227
Views
13
CrossRef citations to date
0
Altmetric
Reviews

The pregnane X receptor in tuberculosis therapeutics

, , &

Bibliography

Papers of special note have been highlighted as:

• either of interest

•• of considerable interest.

  • Lehmann JM, McKee DD, Watson MA, et al. The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. J Clin Investig. 1998;102:1016.
  • Bertilsson G, Heidrich J, Svensson K, et al. Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction. Proc Natl Acad Sci. 1998;95:12208–12213.
  • Ekins S, Erickson JA. A pharmacophore for human pregnane X receptor ligands. Drug Metab Dispos. 2002;30:96–99.
  • Kliewer SA, Moore JT, Wade L, et al. An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell. 1998;92:73–82.
  • Shukla SJ, Sakamuru S, Huang R, et al. Identification of clinically used drugs that activate pregnane X receptors. Drug Metab Dispos. 2011;39:151–159.
  • Jacobs MN, Nolan GT, Hood SR. Lignans, bacteriocides and organochlorine compounds activate the human pregnane X receptor (PXR). Toxicol Appl Pharmacol. 2005;209:123–133.
  • Landes N, Pfluger P, Kluth D, et al. Vitamin E activates gene expression via the pregnane X receptor. Biochem Pharmacol. 2003;65:269–273.
  • Tabb MM, Sun A, Zhou C, et al. Vitamin K2 regulation of bone homeostasis is mediated by the steroid and xenobiotic receptor SXR. J Biol Chem. 2003;278:43919–43927.
  • Tolson AH, Wang H. Regulation of drug-metabolizing enzymes by xenobiotic receptors: PXR and CAR. Adv Drug Deliv Rev. 2010;62:1238–1249.
  • Iyer M, Reschly EJ, Krasowski MD. Functional evolution of the pregnane X receptor. Expert Opin Drug Metab Toxicol. 2006;2:381–397.
  • McGinnity DF, Zhang G, Kenny JR, et al. Evaluation of multiple in vitro systems for assessment of CYP3A4 induction in drug discovery: human hepatocytes, pregnane X receptor reporter gene, and Fa2N-4 and HepaRG cells. Drug Metab Dispos. 2009;37:1259–1268.
  • Ripp SL, Mills JB, Fahmi OA, et al. Use of immortalized human hepatocytes to predict the magnitude of clinical drug-drug interactions caused by CYP3A4 induction. Drug Metab Dispos. 2006;34:1742–1748.
  • Sinz M, Kim S, Zhu Z, et al. Evaluation of 170 xenobiotics as transactivators of human pregnane X receptor (hPXR) and correlation to known CYP3A4 drug interactions. Curr Drug Metab. 2006;7:375–388.
  • Blumberg B, Sabbagh W, Juguilon H, et al. SXR, a novel steroid and xenobioticsensing nuclear receptor. Genes Dev. 1998;12:3195–3205.
  • Dubrac S, Elentner A, Ebner S, et al. Modulation of T lymphocyte function by the pregnane X receptor. J Immunol. 2010;184:2949–2957.
  • Chang H, Wang J, Hsu C, et al. Pxr modulates hepatotoxicity during anti-tuberculosis treatment: sex matters. Am J Respir Crit Care Med. 2014;189:A1026.

•• Research highlights gender difference and the importance of pregnane X receptor (PXR) polymorphisms in tuberculosis (TB) susceptibility and drug-induced liver injury.

  • Guengerich FP. Common and uncommon cytochrome P450 reactions related to metabolism and chemical toxicity. Chem Res Toxicol. 2001;14:611–650.
  • Jancova P, Anzenbacher P, Anzenbacherova E. Phase II drug metabolizing enzymes. Biomed Pap. 2010;154:103–116.
  • Hines RN, McCarver DG. The ontogeny of human drug-metabolizing enzymes: phase I oxidative enzymes. J Pharmacol Exp Ther. 2002;300:355–360.
  • Xu C, Li CY-T, Kong A-NT. Induction of phase I, II and III drug metabolism/transport by xenobiotics. Arch Pharm Res. 2005;28:249–268.
  • Chen Y, Ferguson SS, Negishi M, et al. Induction of human CYP2C9 by rifampicin, hyperforin, and phenobarbital is mediated by the pregnane X receptor. J Pharmacol Exp Ther. 2004;308:495–501.
  • Gerbal-Chaloin S, Pascussi J-M, Pichard-Garcia L, et al. Induction of CYP2C genes in human hepatocytes in primary culture. Drug Metab Dispos. 2001;29:242–251.
  • Goodwin B, Hodgson E, Liddle C. The orphan human pregnane X receptor mediates the transcriptional activation of CYP3A4 by rifampicin through a distal enhancer module. Mol Pharmacol. 1999;56:1329–1339.
  • Pascussi JM, Robert A, Nguyen M, et al. Possible involvement of pregnane X receptor–enhanced CYP24 expression in drug-induced osteomalacia. J Clin Investig. 2005;115:177–186.
  • Falkner K, Pinaire J, Xiao G-H, et al. Regulation of the rat glutathione S-transferase A2 gene by glucocorticoids: involvement of both the glucocorticoid and pregnane X receptors. Mol Pharmacol. 2001;60:611–619.
  • Runge-Morris M, Wu W, Kocarek TA. Regulation of rat hepatic hydroxysteroid sulfotransferase (SULT2-40/41) gene expression by glucocorticoids: evidence for a dual mechanism of transcriptional control. Mol Pharmacol. 1999;56:1198–1206.
  • Chen C, Staudinger JL, Klaassen CD. Nuclear receptor, pregnane X receptor, is required for induction of UDP-glucuronosyltransferases in mouse liver by pregnenolone-16α-carbonitrile. Drug Metab Dispos. 2003;31:908–915.
  • Sugatani J, Nishitani S, Yamakawa K, et al. Transcriptional regulation of human UGT1A1 gene expression: activated glucocorticoid receptor enhances constitutive androstane receptor/pregnane X receptor-mediated UDP-glucuronosyltransferase 1A1 regulation with glucocorticoid receptor-interacting protein 1. Mol Pharmacol. 2005;67:845–855.
  • Xie W, Yeuh M-F, Radominska-Pandya A, et al. Control of steroid, heme, and carcinogen metabolism by nuclear pregnane X receptor and constitutive androstane receptor. Proc Natl Acad Sci. 2003;100:4150–4155.
  • Geick A, Eichelbaum M, Burk O. Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin. J Biol Chem. 2001;276:14581–14587.
  • Mills JB, Rose KA, Sadagopan N, et al. Induction of drug metabolism enzymes and MDR1 using a novel human hepatocyte cell line. J Pharmacol Exp Ther. 2004;309:303–309.
  • Synold TW, Dussault I, Forman BM. The orphan nuclear receptor SXR coordinately regulates drug metabolism and efflux. Nat Med. 2001;7:584–590.
  • Moore LB, Goodwin B, Jones SA, et al. St. John’s wort induces hepatic drug metabolism through activation of the pregnane X receptor. Proc Natl Acad Sci. 2000;97:7500–7502.
  • Chen Y, Tang Y, Wang M-T, et al. Human pregnane X receptor and resistance to chemotherapy in prostate cancer. Cancer Res. 2007;67:10361–10367.
  • Jiang H, Chen K, He J, et al. Association of pregnane X receptor with multidrug resistance-related protein 3 and its role in human colon cancer chemoresistance. J Gastrointest Surg. 2009;13:1831–1838.
  • Dotzlaw H, Leygue E, Watson P, et al. The human orphan receptor PXR messenger RNA is expressed in both normal and neoplastic breast tissue. Clin Cancer Res. 1999;5:2103–2107.
  • Masuyama H, Hiramatsu Y, Kodama J-I, et al. Expression and potential roles of pregnane X receptor in endometrial cancer. J Clin Endocrinol Metab. 2003;88:4446–4454.
  • Miki Y, Suzuki T, Kitada K, et al. Expression of the steroid and xenobiotic receptor and its possible target gene, organic anion transporting polypeptide-A, in human breast carcinoma. Cancer Res. 2006;66:535–542.
  • Bhalla S, Ozalp C, Fang S, et al. Ligand-activated pregnane X receptor interferes with HNF-4 signaling by targeting a common coactivator PGC-1α functional implications in hepatic cholesterol and glucose metabolism. J Biol Chem. 2004;279:45139–45147.
  • Kodama S, Koike C, Negishi M, et al. Nuclear receptors CAR and PXR cross talk with FOXO1 to regulate genes that encode drug-metabolizing and gluconeogenic enzymes. Mol Cell Biol. 2004;24:7931–7940.
  • Kodama S, Moore R, Yamamoto Y, et al. Human nuclear pregnane X receptor cross-talk with CREB to repress cAMP activation of the glucose-6-phosphatase gene. Biochem J. 2007;407:373–381.
  • Nakamura K, Moore R, Negishi M, et al. Nuclear pregnane X receptor cross-talk with FoxA2 to mediate drug-induced regulation of lipid metabolism in fasting mouse liver. J Biol Chem. 2007;282:9768–9776.
  • Kodama S, Negishi M. PXR cross-talks with internal and external signals in physiological and pathophysiological responses. Drug Metab Rev. 2013;45:300–310.

• Review article summarizing the physiological roles of PXR.

  • Zhou J, Zhai Y, Mu Y, et al. A novel pregnane X receptor-mediated and sterol regulatory element-binding protein-independent lipogenic pathway. J Biol Chem. 2006;281:15013–15020.
  • Schote AB, Turner JD, Schiltz J, et al. Nuclear receptors in human immune cells: expression and correlations. Mol Immunol. 2007;44:1436–1445.
  • Gupta S, Grieco MH, Siegel I. Suppression of T-lymphocyte rosettes by rifampinstudies in normals and patients with tuberculosis. Ann Intern Med. 1975;82:484–488.
  • Tsankov N, Grozdev I. Rifampicin in the treatment of psoriasis. J Eur Acad Dermatol Venereol. 2009;23:93–95.
  • Calleja C, Pascussi J, Mani J, et al. The antibiotic rifampicin is a nonsteroidal ligand and activator of the human glucocorticoid receptor. Nat Med. 1998;4:92–96.
  • Hoffmann A, Baltimore D. Circuitry of nuclear factor κB signaling. Immunol Rev. 2006;210:171–186.
  • Zhou C, Tabb MM, Nelson EL, et al. Mutual repression between steroid and xenobiotic receptor and NF-κB signaling pathways links xenobiotic metabolism and inflammation. J Clin Investig. 2006;116:2280–2289.
  • Shah YM, Ma X, Morimura K, et al. Pregnane X receptor activation ameliorates DSS-induced inflammatory bowel disease via inhibition of NF-κB target gene expression. Am J Physiology-Gastrointestinal Liver Physiol. 2007;292:G1114–G22.

•• Research showing the beneficial effect of PXR activation in inflammatory bowel disease.

  • Wehrli W, Staehelin M. Actions of the rifamycins. Bacteriol Rev. 1971;35:290.
  • Luo G, Cunningham M, Kim S, et al. CYP3A4 induction by drugs: correlation between a pregnane X receptor reporter gene assay and CYP3A4 expression in human hepatocytes. Drug Metab Dispos. 2002;30:795–804.
  • Feng HJ, Huang SL, Wang W, et al. The induction effect of rifampicin on activity of mephenytoin 4′‐hydroxylase related to M1 mutation of CYP2C19 and gene dose. Br J Clin Pharmacol. 1998;45:27–29.
  • Goodwin B, Moore LB, Stoltz CM, et al. Regulation of the human CYP2B6 gene by the nuclear pregnane X receptor. Mol Pharmacol. 2001;60:427–431.
  • Aristoff PA, Garcia GA, Kirchhoff PD, et al. Rifamycins–Obstacles and opportunities. Tuberculosis. 2010;90:94–118.
  • Mae T, Hosoe K, Yamamoto T, et al. Effect of a new rifamycin derivative, rifalazil, on liver microsomal enzyme induction in rat and dog. Xenobiotica. 1998;28:759–766.
  • Zhang H, LeCulyse E, Liu L, et al. Rat pregnane X receptor: molecular cloning, tissue distribution, and xenobiotic regulation. Arch Biochem Biophys. 1999;368:14–22.
  • World Health Organization. Treatment of tuberculosis: guidelines. Geneva: World Health Organization; 2010.
  • Li AP, Reith MK, Rasmussen A, et al. Primary human hepatocytes as a tool for the evaluation of structure—activity relationship in cytochrome P450 induction potential of xenobiotics: evaluation of rifampin, rifapentine and rifabutin. Chem Biol Interact. 1997;107:17–30.
  • Burman WJ, Gallicano K, Peloquin C. Comparative pharmacokinetics and pharmacodynamics of the rifamycin antibacterials. Clin Pharmacokinet. 2001;40:327–341.
  • Acocella G, Pagani V, Marchetti M, et al. Kinetic studies on rifampicin. Chemotherapy. 1971;16:356–370.
  • Immanuel C, Jayasankar K, Narayana A, et al. Self-induction of rifampicin metabolism in man. Indian J Med Res. 1985;82:381–387.
  • Venkatesan K. Pharmacokinetic drug interactions with rifampicin. Clin Pharmacokinet. 1992;22:47–65.
  • Evans DAP, Manley KA, McKusick VA. Genetic control of isoniazid metabolism in man. Br Med J. 1960;2:485.
  • Ono Y, Wu X, Noda A, et al. Participation of P450-dependent oxidation of isoniazid in isonicotinic acid formation in rat liver. Biol Pharm Bull. 1998;21:421–425.
  • Sarma GR, Immanuel C, Kailasam S, et al. Rifampin-induced release of hydrazine from isoniazid. A possible cause of hepatitis during treatment of tuberculosis with regimens containing isoniazid and rifampin. Am Rev Respir Dis. 1986;133:1072.
  • Liu K, Li F, Lu J, et al. Role of CYP3A in isoniazid metabolism in vivo. Drug Metab Pharmacokinet. 2014;29:219.
  • Lacroix C, Tranvouez J, Phan HT, et al. Pharmacokinetics of pyrazinamide and its metabolites in patients with hepatic cirrhotic insufficiency. Arzneimittel-Forschung. 1990;40:76–79.
  • Weiner I, Tinker JP. Pharmacology of pyrazinamide: Metabolic and renal function studies related to the mechanism of drug-induced urate retention. J Pharmacol Exp Ther. 1972;180:411–434.
  • Yamamoto T, Moriwaki Y, Suda M, et al. Effect of BOF-4272 on the oxidation of allopurinol and pyrazinamide in vivo: Is xanthine dehydrogenase or aldehyde oxidase more important in oxidizing both allopurinol and pyrazinamide? Biochem Pharmacol. 1993;46:2277–2284.
  • Moriwaki Y, Yamamoto T, Nasako Y, et al. In vitro oxidation of pyrazinamide and allopurinol by rat liver aldehyde oxidase. Biochem Pharmacol. 1993;46:975–981.
  • Hartkoorn RC, Chandler B, Owen A, et al. Differential drug susceptibility of intracellular and extracellular tuberculosis, and the impact of P-glycoprotein. Tuberculosis. 2007;87:248–255.
  • Mahajan R. Bedaquiline: First FDA-approved tuberculosis drug in 40 years. Int J Appl Basic Med Res. 2013;3:1.
  • Andries K, Verhasselt P, Guillemont J, et al. A diarylquinoline drug active on the ATP synthase of Mycobacterium tuberculosis. Science. 2005;307:223–227.
  • Van Heeswijk R, Lachaert R, Leopold L, et al. The effect of CYP3A4 inhibition on the clinical pharmacokinetics of TMC207. 38th Union World Conference on Lung Health; 2007 Nov 8–12; Capetown.
  • Matteelli A, Carvalho AC, Dooley KE, et al. TMC207: the first compound of a new class of potent anti-tuberculosis drugs. Future Microbiol. 2010;5:849–858.
  • Everitt D, Winter H, Egizi E. Pharmacokinetic interaction between the investigational anti-tuberculosis agent TMC207 and rifampicin or rifapentine (Abstract MOAB0304). 19th International AIDS Conference; 2012; Washington, DC.
  • Yew W. Clinically significant interactions with drugs used in the treatment of tuberculosis. Drug Saf. 2002;25:111–113.
  • Yildiz A, Sever MS, Türkmen A, et al. Tuberculosis after renal transplantation: experience of one Turkish centre. Nephrol Dial Transplant. 1998;13:1872–1875.
  • Ernest CS, Hall SD, Jones DR. Mechanism-based inactivation of CYP3A by HIV protease inhibitors. J Pharmacol Exp Ther. 2005;312:583–591.
  • Kumar GN, Jayanti V, Lee RD, et al. In vitro metabolism of the HIV-1 protease inhibitor ABT-378: species comparison and metabolite identification. Drug Metab Dispos. 1999;27:86–91.
  • Singh R, Chang SY, Taylor LC. In vitro metabolism of a potent HIV‐protease inhibitor (141W94) using rat, monkey and human liver S9. Rapid Commun Mass Spectrom. 1996;10:1019–1026.
  • Burman WJ, Gallicano K, Peloquin C. Therapeutic implications of drug interactions in the treatment of human immunodeficiency virus-related tuberculosis. Clin Infect Dis. 1999;28:419–429.
  • Borin MT, Chambers JH, Carel BJ, et al. Pharmacokinetic study of the interaction between rifampin and delavirdine mesylate. Clin Pharmacol Ther. 1997;61:544–553.
  • Borin MT, Chambers JH, Carel BJ, et al. Pharmacokinetic study of the interaction between rifabutin and delavirdine mesylate in HIV-1 infected patients. Antiviral Res. 1997;35:53–63.
  • Erickson DA, Mather G, Trager WF, et al. Characterization of the in vitro biotransformation of the HIV-1 reverse transcriptase inhibitor nevirapine by human hepatic cytochromes P-450. Drug Metab Dispos. 1999;27:1488–1495.
  • Cohen K, Van Cutsem G, Boulle A, et al. Effect of rifampicin-based antitubercular therapy on nevirapine plasma concentrations in South African adults with HIV-associated tuberculosis. J Antimicrob Chemother. 2008;61:389–393.
  • Bhatt NB, Baudin E, Meggi B, et al. Nevirapine or efavirenz for tuberculosis and HIV coinfected patients: exposure and virological failure relationship. J Antimicrob Chemother. 2014;70:225–232.
  • Ogburn ET, Jones DR, Masters AR, et al. Efavirenz primary and secondary metabolism in vitro and in vivo: identification of novel metabolic pathways and cytochrome P450 2A6 as the principal catalyst of efavirenz 7-hydroxylation. Drug Metab Dispos. 2010;38:1218–1229.
  • Ward BA, Gorski JC, Jones DR, et al. The cytochrome P450 2B6 (CYP2B6) is the main catalyst of efavirenz primary and secondary metabolism: implication for HIV/AIDS therapy and utility of efavirenz as a substrate marker of CYP2B6 catalytic activity. J Pharmacol Exp Ther. 2003;306:287–300.
  • Cabrera SE, Cordero M, Iglesias A, et al. Efavirenz–rifampicin interaction: therapeutic drug monitoring to efavirenz dosage optimization in HIV/TBC patients. Aids. 2008;22:2549–2551.
  • Aoyama T, Yamano S, Waxman D, et al. Cytochrome P-450 hPCN3, a novel cytochrome P-450 IIIA gene product that is differentially expressed in adult human liver. cDNA and deduced amino acid sequence and distinct specificities of cDNA-expressed hPCN1 and hPCN3 for the metabolism of steroid hormones and cyclosporine. J Biol Chem. 1989;264:10388–10395.
  • Saeki T, Ueda K, Tanigawara Y, et al. Human P-glycoprotein transports cyclosporin A and FK506. J Biol Chem. 1993;268:6077–6080.
  • Sakhuja V, Jha V, Varma PP, et al. The high incidence of tuberculosis among renal transplant recipients in India. Transplantation. 1996;61:211–215.
  • Sattler M, Guengerich FP, Yun C-H, et al. Cytochrome P-450 3A enzymes are responsible for biotransformation of FK506 and rapamycin in man and rat. Drug Metab Dispos. 1992;20:753–761.
  • Peschke B, Ernst W, Gossmann J, et al. Antituberculous drugs in kidney transplant recipients treated with cyclosporine. Transplantation. 1993;56:236–238.
  • Chenhsu R-Y, Loong -C-C, Chou M-H, et al. Renal allograft dysfunction associated with rifampin–tacrolimus interaction. Ann Pharmacother. 2000;34:27–31.
  • Hebert MF, Fisher RM, Marsh CL, et al. Effects of rifampin on tacrolimus pharmacokinetics in healthy volunteers. J Clin Pharmacol. 1999;39:91–96.
  • Larson AM, Polson J, Fontana RJ, et al. Acetaminophen‐induced acute liver failure: results of a United States multicenter, prospective study. Hepatology. 2005;42:1364–1372.
  • Yue J, Peng R-X, Yang J, et al. CYP2E1 mediated isoniazid-induced hepatotoxicity in rats. Acta Pharmacol Sin. 2004;25:699–704.
  • Cheng J, Ma X, Krausz KW, et al. Rifampicin-activated human pregnane X receptor and CYP3A4 induction enhance acetaminophen-induced toxicity. Drug Metab Dispos. 2009;37:1611–1621.
  • Steele MA, Burk R, DesPrez R. Toxic hepatitis with isoniazid and rifampin. A meta-analysis. Chest J. 1991;99:465–471.
  • Miguet J, Mavier P, Soussy C, et al. Induction of hepatic microsomal enzymes after brief administration of rifampicin in man. Gastroenterology. 1977;72:924–926.
  • Metushi IG, Sanders C, Lee WM, et al. Detection of anti‐isoniazid and anti–cytochrome P450 antibodies in patients with isoniazid‐induced liver failure. Hepatology. 2014;59:1084–1093.
  • Li F, Lu J, Cheng J, et al. Human PXR modulates hepatotoxicity associated with rifampicin and isoniazid co-therapy. Nat Med. 2013;19:418–420.
  • Control CfD, Prevention. Update: fatal and severe liver injuries associated with rifampin and pyrazinamide for latent tuberculosis infection, and revisions in American Thoracic Society/CDC recommendations–United States, 2001. MMWR Morb Mortal Wkly Rep. 2001;50:733.
  • Sarma GR, Acharyulu G, Kannapiran M, et al. Role of rifampicin in arthralgia induced by pyrazinamide. Tubercle. 1983;64:93–100.
  • Fallahi-Sichani M, Kirschner DE, Linderman JJ. NF-κB signaling dynamics play a key role in infection control in tuberculosis. Front Physiol. 2012;3:1–25.
  • Bold TD, Ernst JD. Who benefits from granulomas, mycobacteria or host? Cell. 2009;136:17–19.
  • Ramakrishnan L. Revisiting the role of the granuloma in tuberculosis. Nat Rev Immunol. 2012;12:352–366.
  • Sasindran SJ, Torrelles JB. Mycobacterium tuberculosis infection and inflammation: what is beneficial for the host and for the bacterium? Front Microbiol. 2011;2:2.
  • Davis J, Ramakrishnan L. The role of the granuloma in expansion and dissemination of early tuberculous infection. Cell. 2009;136:37–49.

•• Research discussing the role of inflammation in the early progression of TB disease.

  • Orme IM, Robinson RT, Cooper AM. The balance between protective and pathogenic immune responses in the TB-infected lung. Nat Immunol. 2015;16:57–63.

• Review discussing the role of balancing the immune response in TB disease.

•• Review discussing the benefits of anti-inflammatory agents as add-on therapy in TB disease.

  • Clay H, Volkman HE, Ramakrishnan L. Tumor necrosis factor signaling mediates resistance to mycobacteria by inhibiting bacterial growth and macrophage death. Immunity. 2008;29:283–294.
  • Agarwal N, Lamichhane G, Gupta R, et al. Cyclic AMP intoxication of macrophages by a mycobacterium tuberculosis adenylate cyclase. Nature. 2009;460:98–102.
  • Critchley JA, Young F, Orton L, et al. Corticosteroids for prevention of mortality in people with tuberculosis: a systematic review and meta-analysis. Lancet Infect Dis. 2013;13:223–237.
  • Wallis RS. Corticosteroid effects on sputum culture in pulmonary tuberculosis: a meta-regression analysis. Open forum infectious diseases. Oxford: Oxford University Press; 2014.
  • Litwin A, Brooks SM, Claes F. A pilot study concerning the early immunosuppressive effects of rifampin in man. Chest J. 1974;65:548–551.
  • Serrou B, Solassol C, Joyeux H, et al. Immunosuppressive effect of rifampicin. Transplantation. 1972;14:654–655.
  • Zhang Y, Yew W. Mechanisms of drug resistance in mycobacterium tuberculosis [State of the art series. Drug-resistant tuberculosis. Edited by CY. Chiang. Number 1 in the series]. Int J Tuberculosis Lung Dis. 2009;13:1320–1330.
  • Faustini A, Hall AJ, Perucci CA. Risk factors for multidrug resistant tuberculosis in Europe: a systematic review. Thorax. 2006;61:158–163.
  • Zachariah R, Spielmann M, Harries A, et al. Moderate to severe malnutrition in patients with tuberculosis is a risk factor associated with early death. Trans R Soc Trop Med Hyg. 2002;96:291–294.
  • Cegielski J, McMurray D. The relationship between malnutrition and tuberculosis: evidence from studies in humans and experimental animals. Int J Tuberculosis Lung Dis. 2004;8:286–298.
  • Gumbo T, Louie A, Deziel MR, et al. Concentration-dependent Mycobacterium tuberculosis killing and prevention of resistance by rifampin. Antimicrob Agents Chemother. 2007;51:3781–3788.
  • Goutelle S, Bourguignon L, Maire PH, et al. Population modeling and Monte Carlo simulation study of the pharmacokinetics and antituberculosis pharmacodynamics of rifampin in lungs. Antimicrob Agents Chemother. 2009;53:2974–2981.
  • Gumbo T, Dona CSS, Meek C, et al. Pharmacokinetics-pharmacodynamics of pyrazinamide in a novel in vitro model of tuberculosis for sterilizing effect: a paradigm for faster assessment of new antituberculosis drugs. Antimicrob Agents Chemother. 2009;53:3197–3204.
  • Kjellsson MC, Via LE, Goh A, et al. Pharmacokinetic evaluation of the penetration of antituberculosis agents in rabbit pulmonary lesions. Antimicrob Agents Chemother. 2012;56:446–457.
  • Peloquin C. What is the ‘right’dose of rifampin?[Editorial]. Int J Tuberculosis Lung Dis. 2003;7:3–5.
  • Hukkanen J, Pelkonen O, Hakkola J, et al. Expression and regulation of xenobiotic-metabolizing cytochrome P450 (CYP) enzymes in human lung. CRC Crit Rev Toxicol. 2002;32:391–411.
  • Scheffer G, Pijnenborg A, Smit E, et al. Multidrug resistance related molecules in human and murine lung. J Clin Pathol. 2002;55:332–339.
  • van der Deen M, De Vries E, Timens W, et al. ATP-binding cassette (ABC) transporters in normal and pathological lung. Respir Res. 2005;6:59–74.
  • Chen Y, Tang Y, Chen S, et al. Regulation of drug resistance by human pregnane X receptor in breast cancer. Cancer Biol Ther. 2009;8:1265–1272.
  • Elbireer S, Guwatudde D, Mudiope P, et al. Tuberculosis treatment default among HIV‐TB co‐infected patients in urban Uganda. Trop Med Int Health. 2011;16:981–987.
  • Tobin DM, Roca FJ, Oh SF, et al. Host genotype-specific therapies can optimize the inflammatory response to mycobacterial infections. Cell. 2012;148:434–446.
  • Gardam MA, Keystone EC, Menzies R, et al. Anti-tumour necrosis factor agents and tuberculosis risk: mechanisms of action and clinical management. Lancet Infect Dis. 2003;3:148–155.
  • Imazeki I, Matsuzaki J, Tsuji K, et al. Immunomodulating effect of vitamin D3 derivatives on type-1 cellular immunity. Biomed Res. 2006;27:1–9.
  • Lemire JM. Immunomodulatory role of 1, 25‐dihydroxyvitamin D3. J Cell Biochem. 1992;49:26–31.
  • Vidyarani M, Selvaraj P, Raghavan S, et al. Regulatory role of 1, 25-dihydroxyvitamin D 3 and vitamin D receptor gene variants on intracellular granzyme A expression in pulmonary tuberculosis. Exp Mol Pathol. 2009;86:69–73.
  • Coumoul X, Diry M, Barouki R. PXR-dependent induction of human CYP3A4 gene expression by organochlorine pesticides. Biochem Pharmacol. 2002;64:1513–1519.
  • Korf H, Vander Beken S, Romano M, et al. Liver X receptors contribute to the protective immune response against mycobacterium tuberculosis in mice. J Clin Invest. 2009;119:1626.
  • Mahajan S, Dkhar HK, Chandra V, et al. Mycobacterium tuberculosis modulates macrophage lipid-sensing nuclear receptors PPARγ and TR4 for survival. J Immunol. 2012;188:5593–5603.
  • Rajaram MV, Brooks MN, Morris JD, et al. Mycobacterium tuberculosis activates human macrophage peroxisome proliferator-activated receptor γ linking mannose receptor recognition to regulation of immune responses. J Immunol. 2010;185:929–942.
  • Almeida PE, Silva AR, Maya-Monteiro CM, et al. Mycobacterium bovis bacillus Calmette-Guérin infection induces TLR2-dependent peroxisome proliferator-activated receptor γ expression and activation: functions in inflammation, lipid metabolism, and pathogenesis. J Immunol. 2009;183:1337–1345.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.