Abstract
Severe sepsis is a common, expensive, and fatal condition with as many deaths annually as those from acute myocardial infarctions. The average cost per case seems to exceed $22,000. The increased morbidity and mortality attributed to sepsis could be due to the lack of our understanding of mediators and factors responsible for early cellular alterations and thus could not be intervened which result in progressive deterioration of cell and organ function and even death. It has been well documented that hepatocellular dysfunction occurs early in sepsis and it contributes to multiple organ failure and ultimately death; however the exact mechanism is poorly understood. We and others have shown that cytochrome P-450 (CYP) enzyme system, a superfamily of heme proteins responsible for the metabolism of a variety of endogenous and exogenous substances, plays a crucial role in the prevention of hepatocellular dysfunction in sepsis. In this review, we describe the alterations of CYP enzymes in the experimental model of sepsis and provide the limited information available in septic and severely injured patients. We also review the potential mechanism for the alterations of CYP enzymes in sepsis. Finally, we highlight the importance of future studies needed to understand the regulation of CYP isoforms to develop therapy for hepatocellular dysfunction in sepsis.
Keywords: Cytochrome P-450, CYP enzymes, hepatic dysfunction, liver, sepsis
Introduction
The CYP enzyme system is a superfamily of heme proteins responsible for the metabolism of a vast array of endogenous and exogenous substances. To date, seventeen mammalian CYP gene families have been identified [1]. Within the CYP families, CYP isoforms 1–3 metabolize more than 90% of known drugs [2]. The CYP enzymes are generally found in the hepatocyte endoplasmic reticulum and microsomes while they exist in varying amounts in most extra-hepatic tissues. Environmental factors, foods, social habits and disease conditions are all known to alter CYP enzymes [3]. We [4–6] and others [7] have shown that CYP isoforms are significantly downregulated in sepsis and this decrease is due to the reduction of AhR and Arnt, two critical transcription factors involved in the regulation of CYP1A2 mRNA. Interestingly, our studies further showed that AhR and Arnt expressions were inversely correlated with pro-inflammatory cytokines in sepsis and that exposure of cells with such cytokines downregulated these transcription factors.
Liver dysfunction in sepsis
Sepsis and the ensuing septic shock and multiple organ failure continue to be the most common causes of death in surgical intensive care units [8–12]. Despite advances in the management of trauma victims, the incidence of sepsis and septic shock has increased significantly over the past two decades [11, 13–15]. It has been estimated that in the United States alone more than 750,000 patients develop sepsis and septic shock each year with an overall mortality of 28.6% [16]. Severe sepsis is a common, expensive, and frequently fatal condition, with as many deaths annually as those from acute myocardial infarction. The average costs per case were $22,100, with annual total costs of more than $16 billion nationally [16]. The increased morbidity and mortality attributed to sepsis could be due to the fact that the mediators or factors responsible for early cellular alterations are not fully understood and consequently not prevented, leading to a progressive deterioration of cell and organ function and even death.
A number of animal models such as bacteremia, endotoxemia, and endotoxic shock have been used to study the pathobiology of sepsis and such models have provided valuable information regarding the mechanisms responsible for cell and organ dysfunction under those conditions [17–23]. Other investigators have utilized the model of cecal ligation and puncture (CLP) to produce polymicrobial sepsis [18, 19, 24–32]. The model of CLP mimics many features of clinical sepsis-peritonitis. This model of sepsis is associated with an early, hyperdynamic phase (i.e., 2–10 h after CLP; characterized by increased cardiac output and tissue perfusion, decreased vascular resistance, hyperglycemia and hyperinsulinemia), which is followed by a late, hypodynamic phase (16 h after CLP and later; characterized by reduced cardiac output and tissue perfusion, increased vascular resistance, hypoglycemia and hypo-insulinemia) [30, 33–36]. By using the CLP model of sepsis in the rat, we have shown that hepatocellular dysfunction (as assessed by indocyanine green clearance) occurred early after sepsis despite hyperdynamic circulation including hepatic hyperperfusion [34, 37–40].
Kupffer cells (KCs) play an important role in producing pro-inflammatory cytokines such as TNF-α and IL-1β in sepsis [41, 42]. Administration of recombinant TNF-α in normal animals depressed hepatocellular function [43]. The exact mechanism of hepatocellular dysfunction during early sepsis is poorly understood. We and others have shown that cytochrome P450 (CYP) enzyme system, a superfamily of heme proteins responsible for the metabolism of a variety of endogenous and exogenous substances are significantly altered in sepsis [4–7].
Cytochrome P-450 (CYP) superfamily
The CYP enzyme system is a superfamily of heme proteins responsible for the metabolism of a vast array of endogenous and exogenous substances (xenobiotics) [44–47]. Seventeen mammalian CYP gene families have been identified, encoding approximately 60 distinct CYP isoforms [1]. CYP structure has been deduced and residues required for substrate binding, electron transfer, and heme binding have been identified [48, 49]. Within CYP families, CYP isoforms 1–4 play important roles in metabolizing a variety of xenobiotics [50] and endogenous compounds including steroids, bile acids, fatty acids, eicosanoids and retinoids [51–53]. Nearly all CYPs are designated with the root “CYP” followed by an Arabic numeral for the gene family (i.e. CYP2), a capital letter for subfamily (i.e. CYP2D) and another Arabic number for a certain gene (i.e. CYP2D6). The enzymes in the same family share almost 40% amino acid identity while the members of the subfamily shares almost 55% identity. CYP 1, 2, 3 families are the most important ones because CYP1A2, 2C9, 2C19, 2D6, 2E1 and 3A4 metabolize more than 90% of known drugs [2]. CYP3A4 is a major member of the P450 superfamily that metabolizes much more substrates than any other CYP enzymes including a number of clinically important drugs [54].
The highest concentrations of CYP isoforms are found in hepatocyte endoplasmic reticulum and microsomes, although CYP isoforms exist in varying amounts in most extra-hepatic tissues. The CYP3A family is most abundant in human liver [55] whereas, CYP1A2 is the predominant isoform in the rat liver [56]. CYP1A2 is involved in the metabolism of various exogenous agents such as theophylline, imipramine, and naproxen and can be inhibited by chemicals such as cimetidine and fluoroquinolones [56]. Moreover, CYP1A2 is a major determinant of lidocaine metabolism, a commonly used hepatic CYP function measure [57]. Another member of the CYP1A family is 1A1 and both CYP1A1 and 1A2 coexist in the rat liver.
Alterations of hepatic CYP450 in sepsis
Environmental factors which include medications (e.g., barbiturates, anticonvulsants, rifampin), foods (cruciferous vegetables), social habits (alcohol consumption, cigarette smoking), disease conditions (diabetes, inflammation, and infection) are all known to alter CYP isoforms [3]. Mediators involved in inflammation and sepsis can also alter hepatic CYP's drug metabolizing capabilities [58–60]. In animal models of endotoxemia and in cultured hepatocytes stimulated by endotoxin, CYP isoforms are dramatically decreased [58, 61–66]. The expression of CYP isoforms were downregulated in cultured hepatocytes stimulated by pro-inflammatory cytokines, TNF-α, IL-1β, IFN-γ or IL-6 [67–73]. However, very little work has been performed with hepatic CYP in animal models of sepsis aside from measuring mixed function oxidase [74] and comparing CYP3A expression in sepsis with endotoxic shock [75].
In this regard, we examined whether the major CYP isoform in the rat liver, CYP1A2, is altered during the progression of sepsis and if so, whether reduction of this enzyme plays any role in the inflammatory response. Our results show that rat hepatic CYP1A2 mRNA was significantly downregulated at 10–20h and its proteins decreased at 20h after CLP [4]. Since hepatic perfusion is increased at the early stage of sepsis (e.g., 10h after CLP [30], the observed downregulation in CYP1A2 due to the decreased hepatic perfusion generally observed in severe sepsis and septic shock. Rather, the decreased CYP1A2 can be, in part, due to increase in pro-inflammatory cytokines such as TNF-α and IL-1β [76] which then directly suppress CYP1A2 mRNA [58, 66, 77].
In fact, it has been reported that pro-inflammatory cytokines released from KCs may directly downregulate hepatocyte CYP isoforms [78]. We have confirmed this finding using a KC and hepatocyte coculture system [6]. We have also shown that CYP inhibition by 1-aminobenzotriazole (ABT) in septic animals resulted in a significant increase in serum TNF-α, IL-1β, IL-6 and lactate, and more severe morphological changes in the liver [4]. This result is in agreement with the findings by Carcillo et al. who used ABT to inhibit CYP in the zymosan-induced inflammation which resulted in aggregation of neutrophils and significantly increased mortality [79]. Thus, the CYP enzyme system is essential in sepsis and its inhibition disposes the animal to exacerbated pro-inflammatory response and tissue injury.
Alterations of CYP in septic and severely injured patients
Very little has been known of the changes in CYP during sepsis or severe injury in humans. Shedlofsky et al. [65] reported that adult human volunteers given endotoxin showed decrease in metabolism of the CYP enzyme probe drugs which included antipyrine, hexobarbital and theophylline. The degree of inhibition of drug metabolism correlated with the circulating plasma levels of IL-6 in these endotoxin treated volunteers. Antipyrine metabolism is considered as a “gold standard” measure of mixed CYP450 mediated drug metabolism in humans. Carillo et al., [60] showed that children with sepsis had a two fold reduction in antipyrine clearance and those with multiple organ failure had a fourfold decrease in antipyrine clearance. Interestingly, this antipyrine clearance was inversely correlated with circulating levels of IL-6, nitrite plus nitrate levels and to number of organ failures.
Harbrecht et al., [80] further demonstrated that CYP activity is differentially altered in severely injured patients. In this study, twenty three multiply injured patients admitted to a trauma critical care unit were compared with healthy volunteers. CYP metabolizing activity was measured using the probe drugs mephenytoin (CYP-2C19), chlorzoxazone (CYP-2E1), dapsone (multiple CYP enzymes) and flurbiprofen (CYP-2C9). Mephenytoin metabolism was suppressed after injury and increased during post-injury recovery, whereas chlorozoxane was suppressed to a lesser degree. Measures of dapson and flurbiprofen metabolism were elevated throughout the study. Chlorzoxazone and mephenytoin metabolism correlated with multiple organ failure. Therefore, the metabolism of selected CYP isoforms may have potential for evaluating acute hepatic dysfunction in critically ill trauma patients.
In another study, 42 septic patients divided into survivors and non-survivors were evaluated for the in vivo activity of CYP using the aminopyrine breath test, a clinically well-established assay of hepatic CYP activity [81] prior to surgery and starting at sepsis onset on a daily basis. CYP activity was significantly decreased during the course of sepsis in both survivors and non-survivors group from the preoperative levels. Interestingly, CYP activity returned to normal levels in the survivor patients while they remained low in the non-survivors during the late phase. This study suggests that the aminopyrine breath test is a clinically useful tool for predicting outcome in the early stages of sepsis and helps when early surgical intervention is concerned.
Potential mechanism of CYP450 alterations
Transcriptional activation of CYPs involves the aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor as well as its nuclear translocator (Arnt) and a chaperone protein, heat shock 90 (HSP90) [82, 83]. Nuclear receptors such as AhR represent a superfamily of ligand modulated transcription factors. They mediate a variety of physiological responses to steroids, retinoids, thyroid hormones, and various xenobiotics [84]. These receptors play a key role in development, cell differentiation, and organ physiology [85]. Unlike water-soluble hormones, steroid hormones can enter the cell by simple or facilitated diffusion and transduce their signals to the genome via intracellular receptors [86]. After binding to its receptor, the hormone-receptor complex moves from the cytoplasm to nucleus, undergoing allosteric changes that enable the complex to bind to high affinity sites in the chromatin and modulate gene transcription [87]. More than 70 distinct members of the nuclear receptor superfamily have been identified [88].
Studies have shown that the transcription of CYP1A2 gene is mediated through the AhR signaling pathway [82, 83, 89]. This is further supported by the finding that the human hepatoma cell line SK-Hep-1 which expresses defective AhR, is associated with the lack of CYP1A2 expression [90]. In the absence of stimulation, AhR exists as a non-DNA-binding, ∼300 kDa heteromeric complex, associated with the molecular chaperone HSP90 in a 1:2 ratio in the cytosol [91]. Upon stimulation, AhR-HSP90 complex enters the nucleus and subsequently dissociates, enabling AhR to be phosphorylated by tyrosine kinase. The activated AhR then forms a heterodimer complex with its nuclear translocator Arnt [92]. Within the nucleus, the AhR-Arnt complex recognizes and binds to the specific regulatory sequences known as the dioxin responsive element (DRE) at the promoter region, and initiates the transcription of the CYP1A2 gene [93, 94]. HSP90 appears to be critical for folding of a ligand-binding conformation in AhR and for the ligand's inducibility [95, 96] (Figure 1).
Figure 1.
Schematic representation of the potential mechanism of CYP1A2 downregulation in sepsis: Transcriptional activation of CYPs involves the aryl hydrocarbon receptor (AhR), nuclear translocator (Arnt) and a chaperone protein, heat shock 90 (HSP90). In the absence of stimulation, AhR exists as a non-DNA binding heterometric complex with HSP90. Upon activation, AhR-HSP90 complex enters the nucleus and HSP90 dissociates, enabling AhR to be tyrosine phosphorylated and bind to Arnt. The AhR-Arnt complex then binds to dioxin responsive element (DRE) at the promoter region of CYP1A2 gene and initiates the transcription. During sepsis, extracellular stimuli (e.g., LPS) upregulate both TNF-α and IL-1β via NFκB and/or MAPK pathways and get released from Kupffer cells. The cytokines then bind to there respective receptors on hepatocytes and by mechanisms which are still unknown, downregulate both Arnt and AhR gene and protein thereby, decrease CYP1A2 mRNA expression.
We examined the expression of AhR and Arnt in both in vitro and in vivo conditions. For in vitro experiments, isolated Kupffer cells and hepatocytes together, Kupffer cells and hepatocytes separated by transwell membrane or hepatocytes alone were cultured in the presence of 100 ng/ml LPS for 24h. AhR and Arnt mRNA expressions were analyzed by reverse-transcription-polymerase chain reaction (RT-PCR) and protein levels were measured by Western blotting. Both AhR and Arnt mRNA and protein were significantly decreased in the LPS treated co-culture whereas; either hepatocytes alone or co-culture separated by membrane produced no changes in AhR or Arnt mRNA and protein. The observed downregulation of AhR and Arnt in the LPS treated co-culture were correlated with CYP1A2 and inversely associated with TNF-α and IL-1β [6]. Both AhR and CYP1A2 were decreased in hepatoma cells (H-4-II-E) treated with TNF-α or IL-1β for 48h [5]. For the in vivo studies, adult male rats were subjected to sepsis by cecal ligation and puncture; hepatic tissues were harvested at 5, 10, and 20h after CLP or sham operation. AhR and Arnt mRNA and protein were assessed. AhR mRNA decreased at 5h and remained downregulated at 10 and 20h after CLP. Hepatic CYP1A2 expression was also decreased at 10 and 20h after CLP [4]. That CYP1A2 and AhR were inversely correlated with TNF-α and IL-1β suggest increase in pro-inflammatory cytokines in sepsis play critical role in downregulating the CYP enzymes in sepsis and severe injury (Figure 1).
Cytokines induce inducible nitric oxide synthetase (iNOS) resulting in nitric oxide (NO) production in many cell types [97]. The iNOS mRNA level significantly increases 24 h after CLP [7, 98]. By the use of the NO inhibitor, it was shown that CLP caused decrease in CYP1A1, CYP1A2, and CYP2E1 and was reversed by the NO inhibitors. This suggests NO might contribute to the suppression of CYP in sepsis. It has also been postulated that since NO binds to the heme center of CYP directly and inhibits CYP activity [99], decreased CYP activity in sepsis could also be due to NO-mediated post-translational modification.
Future Studies and Perspectives
The CYP enzymes are crucial in the acquisition of metabolic activation and inactivation of clinically used drugs and toxins. We and others have shown that the CYP enzymes are significantly decreased in sepsis and severe injury. We further demonstrated that by using an experimental model of sepsis, this downregulation in sepsis is due to the decrease in AhR and Arnt, two critical transcription factors involved in the regulation of CYP1A2 mRNA. Our studies further showed that AhR and Arnt expressions were inversely correlated with pro-inflammatory cytokines in sepsis and that exposure to cells with such cytokines downregulated AhR and Arnt. These data collectively suggest that therapies directed towards decreasing pro-inflammatory cytokine release in sepsis, especially from the liver, can prevent the downregulation of the AhR signaling pathway and thereby protect CYP mRNA transcription. Further studies are warranted to examine the regulation of other CYP isoforms in sepsis and severe injury and delineate the role of post-translational modification such as nitrosylation in CYP activity.
Acknowledgments
This work was supported by NIH grants, R01 GM053008 and R01 GM057468 (P. Wang).
References
- 1.Nelson D.R. Cytochrome P450 and the individuality of species. Arch Biochem Biophys. 1999;369:1–10. doi: 10.1006/abbi.1999.1352. [DOI] [PubMed] [Google Scholar]
- 2.Wang J.F., Zhang C.C., Chou K.C., Wei D.Q. Structure of cytochrome p450s and personalized drug. Curr Med Chem. 2009;16:232–244. doi: 10.2174/092986709787002727. [DOI] [PubMed] [Google Scholar]
- 3.De Paepe P., Belpaire F.M., Buylaert W.A. Pharmacokinetic and pharmacodynamic considerations when treating patients with sepsis and septic shock. Clin Pharmacokinet. 2002;41:1135–1151. doi: 10.2165/00003088-200241140-00002. [DOI] [PubMed] [Google Scholar]
- 4.Crawford J.H., Yang S., Zhou M., Simms H.H., Wang P. Down-regulation of hepatic CYP1A2 plays an important role in inflammatory responses in sepsis. Crit Care Med. 2004;32:502–508. doi: 10.1097/01.CCM.0000109453.57709.E2. [DOI] [PubMed] [Google Scholar]
- 5.Zhou M., Maitra S.R., Wang P. The potential role of transcription factor aryl hydrocarbon receptor in downregulation of hepatic cytochrome P-450 during sepsis. Int J Mol Med. 2008;21:423–428. [PMC free article] [PubMed] [Google Scholar]
- 6.Wu R., Cui X., Dong W., Zhou M., Simms H.H., Wang P. Suppression of hepatocyte CYP1A2 expression by Kupffer cells via AhR pathway: the central role of proinflammatory cytokines. Int J Mol Med. 2006;18:339–346. [PubMed] [Google Scholar]
- 7.Lee S.H., Lee S.M. Suppression of hepatic cytochrome p450-mediated drug metabolism during the late stage of sepsis in rats. Shock. 2005;23:144–149. doi: 10.1097/01.shk.0000150778.39484.54. [DOI] [PubMed] [Google Scholar]
- 8.Baue A.E. Sepsis research: what did we do wrong? What would Semmelweis do today? Shock. 2001;16:1–8. doi: 10.1097/00024382-200116010-00001. [DOI] [PubMed] [Google Scholar]
- 9.Bernard G.R., Vincent J.L., Laterre P.F., LaRosa S.P., Dhainaut J.F., Lopez-Rodriguez A., Steingrub J.S., Garber G.E., Helterbrand J.D., Ely E.W., Fisher C.J., Jr Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med. 2001;344:699–709. doi: 10.1056/NEJM200103083441001. [DOI] [PubMed] [Google Scholar]
- 10.Martin G.S., Mannino D.M., Eaton S., Moss M. The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med. 2003;348:1546–1554. doi: 10.1056/NEJMoa022139. [DOI] [PubMed] [Google Scholar]
- 11.van den Berghe G., Wouters P., Weekers F., Verwaest C., Bruyninckx F., Schetz M., Vlasselaers D., Ferdinande P., Lauwers P., Bouillon R. Intensive insulin therapy in the critically ill patients. N Engl J Med. 2001;345:1359–1367. doi: 10.1056/NEJMoa011300. [DOI] [PubMed] [Google Scholar]
- 12.Hotchkiss R.S., Karl I.E. The pathophysiology and treatment of sepsis. N Engl J Med. 2003;348:138–150. doi: 10.1056/NEJMra021333. [DOI] [PubMed] [Google Scholar]
- 13.Oberholzer C., Oberholzer A., Clare-Salzler M., Moldawer L.L. Apoptosis in sepsis: a new target for therapeutic exploration. Faseb J. 2001;15:879–892. doi: 10.1096/fj.00-058rev. [DOI] [PubMed] [Google Scholar]
- 14.Tracey K.J. The inflammatory reflex. Nature. 2002;420:853–859. doi: 10.1038/nature01321. [DOI] [PubMed] [Google Scholar]
- 15.Riedemann N.C., Guo R.F., Ward P.A. Novel strategies for the treatment of sepsis. Nat Med. 2003;9:517–524. doi: 10.1038/nm0503-517. [DOI] [PubMed] [Google Scholar]
- 16.Angus D.C., Linde-Zwirble W.T., Lidicker J., Clermont G., Carcillo J., Pinsky M.R. Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med. 2001;29:1303–1310. doi: 10.1097/00003246-200107000-00002. [DOI] [PubMed] [Google Scholar]
- 17.West M.A., Heagy W. Endotoxin tolerance: A review. Crit Care Med. 2002;30:S64–S73. [PubMed] [Google Scholar]
- 18.Chaudry I.H. Sepsis: lessons learned in the last century and future directions. Arch Surg. 1999;134:922–929. doi: 10.1001/archsurg.134.9.922. [DOI] [PubMed] [Google Scholar]
- 19.Deitch E.A. Animal models of sepsis and shock: a review and lessons learned. Shock. 1998;9:1–11. doi: 10.1097/00024382-199801000-00001. [DOI] [PubMed] [Google Scholar]
- 20.Fink M.P., Heard S.O. Laboratory models of sepsis and septic shock. J Surg Res. 1990;49:186–196. doi: 10.1016/0022-4804(90)90260-9. [DOI] [PubMed] [Google Scholar]
- 21.Opal S.M. The clinical relevance of endotoxin in human sepsis: a critical analysis. J Endotoxin Res. 2002;8:473–476. doi: 10.1179/096805102125001109. [DOI] [PubMed] [Google Scholar]
- 22.Wang H., Yu M., Ochani M., Amella C.A., Tanovic M., Susarla S., Li J.H., Wang H., Yang H., Ulloa L., Al-Abed Y., Czura C.J., Tracey K.J. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature. 2003;421:384–388. doi: 10.1038/nature01339. [DOI] [PubMed] [Google Scholar]
- 23.Van Amersfoort E.S., Van Berkel T.J., Kuiper J. Receptors, mediators, and mechanisms involved in bacterial sepsis and septic shock. Clin Microbiol Rev. 2003;16:379–414. doi: 10.1128/CMR.16.3.379-414.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tinsley K.W., Grayson M.H., Swanson P.E., Drewry A.M., Chang K.C., Karl I.E., Hotchkiss R.S. Sepsis induces apoptosis and profound depletion of splenic interdigitating and follicular dendritic cells. J Immunol. 2003;171:909–914. doi: 10.4049/jimmunol.171.2.909. [DOI] [PubMed] [Google Scholar]
- 25.Remick D.G., Bolgos G.R., Siddiqui J., Shin J., Nemzek J.A. Six at six: interleukin-6 measured 6 h after the initiation of sepsis predicts mortality over 3 days. Shock. 2002;17:463–467. doi: 10.1097/00024382-200206000-00004. [DOI] [PubMed] [Google Scholar]
- 26.Zingarelli B., Sheehan M., Hake P.W., O'Connor M., Denenberg A., Cook J.A. Peroxisome proliferator activator receptor-gamma ligands, 15-deoxy-Delta(12,14)-prostaglandin J2 and ciglitazone, reduce systemic inflammation in polymicrobial sepsis by modulation of signal transduction pathways. J Immunol. 2003;171:6827–6837. doi: 10.4049/jimmunol.171.12.6827. [DOI] [PubMed] [Google Scholar]
- 27.Chung C.S., Song G.Y., Lomas J., Simms H.H., Chaudry I.H., Ayala A. Inhibition of Fas/Fas ligand signaling improves septic survival: differential effects on macrophage apoptotic and functional capacity. J Leukoc Biol. 2003;74:344–351. doi: 10.1189/jlb.0102006. [DOI] [PubMed] [Google Scholar]
- 28.Dong L.W., Wu L.L., Ji Y., Liu M.S. Impairment of the ryanodine-sensitive calcium release channels in the cardiac sarcoplasmic reticulum and its underlying mechanism during the hypodynamic phase of sepsis. Shock. 2001;16:33–39. doi: 10.1097/00024382-200116010-00007. [DOI] [PubMed] [Google Scholar]
- 29.Remick D.G., Call D.R., Ebong S.J., Newcomb D.E., Nybom P., Nemzek J.A., Bolgos G.E. Combination immunotherapy with soluble tumor necrosis factor receptors plus interleukin 1 receptor antagonist decreases sepsis mortality. Crit Care Med. 2001;29:473–481. doi: 10.1097/00003246-200103000-00001. [DOI] [PubMed] [Google Scholar]
- 30.Wang P., Chaudry I.H. Mechanism of hepatocellular dysfunction during hyperdynamic sepsis. Am J Physiol. 1996;270:R927–938. doi: 10.1152/ajpregu.1996.270.5.R927. [DOI] [PubMed] [Google Scholar]
- 31.Ayala A., Chung C.S., Lomas J.L., Song G.Y., Doughty L.A., Gregory S.H., Cioffi W.G., LeBlanc B.W., Reichner J., Simms H.H., Grutkoski P.S. Shock-induced neutrophil mediated priming for acute lung injury in mice: divergent effects of TLR-4 and TLR-4/FasL deficiency. Am J Pathol. 2002;161:2283–2294. doi: 10.1016/S0002-9440(10)64504-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Yang H., Ochani M., Li J., Qiang X., Tanovic M., Harris H.E., Susarla S.M., Ulloa L., Wang H., DiRaimo R., Czura C.J., Wang H., Roth J., Warren H.S., Fink M.P., Fenton M.J., Andersson U., Tracey K.J. Reversing established sepsis with antagonists of endogenous high-mobility group box 1. Proc Natl Acad Sci U S A. 2004;101:296–301. doi: 10.1073/pnas.2434651100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wichterman K.A., Baue A.E., Chaudry I.H. Sepsis and septic shock–a review of laboratory models and a proposal. J Surg Res. 1980;29:189–201. doi: 10.1016/0022-4804(80)90037-2. [DOI] [PubMed] [Google Scholar]
- 34.Yang S., Cioffi W.G., Bland K.I., Chaudry I.H., Wang P. Differential alterations in systemic and regional oxygen delivery and consumption during the early and late stages of sepsis. J Trauma. 1999;47:706–712. doi: 10.1097/00005373-199910000-00015. [DOI] [PubMed] [Google Scholar]
- 35.Yang S., Chung C.S., Ayala A., Chaudry I.H., Wang P. Differential alterations in cardiovascular responses during the progression of polymicrobial sepsis in the mouse. Shock. 2002;17:55–60. doi: 10.1097/00024382-200201000-00010. [DOI] [PubMed] [Google Scholar]
- 36.Chung C.S., Yang S., Song G.Y., Lomas J., Wang P., Simms H.H., Chaudry I.H., Ayala A. Inhibition of Fas signaling prevents hepatic injury and improves organ blood flow during sepsis. Surgery. 2001;130:339–345. doi: 10.1067/msy.2001.116540. [DOI] [PubMed] [Google Scholar]
- 37.Wang P., Ba Z.F., Chaudry I.H. Hepatocellular dysfunction occurs earlier than the onset of hyperdynamic circulation during sepsis. Shock. 1995;3:21–26. [PubMed] [Google Scholar]
- 38.Wang P., Ba Z.F., Chaudry I.H. Hepatic extraction of indocyanine green is depressed early in sepsis despite increased hepatic blood flow and cardiac output. Arch Surg. 1991;126:219–224. doi: 10.1001/archsurg.1991.01410260109015. [DOI] [PubMed] [Google Scholar]
- 39.Wang P., Zhou M., Rana M.W., Ba Z.F., Chaudry I.H. Differential alterations in microvascular perfusion in various organs during early and late sepsis. Am J Physiol. 1992;263:G38–43. doi: 10.1152/ajpgi.1992.263.1.G38. [DOI] [PubMed] [Google Scholar]
- 40.Wang P., Ba Z.F., Chaudry I.H. Mechanism of hepatocellular dysfunction during early sepsis. Key role of increased gene expression and release of proinflammatory cytokines tumor necrosis factor and interleukin-6. Arch Surg. 1997;132:364–369. doi: 10.1001/archsurg.1997.01430280038005. discussion 369–370. [DOI] [PubMed] [Google Scholar]
- 41.Yang S., Zhou M., Fowler D.E., Wang P. Mechanisms of the beneficial effect of adrenomedullin and adrenomedullin-binding protein-1 in sepsis: down-regulation of proinflammatory cytokines. Crit Care Med. 2002;30:2729–2735. doi: 10.1097/00003246-200212000-00018. [DOI] [PubMed] [Google Scholar]
- 42.Koo D.J., Chaudry I.H., Wang P. Kupffer cells are responsible for producing inflammatory cytokines and hepatocellular dysfunction during early sepsis. J Surg Res. 1999;83:151–157. doi: 10.1006/jsre.1999.5584. [DOI] [PubMed] [Google Scholar]
- 43.Wang P., Ayala A., Ba Z.F., Zhou M., Perrin M.M., Chaudry I.H. Tumor necrosis factor-alpha produces hepatocellular dysfunction despite normal cardiac output and hepatic microcirculation. Am J Physiol. 1993;265:G126–132. doi: 10.1152/ajpgi.1993.265.1.G126. [DOI] [PubMed] [Google Scholar]
- 44.Nelson D.R., Koymans L., Kamataki T., Stegeman J.J., Feyereisen R., Waxman D.J., Waterman M.R., Gotoh O., Coon M.J., Estabrook R.W., Gunsalus I.C., Nebert D.W. P450 superfamily: update on new sequences, gene mapping, accession numbers and nomenclature. Pharmacogenetics. 1996;6:1–42. doi: 10.1097/00008571-199602000-00002. [DOI] [PubMed] [Google Scholar]
- 45.Cheng P.Y., Morgan E.T. Hepatic cytochrome P450 regulation in disease states. Curr Drug Metab. 2001;2:165–183. doi: 10.2174/1389200013338676. [DOI] [PubMed] [Google Scholar]
- 46.Fuchs M. Bile acid regulation of hepatic physiology: III. Regulation of bile acid synthesis: past progress and future challenges. Am J Physiol Gastrointest Liver Physiol. 2003;284:G551–557. doi: 10.1152/ajpgi.00468.2002. [DOI] [PubMed] [Google Scholar]
- 47.Lee W.M. Drug-induced hepatotoxicity. N Engl J Med. 2003;349:474–485. doi: 10.1056/NEJMra021844. [DOI] [PubMed] [Google Scholar]
- 48.Negishi M., Uno T., Darden T.A., Sueyoshi T., Pedersen L.G. Structural flexibility and functional versatility of mammalian P450 enzymes. Faseb J. 1996;10:683–689. doi: 10.1096/fasebj.10.7.8635685. [DOI] [PubMed] [Google Scholar]
- 49.Peterson J.A., Graham S.E. A close family resemblance: the importance of structure in understanding cytochromes P450. Structure. 1998;6:1079–1085. doi: 10.1016/s0969-2126(98)00109-9. [DOI] [PubMed] [Google Scholar]
- 50.Rendic S., Di Carlo F.J. Human cytochrome P450 enzymes: a status report summarizing their reactions, substrates, inducers, and inhibitors. Drug Metab Rev. 1997;29:413–580. doi: 10.3109/03602539709037591. [DOI] [PubMed] [Google Scholar]
- 51.Rifkind A.B., Lee C., Chang T.K., Waxman D.J. Arachidonic acid metabolism by human cytochrome P450s 2C8, 2C9, 2E1, and 1A2: regioselective oxygenation and evidence for a role for CYP2C enzymes in arachidonic acid epoxygenation in human liver microsomes. Arch Biochem Biophys. 1995;320:380–389. doi: 10.1016/0003-9861(95)90023-3. [DOI] [PubMed] [Google Scholar]
- 52.Duester G. Involvement of alcohol dehydrogenase, short-chain dehydrogenase/reductase, aldehyde dehydrogenase, and cytochrome P450 in the control of retinoid signaling by activation of retinoic acid synthesis. Biochemistry. 1996;35:12221–12227. doi: 10.1021/bi961176+. [DOI] [PubMed] [Google Scholar]
- 53.Harder D.R., Campbell W.B., Roman R.J. Role of cytochrome P-450 enzymes and metabolites of arachidonic acid in the control of vascular tone. J Vasc Res. 1995;32:79–92. doi: 10.1159/000159080. [DOI] [PubMed] [Google Scholar]
- 54.Anzenbacher P., Anzenbacherova E. Cytochromes P450 and metabolism of xenobiotics. Cell Mol Life Sci. 2001;58:737–747. doi: 10.1007/PL00000897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.de Wildt S.N., Kearns G.L., Leeder J.S., van den Anker J.N. Cytochrome P450 3A: ontogeny and drug disposition. Clin Pharmacokinet. 1999;37:485–505. doi: 10.2165/00003088-199937060-00004. [DOI] [PubMed] [Google Scholar]
- 56.Spatzenegger M., Horsmans Y., Verbeeck R.K. Differential activities of CYP1A isozymes in hepatic and intestinal microsomes of control and 3-methylcholanthrene-induced rats. Pharmacol Toxicol. 2000;86:71–77. doi: 10.1034/j.1600-0773.2000.d01-14.x. [DOI] [PubMed] [Google Scholar]
- 57.Orlando R., Piccoli P., De Martin S., Padrini R., Floreani M., Palatini P. Cytochrome P450 1A2 is a major determinant of lidocaine metabolism in vivo: effects of liver function. Clin Pharmacol Ther. 2004;75:80–88. doi: 10.1016/j.clpt.2003.09.007. [DOI] [PubMed] [Google Scholar]
- 58.Morgan E.T. Regulation of cytochromes P450 during inflammation and infection. Drug Metab Rev. 1997;29:1129–1188. doi: 10.3109/03602539709002246. [DOI] [PubMed] [Google Scholar]
- 59.Piscitelli S.C., Reiss W.G., Figg W.D., Petros W.P. Pharmacokinetic studies with recombinant cytokines. Scientific issues and practical considerations. Clin Pharmacokinet. 1997;32:368–381. doi: 10.2165/00003088-199732050-00003. [DOI] [PubMed] [Google Scholar]
- 60.Carcillo J.A., Doughty L., Kofos D., Frye R.F., Kaplan S.S., Sasser H., Burckart G.J. Cytochrome P450 mediated-drug metabolism is reduced in children with sepsis-induced multiple organ failure. Intensive Care Med. 2003;29:980–984. doi: 10.1007/s00134-003-1758-3. [DOI] [PubMed] [Google Scholar]
- 61.McKindley D.S., Boulet J., Sachdeva K., Wang P., Chichester C. Endotoxic shock alters the pharmacokinetics of lidocaine and monoethylglycinexylidide. Shock. 2002;17:199–204. doi: 10.1097/00024382-200203000-00007. [DOI] [PubMed] [Google Scholar]
- 62.Oyekan A.O. The suppression by lipopolysaccharide of cytochrome P450-dependent renal vasodilation in the rat is mediated by nitric oxide. Eur J Pharmacol. 1995;277:123–132. doi: 10.1016/0014-2999(95)00044-l. [DOI] [PubMed] [Google Scholar]
- 63.Monshouwer M., McLellan R.A., Delaporte E., Witkamp R.F., van Miert A.S., Renton K.W. Differential effect of pentoxifylline on lipopolysaccharide-induced downregulation of cytochrome P450. Biochem Pharmacol. 1996;52:1195–1200. doi: 10.1016/0006-2952(96)00468-6. [DOI] [PubMed] [Google Scholar]
- 64.Iber H., Sewer M.B., Barclay T.B., Mitchell S.R., Li T., Morgan E.T. Modulation of drug metabolism in infectious and inflammatory diseases. Drug Metab Rev. 1999;31:29–41. doi: 10.1081/dmr-100101906. [DOI] [PubMed] [Google Scholar]
- 65.Shedlofsky S.I., Israel B.C., McClain C.J., Hill D.B., Blouin R.A. Endotoxin administration to humans inhibits hepatic cytochrome P450-mediated drug metabolism. J Clin Invest. 1994;94:2209–2214. doi: 10.1172/JCI117582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Morgan E.T. Regulation of cytochrome p450 by inflammatory mediators: why and how? Drug Metab Dispos. 2001;29:207–212. [PubMed] [Google Scholar]
- 67.Abdel-Razzak Z., Loyer P., Fautrel A., Gautier J.C., Corcos L., Turlin B., Beaune P., Guillouzo A. Cytokines down-regulate expression of major cytochrome P-450 enzymes in adult human hepatocytes in primary culture. Mol Pharmacol. 1993;44:707–715. [PubMed] [Google Scholar]
- 68.Muntane-Relat J., Ourlin J.C., Domergue J., Maurel P. Differential effects of cytokines on the inducible expression of CYP1A1, CYP1A2, and CYP3A4 in human hepatocytes in primary culture. Hepatology. 1995;22:1143–1153. [PubMed] [Google Scholar]
- 69.Morgan E.T. Suppression of constitutive cytochrome P-450 gene expression in livers of rats undergoing an acute phase response to endotoxin. Mol Pharmacol. 1989;36:699–707. [PubMed] [Google Scholar]
- 70.Bertini R., Bianchi M., Erroi A., Villa P., Ghezzi P. Dexamethasone modulation of in vivo effects of endotoxin, tumor necrosis factor, and interleukin-1 on liver cytochrome P-450, plasma fibrinogen, and serum iron. J Leukoc Biol. 1989;46:254–262. doi: 10.1002/jlb.46.3.254. [DOI] [PubMed] [Google Scholar]
- 71.Barker C.W., Fagan J.B., Pasco D.S. Interleukin-1 beta suppresses the induction of P4501A1 and P4501A2 mRNAs in isolated hepatocytes. J Biol Chem. 1992;267:8050–8055. [PubMed] [Google Scholar]
- 72.Sujita K., Okuno F., Tanaka Y., Hirano Y., Inamoto Y., Eto S., Arai M. Effect of interleukin 1 (IL-1) on the levels of cytochrome P-450 involving IL-1 receptor on the isolated hepatocytes of rat. Biochem Biophys Res Commun. 1990;168:1217–1222. doi: 10.1016/0006-291x(90)91158-o. [DOI] [PubMed] [Google Scholar]
- 73.Fukuda Y., Ishida N., Noguchi T., Kappas A., Sassa S. Interleukin-6 down regulates the expression of transcripts encoding cytochrome P450 IA1, IA2 and IIIA3 in human hepatoma cells. Biochem Biophys Res Commun. 1992;184:960–965. doi: 10.1016/0006-291x(92)90684-d. [DOI] [PubMed] [Google Scholar]
- 74.Godellas C.V., Williams J.F., Fabri P.J. Mixed-function oxidase activity in sepsis. J Surg Res. 1995;59:783–786. doi: 10.1006/jsre.1995.1240. [DOI] [PubMed] [Google Scholar]
- 75.Sachdeva K., Yan B., Chichester C.O. Lipopolysaccharide and cecal ligation/puncture differentially affect the subcellular distribution of the pregnane X receptor but consistently cause suppression of its target genes CYP3A. Shock. 2003;19:469–474. doi: 10.1097/01.shk.0000048903.46342.ec. [DOI] [PubMed] [Google Scholar]
- 76.Ertel W., Morrison M.H., Wang P., Ba Z.F., Ayala A., Chaudry I.H. The complex pattern of cytokines in sepsis. Association between prostaglandins, cachectin, and interleukins. Ann Surg. 1991;214:141–148. doi: 10.1097/00000658-199108000-00008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Nicholson T.E., Renton K.W. Role of cytokines in the lipopolysaccharide-evoked depression of cytochrome P450 in the brain and liver. Biochem Pharmacol. 2001;62:1709–1717. doi: 10.1016/s0006-2952(01)00859-0. [DOI] [PubMed] [Google Scholar]
- 78.Milosevic N., Schawalder H., Maier P. Kupffer cell-mediated differential down-regulation of cytochrome P450 metabolism in rat hepatocytes. Eur J Pharmacol. 1999;368:75–87. doi: 10.1016/s0014-2999(98)00988-1. [DOI] [PubMed] [Google Scholar]
- 79.Carcillo J.A., Korzekwa K.R., Jones G.S., Parise R.A., Gillespie D.G., Whalen M.J., Kochanek P.M., Branch R.A., Kost C.K., Jr The cytochrome P450 suicide inhibitor, 1-aminobenzotriazole, sensitizes rats to zymosan-induced toxicity. Res Commun Mol Pathol Pharmacol. 1998;102:57–68. [PubMed] [Google Scholar]
- 80.Harbrecht B.G., Frye R.F., Zenati M.S., Branch R.A., Peitzman A.B. Cytochrome P-450 activity is differentially altered in severely injured patients. Crit Care Med. 2005;33:541–546. doi: 10.1097/01.ccm.0000155989.54344.e0. [DOI] [PubMed] [Google Scholar]
- 81.Baker A.L., Kotake A.N., Schoeller D.A. Clinical utility of breath tests for the assessment of hepatic function. Semin Liver Dis. 1983;3:318–329. doi: 10.1055/s-2008-1040784. [DOI] [PubMed] [Google Scholar]
- 82.Honkakoski P., Negishi M. Regulation of cytochrome P450 (CYP) genes by nuclear receptors. Biochem J. 2000;347:321–337. doi: 10.1042/0264-6021:3470321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Waxman D.J. P450 gene induction by structurally diverse xenochemicals: central role of nuclear receptors CAR, PXR, and PPAR. Arch Biochem Biophys. 1999;369:11–23. doi: 10.1006/abbi.1999.1351. [DOI] [PubMed] [Google Scholar]
- 84.Greschik H., Moras D. Structure-activity relationship of nuclear receptor-ligand interactions. Curr Top Med Chem. 2003;3:1573–1599. doi: 10.2174/1568026033451736. [DOI] [PubMed] [Google Scholar]
- 85.Beato M., Herrlich P., Schutz G. Steroid hormone receptors: many actors in search of a plot. Cell. 1995;83:851–857. doi: 10.1016/0092-8674(95)90201-5. [DOI] [PubMed] [Google Scholar]
- 86.Mangelsdorf D.J., Thummel C., Beato M., Herrlich P., Schutz G., Umesono K., Blumberg B., Kastner P., Mark M., Chambon P., Evans R.M. The nuclear receptor superfamily: the second decade. Cell. 1995;83:835–839. doi: 10.1016/0092-8674(95)90199-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Forman B.M., Chen J., Evans R.M. The peroxisome proliferator-activated receptors: ligands and activators. Ann N Y Acad Sci. 1996;804:266–275. doi: 10.1111/j.1749-6632.1996.tb18621.x. [DOI] [PubMed] [Google Scholar]
- 88.Handschin C., Meyer U.A. Induction of drug metabolism: the role of nuclear receptors. Pharmacol Rev. 2003;55:649–673. doi: 10.1124/pr.55.4.2. [DOI] [PubMed] [Google Scholar]
- 89.Hankinson O. The aryl hydrocarbon receptor complex. Annu Rev Pharmacol Toxicol. 1995;35:307–340. doi: 10.1146/annurev.pa.35.040195.001515. [DOI] [PubMed] [Google Scholar]
- 90.Roberts E.A., Harper P.A., Wong J.M., Wang Y., Yang S. Failure of Ah receptor to mediate induction of cytochromes P450 in the CYP1 family in the human hepatoma line SK-Hep-1. Arch Biochem Biophys. 2000;384:190–198. doi: 10.1006/abbi.2000.2059. [DOI] [PubMed] [Google Scholar]
- 91.Gradin K., McGuire J., Wenger R.H., Kvietikova I., fhitelaw M.L., Toftgard R., Tora L., Gassmann M., Poellinger L. Functional interference between hypoxia and dioxin signal transduction pathways: competition for recruitment of the Arnt transcription factor. Mol Cell Biol. 1996;16:5221–5231. doi: 10.1128/mcb.16.10.5221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Tomita S., Sinal C.J., Yim S.H., Gonzalez F.J. Conditional disruption of the aryl hydrocarbon receptor nuclear translocator (Arnt) gene leads to loss of target gene induction by the aryl hydrocarbon receptor and hypoxia-inducible factor 1alpha. Mol Endocrinol. 2000;14:1674–1681. doi: 10.1210/mend.14.10.0533. [DOI] [PubMed] [Google Scholar]
- 93.Reyes H., Reisz-Porszasz S., Hankinson O. Identification of the Ah receptor nuclear translocator protein (Arnt) as a component of the DNA binding form of the Ah receptor. Science. 1992;256:1193–1195. doi: 10.1126/science.256.5060.1193. [DOI] [PubMed] [Google Scholar]
- 94.Whitelaw M., Pongratz I., Wilhelmsson A., Gustafsson J.A., Poellinger L. Ligand-dependent recruitment of the Arnt coregulator determines DNA recognition by the dioxin receptor. Mol Cell Biol. 1993;13:2504–2514. doi: 10.1128/mcb.13.4.2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Coumailleau P., Poellinger L., Gustafsson J.A., Whitelaw M.L. Definition of a minimal domain of the dioxin receptor that is associated with Hsp90 and maintains wild type ligand binding affinity and specificity. J Biol Chem. 1995;270:25291–25300. doi: 10.1074/jbc.270.42.25291. [DOI] [PubMed] [Google Scholar]
- 96.Whitelaw M.L., McGuire J., Picard D., Gustafsson J.A., Poellinger L. Heat shock protein hsp90 regulates dioxin receptor function in vivo. Proc Natl Acad Sci U S A. 1995;92:4437–4441. doi: 10.1073/pnas.92.10.4437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Carlson T.J., Billings R.E. Role of nitric oxide in the cytokine-mediated regulation of cytochrome P-450. Mol Pharmacol. 1996;49:796–801. [PubMed] [Google Scholar]
- 98.Shieh P., Zhou M., Ornan D.A., Chaudry I.H., Wang P. Upregulation of inducible nitric oxide synthase and nitric oxide occurs later than the onset of the hyperdynamic response during sepsis. Shock. 2000;13:325–329. doi: 10.1097/00024382-200004000-00012. [DOI] [PubMed] [Google Scholar]
- 99.Minamiyama Y., Takemura S., Imaoka S., Funae Y., Tanimoto Y., Inoue M. Irreversible inhibition of cytochrome P450 by nitric oxide. J Pharmacol Exp Ther. 1997;283:1479–1485. [PubMed] [Google Scholar]