARgamma antagonists reverse the inhibition of neural antigen-specific Th1 response and experimental allergic encephalomyelitis by Ciglitazone and 15-deoxy-Delta12,14-prostaglandin J2. J Neuroimmunol 178: 76–86.
    1. Heneka MT, Landreth GE, Hüll M (2007) Drug insight: effects mediated by peroxisome proliferator-activated receptor-gamma in CNS disorders. Nat Cli Pract Neurol 3: 496–504.
    1. Bernardo A, Minghetti L (2008) Regulation of Glial Cell Functions by PPAR gamma. Natural and Synthetic Agonists. PPAR Res 2008: 864140.
    1. Landreth G, Jiang Q, Mandrekar S, Heneka M (2008) PPARgamma agonists as therapeutics for the treatment of Alzheimer’s disease. Neurotherapeutics 5: 481–489.
    1. Lee YJ, Han SB, Nam SY, Oh KW, Hong JT (2010) Inflammation and Alzheimer’s disease. Arch Pharm Res 33: 1539–1556.
    1. Swanson CR, Bondarenko VJV, Brunner K, Simmons HA, Ziegler TE, et al. (2011) The PPAR-γ agonist pioglitazone modulates inflammation and induces neuroprotection in parkinsonian monkeys Journal of Neuroinflammation. 8: 91.
    1. Napolitano M, Costa L, Palermo R, Giovenco A, Vacca A, et al. (2011) Protective effect of pioglitazone, a PPARgamma ligand, in a 3 nitropropionic acid model of Huntington’s disease. Brain Research Bulletin 85: 231–237.
    1. Benedusi V, Martorana F, Brambilla L, Maggi A, Rossi D (2012) The Peroxisome Proliferator-activated Receptor gamma (PPARgamma) Controls Natural Protective Mechanisms against Lipid Peroxidation in Amyotrophic Lateral Sclerosis. The Journal Of Biological Chemistry 287: 35899–35911.
    1. Lourenco MV, Ledo JH (2013) Targeting Alzheimer’s pathology through PPARγ signaling: modulation of microglial function. J Neurosci 33: 5083–5084.
    1. McCarty KD, de Vellis J (1980) Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue. J Cell Biol 85: 890–902.
    1. Perdomo MC, Santos JE, Badinga L (2011) Trans-10, cis-12 conjugated linoleic acid and the PPAR-γ agonist rosiglitazone attenuate lipopolysaccharide-induced TNF-α production by bovine immune cells. Domest Anim Endocrinol 41: 118–25.
    1. Ye F, Zhang ZS, Luo HB, Shen JH, Chen KX, et al. (2006) The dipeptide H-Trp-Glu-OH shows highly antagonistic activity against PPARgamma: bioassay with molecular modeling simulation. Chembiochem 7: 74–82.
    1. Cavaletti G, Tredici G, Petruccioli MG, Dondè E, Tredici P, et al. (2001) Effects of different schedules of oxaliplatin treatment on the peripheral nervous system of the rat. Eur J Cancer 37: 2457–2463.
    1. Leighton GE, Rodriguez RE, Hill RG, Hughes J (1988) kappa-Opioid agonists produce antinociception after i.v. and i.c.v. but not intrathecal administration in the rat. Br J Pharmacol 93: 553–560.
    1. Goullé JP, Mahieu L, Castermant J, Neveu N, Bonneau L, et al. (2005) Metal and metalloid multi-elementary ICP MS validation in whole blood, plasma, urine and hair. Reference values. Forensic Sci Int 153: 39–44.
    1. Morrison JG, White P, McDougall S, Firth JW, Woolfrey SG, et al. (2000) Validation of a highly sensitive ICP-MS method for the determination of platinum in biofluids: application to clinicalpharmacokinetic studies with oxaliplatin. J Pharm Biomed Anal 24: 1–10.
    1. Graham MA, Lockwood GF, Greenslade D, Brienza S, Bayssas M, et al. (2000) Clinical pharmacokinetics of oxaliplatin: a critical review. Clin Cancer Res 6: 1205–1218.
    1. Han CH, Khwaounjoo P, Kilfoyle DH, Hill A, McKeage MJ (2013) Phase I drug-interaction study of effects of calcium and magnesium infusions on oxaliplatin pharmacokinetics and acute neurotoxicity in colorectal cancer patients. BMC Cancer 13: 495.
    1. Freireich EJ, Gehan EA, Rall DP, Schmidt LH, Skipper HE (1996) Quantitative comparison of toxicity of anticancer agents in mouse, rat, hamster, dog, monkey, and man. Cancer Chemother Rep 50: 219–244.
    1. Reagan-Shaw S, Nihal M, Ahamad N (2008) Dose translation from animal to human studies revisited. FASEB J 22: 659–661.
    1. Grisold W, Cavaletti G, Windebank AJ (2012) Peripheral neuropathies from chemotherapeutics and targeted agents: diagnosis, treatment, and prevention. Neuro Oncol Suppl 4: iv 45–54.
    1. Wang B, Veldhoven PP, Brees C, Rubio C, Nordgren M, et al. (2013) Mitochondria are targets for peroxisome-derived oxidative stress in cultured mammalian cells. Free Radic Biol Med 65: 882–894.
    1. Zanardelli M, Di Cesare Mannelli L, Micheli L, Cinci L, Baptista De Souza D, et al. (2013) oxaliplatin-dependent apoptosis: different apoptotic process in primary astrocytes vs colorectal cancer cells. Proceeding of 36°Congress of the Italian Society of Pharmacology, Turin, 23–26 October. Available: .
    1. Alberts B, Johnson A, Lewis J, Raff M, Roberts K, et al. (2002) Intracellular Compartments and Protein Sorting. In: Garland Science, editor. Molecular biology of the cell, 4th edition. New York.
    1. Powers SK, Lennon SL (1999) Analysis of cellular responses to free radicals: focus on exercise and skeletal muscle. Proc Nutr Soc 58: 1025–1033.
    1. Venkateshappa C, Harish G, Mahadevan A, Srinivas Bharath MM, Shankar SK (2012) Elevated oxidative stress and decreased antioxidant function in the human hippocampus and frontal cortex with increasing age: implications for neurodegeneration in Alzheimer’s disease. Neurochem Res 37: 1601–1614.
    1. Khan MM, Ahmad A, Ishrat T, Khan MB, Hoda MN, et al. (2010) Resveratrol attenuates 6-hydroxydopamine-induced oxidative damage and dopamine depletion in rat model of Parkinson’s disease. Brain Res 1328: 139–151.
    1. Kamboj SS, Vasishta RK, Sandir R (2010) N-acetylcysteine inhibits hyperglycemia-induced oxidative stress and apoptosis markers in diabetic neuropathy. J Neurochem 112: 77–91.
    1. Masoud A, Sandhir R (2012) Increased oxidative stress is associated with the development of organophosphate-induced delayed neuropathy. Hum Exp Toxicol 31: 1214–1227.
    1. Gao YJ, Ji RR (2010) Targeting astrocyte signaling for chronic pain. Neurotherapeutics 7: 482–493.
    1. Salvemini D, Neumann W (2010) Targeting peroxynitrite driven nitroxidative stress with synzymes: A novel therapeutic approach in chronic pain management. Life Sci 86: 604–614.
    1. Ribeiro D, Castro I, Fahimi HD, Schrader M (2012) Peroxisome morphology in pathology. Histol Histopathol 27: 661–676.
    1. Chen Y, Chan PH, Swanson RA (2001) Astrocytes overexpressing Cu, Zn superoxide dismutase have increased resistance to oxidative injury. Glia 33: 343–347.
    1. Choi JH, Kim DH, Yun IJ, Chang JH, Chun BG, et al. (2007) Zaprinast inhibits hydrogen peroxide-induced lysosomal destabilization and cell death in astrocytes. Eur J Pharmacol 571: 106–115.
    1. Hunter RL, Dragicevic N, Seifert K, Choi DY, Liu M, et al. (2007) Inflammation induces mitochondrial dysfunction and dopaminergic neurodegeneration in the nigrostriatal system. J Neurochem 100: 1375–1386.
    1. Sarruf DA, Yu F, Nguyen HT, Williams DL, Printz RL, et al. (2009) Expression of peroxisome proliferator-activated receptor-gamma in key neuronal subsets regulating glucose metabolism and energy homeostasis. Endocrinology 150: 707–712.
    1. Carniglia L, Durand D, Caruso C, Lasaga M (2013) Effect of NDP-α-MSH on PPAR-γ and -β expression and anti-inflammatory cytokine release in rat astrocytes and microglia. PLoS One 8: e57313.
    1. Gray E, Ginty M, Kemp K, Scolding N, Wilkings A (2012) The PPAR- γ agonist pioglitazone protects cortical neurons from inflammatory mediators via improvement in peroxisomal function. J Neuroinflammation 9: 63.
    1. Scholz J, Woolf CJ (2007) The neuropathic pain triad: neurons, immune cells and glia. Nat Neurosci 10: 1361–1368.
    1. Milligan ED, Watkins LR (2009) Pathological and protective roles of glia in chronic pain. Nat Rev Neurosci 10: 23–36.
    1. Boivin A, Pineau I, Barrette B, Filali M, Vallières N, et al. (2007) Toll-like receptor signaling is critical for Wallerian degeneration and functional recovery after peripheral nerve injury. J Neurosci 27: 12565–12576.
    1. Hong S, Xin Y, HaiQin W, GuiLian Z, Ru Z, et al. (2012) The PPARγ agonist rosiglitazone prevents cognitive impairment by inhibiting astrocyte activation and oxidative stress following pilocarpine-induced status epilepticus. Neurol Sci 33: 559–566.
    1. Lee EY, Lee JE, Park JH, Shin IC, Koh HC (2012) Rosiglitazone, a PPAR-γ agonist, protects against striatal dopaminergic neurodegeneration induced by 6-OHDA lesions in the substantia nigra of rats. Toxicol Lett 213: 332–344.
    1. Festuccia WT, Oztezcan S, Laplante M, Berthiaume M, Michel C, et al. (2008) Peroxisome proliferato-activated receptor-gamma-mediated positive energy balance in the rat is associated with reduced sympathetic drive to adipose tissues and thyroid status. Endocrinology 149: 2121–2130.
    1. Strum JC, Shehee R, Virley D, Richardson J, Mattie M, et al. (2007) Rosiglitazone induces mitochondrial biogenesis in mouse brain. J Alzheimers Dis 11: 45–51.
    1. Sheu WH, Chuang HC, Cheng SM, Lee MR, Chou CC, et al. (2011) Microdialysis combined blood sampling technique for the determination of rosiglitazone and glucose in brain and blood of gerbils subjected to cerebral ischemia. J Pharm Biomed Anal 54: 759–764.
    1. Lu M, Sarruf DA, Talukdar S, Sharma S, Li P, et al. (2011) Brain PPAR-γ promotes obesity and is required for the insulin-sensitizing effect of thiazolidinediones. Nat Med 17: 618–622.
    1. Blanquicett C, Roman J, Hart CM (2008) Thiazolidinediones as anti-cancer agents. Cancer Ther 6(A): 25–34.
    1. Joshi H, Pal T, Ramaa CS (2014) A new dawn for the use of thiazolidinediones in cancer therapy. Expert Opin Investig Drugs 23: 501–510.

Source: PubMed

3
Se inscrever