throbber
Downregulation of Hepatic Cytochrome P450 in Chronic
`Renal Failure
`
`J Am Soc Nephrol 12: 326 –332, 2001
`
`FRANCOIS LEBLOND,* CARL GUÉVIN,* CHRISTIAN DEMERS,†
`ISABELLE PELLERIN,* MARIELLE GASCON-BARRÉ,† and VINCENT PICHETTE*
`*Service de Néphrologie et Centre de Recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, and †Centre
`Hospitalier Universitaire de Montréal, Faculté de Médecine, Université de Montréal, Québec, Canada.
`
`Abstract. Chronic renal failure (CRF) is associated with a
`decrease in drug metabolism. The mechanism remains poorly
`understood. The present study investigated the repercussions of
`CRF on liver cytochrome P450 (CYP450). Three groups of rats
`were defined: control, control paired-fed, and CRF. Total
`CYP450 activity, protein expression of several CYP450 iso-
`forms as well as their mRNA, and the in vitro N-demethylation
`of erythromycin were assessed in liver microsomes. The reg-
`ulation of liver CYP450 by dexamethasone and phenobarbital
`was assessed in CRF rats. Compared with control and control
`paired-fed rats, creatinine clearance was reduced by 60% (P ,
`0.01) in CRF rats. Weight was reduced by 30% (P , 0.01) in
`control paired-fed and CRF rats, compared with control ani-
`mals. There was no difference in the CYP450 parameters
`between control and control paired-fed. Compared with control
`
`paired-fed rats, total CYP450 was reduced by 47% (P , 0.001)
`in CRF rats. Protein expression of CYP2C11, CYP3A1, and
`CYP3A2 were considerably reduced (.40%, P , 0.001) in
`rats with CRF. The levels of CYP1A2, CYP2C6, CYP2D, and
`CYP2E1 were the same in the three groups. Northern blot
`analysis revealed a marked downregulation in gene expression
`of CYP2C11, 3A1, and 3A2 in CRF rats. Although liver
`CYP450 was reduced in CRF, its induction by dexamethasone
`and phenobarbital was present. N-demethylation of erythromy-
`cin was decreased by 50% in CRF rats compared with control
`(P , 0.001). In conclusion, CRF in rats is associated with a
`decrease in liver cytochrome P450 activity (mainly in
`CYP2C11, CYP3A1, and 3A2), secondary to reduced gene
`expression.
`
`Reduction in renal function alters the disposition of many
`drugs mainly by decreasing the elimination of those excreted
`by the kidney (1,2). However, drug metabolism by the liver
`may also be altered in patients with chronic renal failure (CRF)
`(3). Indeed, several studies have shown that the metabolic
`clearance of various substrates is reduced in patients with CRF
`(1,3,4). The severity of the inhibition of drug metabolism is
`variable (from 17 to 85%), depending on the metabolic path-
`way involved, but drugs metabolized by hepatic cytochrome
`P450 seem particularly vulnerable (3,5). Supporting the hy-
`pothesis that CRF inhibits liver P450 is the reduction in hepatic
`P450 as well as other cytosolic enzymes in rats with experi-
`mental renal failure (6 –9).
`Rat hepatic cytochrome P450 is composed of several iso-
`forms. Those involved in drug metabolism processes include
`CYP1A2, CYP2C11, CYP2D, CYP2E1, and CYP3A1/3A2
`(10). The knowledge of which isoforms are reduced by CRF is
`critical to predict which drugs are at risk for accumulation.
`Although previous work has focused on the repercussions of
`
`CRF on total hepatic P450 content, only one has studied
`whether the reduction in P450 involves all isoforms. Uchida et
`al. (11) demonstrated that in CRF rats, protein expression of
`some liver P450 isoforms were decreased (CYP2C6, 2C11, and
`3A2) while another was increased (CYP1A1). However, the
`relation between reduced protein expression in P450 isoforms
`and their metabolic activities has not been studied. Further-
`more, the mechanism of liver P450 reduction remains poorly
`understood; it may be secondary to a decrease in synthesis or
`an increase in degradation.
`The objectives of this study were to determine the effects of
`CRF on hepatic P450 and to define the mechanisms leading to
`its downregulation. For this purpose, in control, control paired-
`fed, and CRF rats, we measured the following: (1) liver cyto-
`chrome P450 total activity; (2) the main P450 isoforms in-
`volved in drug metabolism, e.g., CYP1A1/1A2, 2C6, 2C11,
`2D1, 2E1, 3A1, and 3A2, as well as some of their specific
`metabolic activities; and (3) the mRNA encoding for these
`specific isoforms. Finally, the effect of CRF on liver cyto-
`chrome P450 was also studied in rats treated with known
`inducers of P450, dexamethasone, and phenobarbital (12).
`
`Received February 2, 2000. Accepted June 21, 2000.
`Correspondence to Dr. Vincent Pichette, Centre de Recherche Guy-Bernier,
`Hôpital Maisonneuve-Rosemont, 5415 boul. de l’Assomption, Montréal, Qué-
`bec H1T 2M4, Canada. Phone: 514-252-3489; Fax: 514-255-3026; E-mail:
`vincent.pichette@hmr.qc.ca
`1046-6673/1202-0326
`Journal of the American Society of Nephrology
`Copyright © 2001 by the American Society of Nephrology
`
`Materials and Methods
`Experimental Model
`Male Sprague-Dawley rats (Charles River, Saint-Charles, Québec,
`Canada), weighing 200 to 300 g, were housed in the Research Center
`animal care facility and maintained on Purina rat pellets (Ralston-
`Purina, St. Louis, MO) and water ad libitum. The animals were
`
`Boehringer Ex. 2031
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 1
`
`

`
`J Am Soc Nephrol 12: 326 –332, 2001
`
`Decreased Cytochrome P450 in Renal Failure
`
`327
`
`allowed an acclimatization period of at least 3 d before any experi-
`mental work was undertaken. All of the experiments were conducted
`according to the Canadian Council on Animal Care guidelines for care
`and use of laboratory animals.
`
`Experimental Protocol
`Studies were performed in three groups of 10 animals each: control,
`control paired-fed, and CRF. To evaluate further the effects of CRF on
`the regulation of liver P450, three other groups (n 5 6 in each group)
`were studied: CRF, CRF treated with dexamethasone, or CRF treated
`with phenobarbital. Dexamethasone and phenobarbital are potent in-
`ducers of liver P450 (12).
`Chronic renal failure was induced by a two-staged, five-sixths
`nephrectomy. Briefly, the rats underwent a two-thirds nephrectomy of
`the left kidney through a midline incision, and 7 d later, the right
`nephrectomy was done. Rats from both control groups underwent two
`sham laparotomies. Pentobarbital was used for anesthesia (60 mg/kg
`via intraperitoneal injection). After surgery, CRF animals were fed
`Purina rat chow and water ad libitum. Control rats were fed ad
`libitum. Control paired-fed rats were fed the same amount of rat chow
`that was ingested by the CRF rats on the previous day to assess the
`effect of CRF-induced malnutrition. Body weight was measured every
`other day for the duration of the study. At day 41 after the nephrec-
`tomy, the rats were housed in metabolic cages and urine was collected
`for 24 h to determine the clearance of creatinine. Rats were killed by
`decapitation at 42 d after nephrectomy. Blood was collected for the
`measurement of serum creatinine and urea. Enzyme induction of liver
`P450 was achieved using an intraperitoneal injection of dexametha-
`sone (100 mg/kg per d) or phenobarbital (80 mg/kg) on days 38 to 41
`(12).
`
`Preparation of Liver Microsomes
`The rat livers were immediately excised after death, and micro-
`somes were isolated by differential centrifugation (13). Samples were
`maintained at 4°C during microsome preparation. Briefly, 5 g ofliver
`was homogenized, using a Potter-Elvehjem tissue grinder (Wheaton
`Science Products, Millville, NJ), in 25 ml of 0.25 M sucrose and was
`centrifuged at 12,000 3 g. To the 12,000 3 g supernatant, 1 M CaCl2
`was added (10% vol/vol) and further centrifuged at 27,000 3 g. The
`pellet containing the microsomes was stored at 280°C in Tris 0.1 M
`(pH 7.4), glycerol 20%, ethylenediaminetetraacetate (EDTA) 10 mM
`up to analysis.
`
`Determination of Total Cytochrome P450 Activity
`Microsomal protein content was determined by the method of
`Lowry et al. (14), using bovine serum albumin as standard protein.
`Total cytochrome P450 activity was measured from the difference
`spectrum of the reduced protein according to a previously published
`method (15).
`
`Western Blot Analysis
`The major cytochrome P450 isoforms implicated in the metabolism
`of drugs were assessed by Western blot analysis: CYP1A2, CYP2C6,
`CYP2C11, CYP2D, CYP2E1, CYP3A1, and CYP3A2. Forty mg of
`protein was electrophoresed in a 7.5% polyacrylamide gel containing
`0.1% sodium dodecyl sulfate (SDS), and separated proteins were
`electrophoretically transferred on nitrocellulose (16,17). Immunoblots
`for respective isoforms were performed in 5% low-fat milk in phos-
`phate-buffered saline and washed with 0.1% Tween 20 in phosphate-
`buffered saline. CYP1A2 was detected using a polyclonal goat anti-rat
`1A2 (Gentest Corporation, Woburn, MA). CYP2C6 and CYP2C11
`
`were detected using a goat anti-rat 2C6 and 2C11 (Gentest), respec-
`tively. CYP2D was detected using a rabbit anti-human 2D (Oxford
`Biochemical Research Inc., Oxford, MI). CYP2E1 and CYP3A1 were
`detected using a monoclonal mouse anti-rat 2E1 and 3A1 (Oxford),
`respectively. CYP3A2 was detected using a goat anti-rat 3A2 (Gen-
`test). Immune complexes were revealed by secondary antibody (swine
`anti-goat IgG and goat anti-rabbit IgG [Biosource International, Cam-
`marillo, CA], as well as goat anti-mouse IgG [Sigma Chemicals, St.
`Louis, MO]) coupled to peroxidase and the Luminol derivative of
`Lumi-Light Western blotting substrate (Roche Diagnostics, Laval,
`Québec, Canada). Immune reaction intensity was determined by com-
`puter-assisted densitometry on exposed Biomax MR film (Scientific
`Imaging Systems, Eastman Kodak Co., Rochester, NY).
`
`mRNA Analysis
`At the time of death, biopsies of liver were rinsed in ice-cold saline
`and flash-frozen in liquid nitrogen. Samples were kept at 280°C until
`RNA extraction. The RNA encoding for CYP1A2, CYP2C11,
`CYP3A1, and CYP3A2 was evaluated by Northern blot analysis.
`Total RNA was extracted from frozen tissue by the RNeasy kit
`(Qiagen, Mississauga, Ontario, Canada). RNA concentrations were
`determined by measuring absorbance at a wavelength of 260 nm.
`Total RNA samples were denatured by heating at 65°C in buffer
`containing 42% deionized formamide, 30 mM 4-morpholinepropane-
`sulfonic acid, and 8.5% formaldehyde. RNAs (30 mg total RNA) were
`separated by electrophoresis in 1% agarose-1.7% formaldehyde gel
`submerged in buffer (pH 7.2), containing 20 mM 4-morpholinepro-
`panesulfonic acid, 8 mM sodium acetate, and 1 mM EDTA. Separate
`RNA were transferred to nylon membranes (Qiabrane, Qiagen), using
`the standard capillary technique with 10 3 SSC (1.5 M NaCl and 0.15
`M sodium citrate, pH 7.0) and fixed under ultraviolet lamp at 0.6
`J/cm2. Prehybridizations were performed at 52°C in buffer composed
`of 0.5 M NaPO4 (pH 7.2), 7% SDS, 1% bovine serum albumin, 1%
`dextran sulfate, 1 mM EDTA, and 250 mg/ml denatured herring sperm
`DNA (Roche Diagnostics). The blots were hybridized with oligonu-
`cleotide probes specific for each P450 mRNA: for CYP1A2, a syn-
`thetic 20-mer oligonucleotide complementary to bases 1580 to 1599
`of the P450 1A2 cDNA sequence (18); for CYP2C11, a 30-mer
`oligonucleotide corresponding to the complement of nucleotides 945
`to 974 of the coding sequences of P450 2C11 (19); for CYP3A1, a
`synthetic 32-mer oligonucleotide complementary to bases 1593 to
`1624 of the P450PCN1 nucleotide sequence (20); for CYP3A2, a
`synthetic 24-mer oligonucleotide complementary to bases 1652 to
`1675 of the 6b-A nucleotide sequence (21); for 18S ribosomal RNA,
`a 1.5-kb human cDNA insert from EcoRI site of the pBluescript
`SK-vector ATCC no.77242 (MBI Fermentas). Oligonucleotides were
`prepared by the Sheldon Biotechnology Center (McGill University,
`Montreal, Québec, Canada). They were end labeled with [g-32P]ATP
`(3000 Ci/mmol), using T4 polynucleotide kinase (BRL, Burlington,
`Ontario, Canada). The 18S cDNA probe was
`labeled using
`[a-32P]dCTP (3000 Ci/mmol) and Klenow according to the random
`oligo-priming method. Hybridization was then performed at 52°C for
`24 h in prehybridization buffer to which were added the labeled
`probes. The membranes were then washed in 0.2 M NaPO4 (pH 7.2),
`1% SDS, 1 mM EDTA at room temperature and at 52°C. Washed
`membranes were exposed to autoradiography film (Biomax MS;
`Kodak) with Biomax TranScreen-HE intensifying screen (Kodak) at
`280°C for 3 to 10 d. Hybridization signals were quantified by
`computer-assisted densitometer. mRNA levels were expressed as ar-
`bitrary densitometric units and standardized by comparison with hy-
`bridization results obtained with 18S ribosomal RNA probe.
`
`Boehringer Ex. 2031
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 2
`
`

`
`328
`
`Journal of the American Society of Nephrology
`
`J Am Soc Nephrol 12: 326 –332, 2001
`
`In Vitro Metabolism of Erythromycin
`To evaluate the metabolic activity of CYP3A2 in liver microsomes,
`erythromycin N-demethylation was determined as described by Wang
`et al. (22). Erythromycin (250 mM) (Sigma Chemicals) was incubated
`with 2.0 mg of rat liver microsomes (either from control paired-fed or
`CRF) at 37°C for 15 min in the presence of an NADPH-generating
`system consisting of the following: 10 mM glucose 6-phosphate, 1
`mM NADP, and 0.35 units glucose-6-phosphate dehydrogenase (Sig-
`ma Chemicals) in a total volume of 0.5 ml. Reactions were quenched
`with 0.05 ml of 25% ZnSO4 and 0.05 ml of 0.3N Ba(OH)2. The
`samples were then centrifuged at 14,000 3 g for 10 min, and 0.35 ml
`of the supernatant was transferred and mixed with 0.15 ml of Nash
`reagent (23). The mixture was incubated at 56°C for 30 min, and
`samples were analyzed by spectrophotometry (absorbance 405 nm) to
`determine the formation of formaldehyde (23).
`
`Other Assays
`Blood and urine chemistries were determined with a Hitachi 717
`autoanalyser
`(Boehringer Mannheim Canada, Laval, Québec,
`Canada).
`
`Statistical Analyses
`The results are expressed as mean 6 SEM. Differences between
`groups were assessed by using an unpaired t test or an ANOVA test.
`Significant ANOVA was followed by Fisher least significant differ-
`ence multiple comparisons procedure. The threshold of significance
`was P , 0.05
`
`Results
`Biochemical Parameters and Body Weight in Control,
`Control Paired-Fed, and CRF Rats
`Table 1 presents the biochemicals and body weight of the
`three groups of animals studied. Compared with control and
`control paired-fed animals, CRF rats had higher levels of
`plasma creatinine and urea and lower values of creatinine
`clearance (reduced by 60%; P , 0.001). Body weight in
`control paired-fed and CRF rats was reduced by 30% compared
`with control animals. However, there was no difference in
`body weight between control paired-fed and CRF rats.
`
`Liver Total Cytochrome P450 Activity in Control,
`Control Paired-Fed, and CRF Rats
`No difference was observed in total cytochrome P450 ac-
`tivity between control and control paired rats (0.61 6 0.01 and
`0.59 6 0.04 nmol/mg protein; Table 1). Thus, malnutrition as
`
`produced by CRF has no effect on liver P450. In CRF rats, total
`cytochrome P450 activity was significantly reduced by 47%
`compared with both control groups (Table 1). The P450 activ-
`ity was negatively correlated with creatinine clearance (r 5
`0.68, P , 0.001). Similar correlations were found between
`P450 activity and blood urea and creatinine.
`
`Protein Expression of Liver Cytochrome P450 Isoforms
`in Control, Control Paired-Fed, and CRF Rats
`No differences in the different isoforms between control and
`control paired-fed rats were observed (data not shown). The
`levels of CYP2C11, 3A1, and 3A2 in CRF rats were reduced
`by 40, 75, and 65%, respectively, in CRF rats compared with
`control paired-fed animals (P , 0.001; Figure 1). Conversely,
`the levels of CYP1A2, 2C6, 2D1, and 2E1 were not modified
`in CRF rats compared with control rats.
`
`mRNA Encoding Liver Cytochrome P450 Isoforms in
`Control, Control Paired-Fed, and CRF Rats
`To determine whether liver cytochrome P450 isoforms in
`CRF were downregulated secondary to a decrease in their
`synthesis or an increase in their degradation, we evaluated
`mRNA encoding the different isoforms by Northern blot anal-
`ysis. Again, there was no difference in the level of mRNA
`between control and control paired-fed rats (data not shown).
`However,
`a
`significant decrease
`in mRNA encoding
`CYP2C11, 3A1, and 3A2 isoforms was observed in CRF
`compared with control paired-fed animals (Figure 2). Thus, the
`decrease in protein expression of the different isoforms of P450
`observed in CRF is secondary to reduced gene expression.
`
`In Vitro Metabolism of Erythromycin in Control,
`Control Paired-Fed, and CRF Rats
`To determine the repercussion of cytochrome P450 reduction in
`CRF on the metabolism of drugs, we assessed the in vitro N-
`demethylation of erythromycin in liver. This enzymatic reaction is
`mediated primarily by the CYP3A family. No differences be-
`tween control and control paired-fed rats were observed (data not
`shown). The N-demethylation of erythromycin was decreased by
`more than 50% in rats with CRF, compared with control paired-
`fed animals (P , 0.001; Figure 3).
`
`Table 1. Characteristics of the control, control paired-fed and CRF ratsa
`
`Control
`
`Control Paired-Fed
`
`CRF
`
`Body weight (g)
`Serum creatinine (mmol/L)
`Creatinine clearance (ml/100 g of body weight/min)
`Serum urea (mmol/L)
`Liver total cytochrome P450 activity (nmol/mg of proteins)
`
`459 6 3.7
`52 6 1
`406 6 18
`4.8 6 0.2
`0.61 6 0.01
`
`318 6 9.4b
`56 6 2
`390 6 17
`5.9 6 0.5
`0.59 6 0.04
`
`327 6 8.5b
`141 6 9b,c
`178 6 25b,c
`24.5 6 5.8b,c
`0.34 6 0.04b,c
`
`a CRF, chronic renal failure. Data are the mean 6 SEM. Measurements were made 41 d after the first surgery.
`b P , 0.01 compared with control rats.
`c P , 0.01 compared with control paired-fed animals.
`
`Boehringer Ex. 2031
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 3
`
`

`
`J Am Soc Nephrol 12: 326 –332, 2001
`
`Decreased Cytochrome P450 in Renal Failure
`
`329
`
`Figure 1. Protein expression of liver cytochrome P450 isoforms in control paired-fed (e) and chronic renal failure (CRF) rats (f). Protein bands
`are expressed in densitometry units (%). The densitometry units of control paired-fed rats were arbitrarily defined as 100%. Data are the mean
`6 SEM of 10 rats in each group. *, P , 0.001 as compared with control paired-fed rats. Representative blots are also shown.
`
`Figure 2. mRNA encoding liver cytochrome P450 isoforms (CYP1A2, CYP2C11, CYP3A1, and 3A2) in control paired-fed (e) and CRF rats
`(f). mRNA bands are expressed in standardized densitometry units (%). The densitometry values for cytochrome P450 isoforms were
`standardized by dividing these values by the values for 18S ribosomal RNA. The standardized densitometry units of control paired-fed rats were
`arbitrarily defined as 100%. Data are the mean 6 SEM of six rats in each group. *, P , 0.01 as compared with control paired-fed rats.
`Representative blots are also shown.
`
`Induction of Cytochrome P450 in Rats with CRF by
`Dexamethasone or Phenobarbital
`Although our results showed that the decrease in liver cy-
`tochrome P450 in CRF is secondary to reduced gene expres-
`sion, we were also interested to know whether P450 was still
`inducible, despite its inhibition by CRF. We studied the effect
`of dexamethasone and phenobarbital (which are potent induc-
`ers of the CYP3A family) in CRF rats on CYP3A1 and 3A2
`protein expressions as well as on their mRNA levels (Figure 4).
`In CRF rats, CYP3A1 and 3A2 were greatly enhanced by
`dexamethasone and phenobarbital. This was secondary to an
`upregulation of mRNA encoding these proteins. We further
`studied the effect of dexamethasone on N-demethylation of
`
`erythromycin. In CRF rats, N-demethylation of erythromycin
`was also increased by dexamethasone (0.28 6 0.02 versus 1.38
`6 0.07 nmol/mg of protein/min, P , 0.001).
`
`Discussion
`This study demonstrates that in the rat, CRF induces a
`marked decrease in liver total cytochrome P450 activity sec-
`ondary to reduced protein expression of selective cytochrome
`P450 isoforms, namely CYP2C11, 3A1, and 3A2. The mech-
`anism underlying this downregulation is a reduction in the
`mRNA levels encoding these proteins. The repercussions on
`the metabolism of drugs by the liver are important in that we
`observed a 50% reduction of erythromycin biotransformation
`
`Boehringer Ex. 2031
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 4
`
`

`
`330
`
`Journal of the American Society of Nephrology
`
`J Am Soc Nephrol 12: 326 –332, 2001
`
`Our results demonstrate that there is an association between
`lower levels of mRNA and protein for some isoforms of
`cytochrome P450, namely CYP2C11, 3A1, and 3A2 (Figure
`2). This suggests that there is reduced gene expression. The
`mechanisms responsible for the diminished liver CYP2C11,
`3A1, and 3A2 gene expression in CRF are not known. Caloric
`restriction, as seen in CRF rats, downregulates hepatic genes of
`drug metabolizing enzymes in the mouse and in the rat (25,26).
`However, in our control paired-fed rats, we did not observe any
`modification in cytochrome P450 levels despite a similar
`weight loss as in CRF rats. Further studies will require evalu-
`ation of uremia on the function of the CYP450 promoters.
`A number of studies indicate that animals with CRF display
`impaired protein synthesis, by reduced gene expression, in the
`liver as well as in the skeletal muscle (27,28). For instance, the
`mRNA of hepatic lipase and insulin-like growth factor 1 re-
`ceptor are decreased in hepatocytes and skeletal muscle, re-
`spectively, of rats with CRF (27,28). CRF is associated with
`sustained elevations in calcium in many cell types, including
`the hepatocytes as well as the skeletal muscle, and this high
`intracellular calcium seems to be a major factor underlying cell
`dysfunction and reduced protein synthesis in CRF (29,30).
`However, the effects of intracellular calcium on the regulation
`of P450 remain poorly defined (31,32). Whether increased
`intracellular calcium is implicated in the downregulation of
`liver P450 in CRF remains to be studied.
`In the present study, downregulation of cytochrome P450 in
`CRF was overcome by dexamethasone and also by phenobar-
`bital, which are potent inducers of CYP3A isoforms (Figure 4).
`This result suggests that although liver cytochrome P450 is
`decreased in CRF, its dexamethasone and phenobarbital regu-
`lation is still present. The clinical significance of this finding
`remains to be defined. However, one can anticipate that in
`CRF, liver cytochrome P450 could be modulated by known
`P450 inducers, e.g., steroids, phenobarbital.
`Several investigators have demonstrated that in patients with
`renal failure, there is also a decrease in the metabolic clearance
`of many drugs (ranging from 17 to 85%) (1,3,4). The vast
`majority of these drugs are metabolized by the liver through the
`cytochrome P450 pathway. Unfortunately, we are not aware of
`human studies demonstrating a reduction in liver cytochrome
`P450 isoforms in CRF. However, several methods have been
`described to assess in vivo the CYP activity in humans, and the
`most widely used is the administration of probe drugs that are
`selectively metabolized by a specific CYP isoform. Kevorkian
`et al. (33) reported that assessment of CYP2D6 activity by the
`use of dextromethorphan or sparteine was possible in patients
`with CRF . The results of their study revealed that there was a
`decrease in the metabolic clearance of sparteine suggesting a
`decrease in CYP2D6 activity in CRF patients. There seems to
`be a correlation between the decrease in the metabolism of
`drugs and the severity of renal failure in humans (34). Inter-
`estingly, in the rat, we found a significant correlation between
`the decrease in renal function and the reduction in liver cyto-
`chrome P450 and also with the reduction in the N-demethyl-
`ation of erythromycin. These results suggest
`that as CRF
`
`Figure 3. In vitro metabolism of erythromycin (N-demethylation) in
`control paired-fed (e) and CRF rats (f). Incubations were performed
`with 2 mg of rat liver microsome at 37°C for 15 min. Data are the
`mean 6 SEM of six rats in each group. *, P , 0.001 as compared with
`control paired-fed rats.
`
`mediated by the CYP3A family. Although liver cytochrome
`P450 is decreased in CRF, its dexamethasone and phenobar-
`bital regulation was still present as shown by the induction of
`CYP3A by dexamethasone or phenobarbital.
`Renal failure has been generally thought to decrease only the
`renal clearance of drugs (24). However, several studies have
`demonstrated that animals with CRF also present decreased
`hepatic drug metabolism (5). Because the P450 is the major
`enzymatic system involved in drug metabolism, most studies
`have focused on liver P450. The results of these studies show
`that in CRF rats there is a 18.6 to 42.8% decrease in liver total
`P450 (6 –9,11). Furthermore, important reductions in enzy-
`matic reactions normally carried by the liver P450 have been
`reported: N-demethylation of aminopyrine and ethylmorphine,
`O-demethylation of codeine, and hydroxylation of aniline (5).
`In the present study, we found a 47% reduction in total P450
`activity as well as a significant reduction in the N-demethyl-
`ation of erythromycin.
`Few studies have focused on the specific P450 isoforms
`reduced in CRF (11). Knowledge of which isoform is reduced
`by CRF is critical to predict which drugs are at risk for
`accumulation when used in CRF. Recently, Uchida et al. (11)
`reported a reduction in the levels of hepatic CYP2C6,
`CYP2C11, and CYP3A2 and a slight increase in CYP1A2 in
`rats with CRF. Our results demonstrate that only CYP2C11,
`CYP3A1, and CYP3A2 are significantly reduced, while no
`isoform induction was noted. Furthermore, the level of reduc-
`tion was far more important in the present study. This could
`reflect a more pronounced degree of uremia obtained in our
`rats (50% increase in plasma creatinine versus 65% in the
`present study) and also a longer period of uremia (21 d versus
`42 d in the present study). Interestingly, CYP3A1 and 3A2 in
`the rat correspond to CYP3A4 in humans. Because this isoform
`is responsible for the metabolism of several drugs commonly
`used in CRF patients, patients with CRF could be at risk for
`drug accumulation and toxicity.
`
`Boehringer Ex. 2031
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 5
`
`

`
`J Am Soc Nephrol 12: 326 –332, 2001
`
`Decreased Cytochrome P450 in Renal Failure
`
`331
`
`Figure 4. Changes in liver microsome levels of CYP3A2 protein expression and specific mRNA in response to treatment with dexamethasone
`(100 mg/kg intraperitoneally daily during 4 d) (f) or with phenobarbital (80 mg/kg intraperitoneally daily during 4 d; &U25A8;). Data are the
`mean 6 SEM of six rats in each group. *, P , 0.001 as compared with CRF without inducer treatment (e). Representative blots are also shown.
`
`worsened, patients were at risk of drug accumulation and
`toxicity, secondary to reduction in their metabolism.
`In conclusion, CRF is associated with a decrease in liver
`cytochrome P450 isoforms
`in rats
`(mainly CYP2C11,
`CYP3A1, and 3A2), secondary to reduced mRNA levels. Drug
`metabolism activity, assessed by the N-demethylation of eryth-
`romycin, is also greatly depressed in CRF rats. Liver cyto-
`chrome P450 downregulation is correlated with the degree of
`renal failure. This decrease could explain the reduction in drug
`metabolism observed in CRF patients because these isoforms
`(especially CYP3A1 and 3A2) correspond to CYP3A4 in hu-
`mans, which is responsible for the metabolism of several drugs
`commonly used in CRF patients.
`
`Acknowledgments
`This work was supported by the Kidney Foundation of Canada and
`Fonds de la Recherche en Santé du Québec. Part of this work has been
`presented at the 31st and 32nd annual meetings of the American
`Society of Nephrology in Philadelphia and Miami, respectively. Vin-
`cent Pichette is a scholar of the Kidney Foundation of Canada.
`
`References
`1. Matzke GR, Frye RF: Drug administration in patients with renal
`insufficiency. Minimizing renal and extrarenal toxicity. Drug Saf
`16: 205–231, 1997
`2. Lam YW, Banerji S, Hatfield C, Talbert RL: Principles of drug
`administration in renal insufficiency. Clin Pharmacokinet 32:
`30 –57, 1997
`3. Touchette MA, Slaughter RL: The effect of renal failure on
`hepatic drug clearance. DICP 25: 1214 –24, 1991
`4. Talbert RL: Drug dosing in renal insufficiency. J Clin Pharmacol
`34: 99 –110, 1994
`
`5. Gibson TP: Renal disease and drug metabolism: An overview.
`Am J Kidney Dis 8: 7–17, 1986
`6. Leber HW, Schutterle G: Oxidative drug metabolism in liver
`microsomes from uremic rats. Kidney Int 2: 152–158, 1972
`7. Van Peer AP, Belpaire FM: Hepatic oxidative drug metabolism
`in rats with experimental renal failure. Arch Int Pharmacodyn
`Ther 228: 180 –183, 1977
`8. Leber HW, Gleumes L, Schutterle G: Enzyme induction in the
`uremic liver. Kidney Int Suppl 8: S43–S48, 1978
`9. Patterson SE, Cohn VH: Hepatic drug metabolism in rats with
`experimental chronic renal failure. Biochem Pharmacol 33: 711–
`716, 1984
`10. Wrighton SA, Stevens JC: The human hepatic cytochromes P450
`involved in drug metabolism. Crit Rev Toxicol 22: 1–21,
`1992
`11. Uchida N, Kurata N, Shimada K, Nishimura Y, Yasuda K,
`Hashimoto M, Uchida E, Yasuhara H: Changes of hepatic mi-
`crosomal oxidative drug metabolizing enzymes in chronic renal
`failure (CRF) rats by partial nephrectomy. Jpn J Pharmacol 68:
`431– 439, 1995
`12. Correia MA: Rat and human liver cytochrome P450: Substrate
`and inhibitor specificities and functional markers. In: Cyto-
`chrome P450: Structure, Mechanism, and Biochemistry, 2nd Ed.,
`edited by Montellano OD, New York, Plenum Press, 1995, pp
`607– 630
`13. Cinti DL, Moldeus P, Schenkman JB: Kinetic parameters of
`drug-metabolizing enzymes in Ca21-sedimented microsomes
`from rat liver. Biochem Pharmacol 21: 3249 –3256, 1972
`14. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ: Protein mea-
`surement with the folin phenol reagent. J Biol Chem 193: 265–
`275, 1951
`15. Omura T, Sato R: The carbon monoxide-binding pigment of liver
`microsomes. J Biol Chem 239: 2370 –2378, 1964
`
`Boehringer Ex. 2031
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 6
`
`

`
`332
`
`Journal of the American Society of Nephrology
`
`J Am Soc Nephrol 12: 326 –332, 2001
`
`16. Laemmli UK: Cleavage of structural proteins during the assem-
`bly of the head of bacteriophage T4. Nature 227: 680 – 685, 1970
`17. Towbin H, Staehlin T, Gordon J: Electrophoretic transfer of
`proteins from polyacrylamide gels to nitrocellulose sheets. Proc
`Natl Acad Sci USA 76: 4350 – 4354, 1979
`18. Kim H, Reddy S, Novak RF: 3-Methylcholanthrene and pyridine
`effects on CYP1A1 and CYP1A2 expression in rat renal tissue.
`Drug Metab Dispos 23: 818 – 824, 1995
`19. Chen J, Murray M, Liddle C, Jiang XM, Farrell GC: Downregu-
`lation of male-specific cytochrome P450s 2C11 and 3A2 in bile
`duct-ligated male rats: Importance to reduced hepatic content of
`cytochrome P450 in cholestasis. Hepatology 22: 580 –587, 1995
`20. Gonzalez FJ, Song BJ, Hardwick JP: Pregnenolone 16 alpha-
`carbonitrile-inducible P-450 gene family: Gene conversion and
`differential regulation. Mol Cell Biol 6: 2969 –2976, 1986
`21. Miyata M, Nagata K, Shimada M, Yamazoe Y, Kato R: Structure of
`a gene and cDNA of a major constitutive form of testosterone 6
`beta-hydroxylase (P450/6 beta A) encoding CYP3A2: Comparison
`of the cDNA with P450PCN2. Arch Biochem Biophys 314: 351–
`359, 1994
`22. Wang RW, Newton DJ, Scheri TD, Lu AY: Human cytochrome
`P450 3A4-catalyzed testosterone 6 beta-hydroxylation and eryth-
`romycin N-demethylation. Competition during catalysis. Drug
`Metab Dispos 25: 502–507, 1997
`23. Nash T: The calorimetric estimation of formaldehyde by means
`of the Hantzsh reaction. Biochem J 55: 416 – 421, 1953
`24. Cantu TG, Ellerbeck EF, Yun SW, Castine SD, Kornhauser DM:
`Drug prescribing for patients with changing renal function. Am
`J Hosp Pharm 49: 2944 –2948, 1992
`25. Manjgaladze M, Chen S, Frame LT, Seng JE, Duffy PH, Feuers
`RJ, Hart RW, Leakey JE: Effects of caloric restriction on rodent
`drug and carcinogen metabolizing enzymes: Implications for
`mutagenesis and cancer. Mutat Res 295: 201–222, 1993
`
`26. Mote PL, Grizzle JM, Walford RL, Spindler SR: Influence of age
`and caloric restriction on expression of hepatic genes for xeno-
`biotic and oxygen metabolizing enzymes in the mouse. J Ger-
`ontol 46: B95–B100, 1991
`27. Klin M, Smogorzewski M, Ni Z, Zhang G, Massry SG:
`Abnormalities in hepatic lipase in chronic renal failure: Role
`of excess parathyroid hormone. J Clin Invest 97: 2167–2173,
`1996
`28. Ding H, Gao XL, Hirschberg R, Vadgama JV, Kopple JD:
`Impaired actions of insulin-like growth factor 1 on protein syn-
`thesis and degradation in skeletal muscle of rats with chronic
`renal failure. Evidence for a postreceptor defect. J Clin Invest 97:
`1064 –1075, 1996
`29. Massry SG, Fadda GZ: Chronic renal failure is a state of cellular
`calcium toxicity. Am J Kidney Dis 21: 81– 86, 1993
`30. Klin M, Smogorzewski

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket