Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
Drug Metab Pharmacokinet ; 27(5): 548-52, 2012.
Article in English | MEDLINE | ID: mdl-22498644

ABSTRACT

P-Glycoprotein (P-gp), one of the drug efflux pumps, is expressed in some tissues and may affect the pharmacokinetics of its substrates. We have previously reported that a decrease of intestinal P-gp expression affects the pharmacokinetics of orally-administered P-gp substrate drugs in a streptozotocin (STZ)-induced type 1 diabetic mice model. Although we have found the participation of nitric oxide synthase (NOS) activation as a mechanism of the decrease in intestinal P-gp expression under diabetic conditions, more detailed mechanisms other than NOS remain unknown. Here, we studied the involvement of the ubiquitin-proteasome system in the mechanism of the decrease in intestinal P-gp expression under diabetic conditions. Nine days after STZ administration (diabetic condition), ubiquitination levels of ileal P-gp were significantly increased, accompanied by an decrease of intestinal P-gp protein expression levels. Furthermore, treatment with an NO donor could increase the intestinal ubiquitinated P-gp levels. On the other hand, activity of 26S proteasome, an important enzyme in ubiquitin-proteasome system, did not change, suggesting the first step of the system (i.e., ubiquitination) but not the second step (i.e., degradation)-specific up-regulation under diabetic conditions. Our results reveal the participation of the acceleration of the ubiquitin-preotasome system by NO in the decrease of intestinal P-gp expression levels under diabetic conditions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Diabetes Mellitus, Experimental/metabolism , Ileum/metabolism , Ubiquitination , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Diabetes Mellitus, Experimental/genetics , Disease Models, Animal , Ileum/drug effects , Male , Mice , Nitric Oxide/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Up-Regulation/drug effects
2.
J Pharm Pharmacol ; 64(4): 496-504, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22420656

ABSTRACT

OBJECTIVES: Opioids and anticancer compounds such as etoposide (ETP) are substrates of P-glycoprotein (P-gp), an ATP-dependent efflux pump. Chemotherapy compounds may impact on the analgesic effect of opioids such as morphine when the two drugs are co-administered. In this study, we used a mouse model to determine if there is a pharmacological interaction between ETP and morphine, focusing on the involvement of intestinal P-gp. METHODS: P-gp drug efflux activity was measured by an in-situ closed loop method with Rhodamine 123, a P-gp substrate. The analgesic effect of morphine was determined by the tail-flick test. Intestinal P-gp expression levels were determined by Western blot. KEY FINDINGS: ETP and morphine significantly decreased the intestinal Rhodamine 123 efflux activity of P-gp. Oral morphine analgesia was significantly enhanced when co-administered with ETP. However, repeated pretreatment (7 days) with oral ETP significantly decreased the oral morphine-induced analgesia, in a cyclosporine A (a P-gp inhibitor) reversible manner. Furthermore, repeated ETP significantly up-regulated intestinal P-gp expression. CONCLUSIONS: It may be important to consider aspects of therapeutic design such as the administration route or scheduling of drugs in patients receiving concurrent chemotherapy and opioid therapy to avoid pharmacokinetic interactions between the two agents.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Analgesics, Opioid/pharmacology , Etoposide/pharmacology , Morphine/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Administration, Oral , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Cyclosporine/pharmacology , Disease Models, Animal , Drug Administration Schedule , Drug Interactions , Etoposide/administration & dosage , Intestinal Mucosa/metabolism , Male , Mice , Morphine/administration & dosage , Morphine/pharmacokinetics , Pain/drug therapy , Rhodamine 123/metabolism , Up-Regulation/drug effects
3.
Yakugaku Zasshi ; 132(2): 161-6, 2012.
Article in Japanese | MEDLINE | ID: mdl-22293693

ABSTRACT

P-glycoprotein (P-gp), one of the important drug-efflux pumps, is known to affect pharmacokinetics and pharmacodynamics of P-gp substrate drugs. We have previously reported that intestinal P-gp expression levels are transiently decreased in streptozotocin (STZ)-induced type 1 diabetic mouse model. Herein, we examined the analgesic effects of orally administered morphine and its pharmacokinetic properties under diabetic conditions, specifically focusing on the involvement of intestinal P-gp in a type 1 diabetic mouse model. Type 1 diabetes was induced in male ddY mice by an i.p. injection of STZ (230 mg/kg). We assessed the oral morphine analgesia using the tail-flick test. Serum and brain morphine content were determined on a HPLC-ECD system. Intestinal P-gp expression levels were significantly decreased on day 9 after STZ administration. On the other hands, oral morphine analgesia, and serum and brain morphine content were significantly increased on day 9 after STZ administration. The decrease in the intestinal P-gp expression levels were suppressed by aminoguanidine, a specific iNOS inhibitor. Interestingly, the increase in the analgesic effect of morphine, as well as serum and brain morphine content, was suppressed by aminoguanidine. Conversely, there was no change in the analgesic effect obtained with subcutaneous morphine in STZ-treated mice. In conclusions, our findings suggest that the oral morphine analgesia is dependent on intestinal P-gp expression, and that may be one of the problems against obtaining stable pharmacological effects of morphine in diabetic patients.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Analgesics, Opioid/pharmacokinetics , Diabetes Mellitus, Experimental/metabolism , Intestinal Mucosa/metabolism , Morphine/pharmacokinetics , Administration, Ophthalmic , Analgesics, Opioid/administration & dosage , Animals , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Male , Mice , Morphine/administration & dosage , Nitric Oxide Synthase Type II/antagonists & inhibitors , Time Factors , Tissue Distribution
4.
Life Sci ; 89(23-24): 834-8, 2011 Dec 05.
Article in English | MEDLINE | ID: mdl-21983297

ABSTRACT

AIMS: P-glycoprotein (P-gp) is an important drug efflux transporter located in many tissues such as the blood-brain barrier, intestines, liver and kidneys. We have previously reported that ileal P-gp expression levels decrease via a nitric oxide synthase (NOS)-mediated pathway in a streptozotocin (STZ)-induced type 1 diabetic mouse model. Herein, our objective was to assess whether there are differences in the expression of intestinal P-gp in an obesity-induced hyperglycemic mouse model versus the type 1 diabetic mouse model. MAIN METHODS: The hyperglycemia-accompanied obese mouse model was developed through an injection of monosodium glutamate (MSG). We analyzed intestinal P-gp expression using Western blot analysis. KEY FINDINGS: Body weight, body mass index, blood glucose levels and serum insulin levels increased significantly with age in the MSG-treated mice. Furthermore, in 24week-old MSG-treated mice, while intestinal P-gp expression levels were tended to increase P-gp expression in the duodenum, it was only significant in the jejunum, but not in the ileum. Additionally, the hyperglycemia-accompanied increase in intestinal NOS activity of STZ-treated mice was not evident in the MSG-treated mice. SIGNIFICANCE: Our results suggest that P-gp expression levels in the upper part of the intestine increase with age in a hyperglycemia/hyperinsulinemia (i.e. type 2 diabetes) -associated MSG-treated obese mouse model, and that these results completely differ from those found in the STZ-induced type 1 diabetic mouse model.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Intestines/drug effects , Mice, Obese/metabolism , Sodium Glutamate/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Animals , Blood Glucose/analysis , Blotting, Western , Body Mass Index , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Insulin/blood , Intestinal Mucosa/metabolism , Intestines/chemistry , Male , Mice , Nitric Oxide Synthase/metabolism
5.
Drug Metab Pharmacokinet ; 26(6): 584-91, 2011.
Article in English | MEDLINE | ID: mdl-21878740

ABSTRACT

Morphine is one of the strongest analgesics and is commonly used for the treatment of chronic pain. The pharmacokinetic properties of morphine are, in part, modulated by P-glycoprotein (P-gp). We previously reported that intestinal P-gp expression levels are influenced via the activation of inducible nitric oxide synthase (iNOS) in streptozotocin (STZ)-induced diabetic mice. Herein, we examine the analgesic effects of orally administered morphine and its pharmacokinetic properties under diabetic conditions, specifically we focusing on the involvement of intestinal P-gp in a type 1 diabetic mouse model. We assessed the analgesic effect of morphine using the tail-flick test. Serum and brain morphine levels were determined using a HPLC-ECD system. Oral morphine analgesic effects and serum and brain morphine content were significantly increased 9 days after STZ administration. The increase in the analgesic effects of morphine, as well as serum and brain morphine content, was suppressed by aminoguanidine, a specific iNOS inhibitor. Conversely, there were no changes in the analgesic effects obtained with subcutaneous morphine in STZ-treated mice. Our findings suggest that the analgesic effects of oral morphine are dependent on intestinal P-gp expression, and this may be one of the reasons that it is difficult to obtain stable pharmacological effects of morphine in diabetic patients.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Analgesics/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Intestinal Mucosa/metabolism , Morphine/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Analgesics/blood , Analgesics/pharmacokinetics , Animals , Brain/drug effects , Brain/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Guanidines/pharmacology , Intestines/drug effects , Male , Mice , Morphine/blood , Morphine/pharmacokinetics , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Pain/drug therapy
6.
Yakugaku Zasshi ; 131(4): 487-92, 2011 Apr.
Article in Japanese | MEDLINE | ID: mdl-21467785

ABSTRACT

P-glycoprotein (P-gp), one of the important drug-efflux pumps, is known to be affected by pathological conditions such as inflammation or infection. Recently, it is reported that high glucose or hyperglycemia can alter P-gp expression levels at the blood-brain barrier or in kidney, although the details are still unknown. Here, we analyzed the alteration of intestinal P-gp expression and function in the development of diabetes and elucidated the mechanisms. Type 1 diabetes was induced in male ddY mice by an i.p. injection of streptozotocin (STZ) (230 mg/kg). We analyzed ileal P-gp expression and drug efflux activity using western blot analysis and an in situ closed loop method, respectively. Additionally, we analyzed ileal nitric oxide synthase (NOS) activity using colorimetric method. A significant reduction of P-gp expression level in ileum was found on day 9 after STZ administration. In contrast, a remarkable decrease in drug efflux activity was observed on days 3 and 9. Interestingly, NOS activity in ilea was significantly increased on day 9. The decrease of P-gp expression levels observed on day 9 was completely suppressed by L-NG-nitroarginine methyl ester (L-NAME), a broad range NOS inhibitor, or aminoguanidine, a specific inducible NOS (iNOS) inhibitor. In addition, P-gp expression level in ileum was significantly decreased by administration of NOR5, a NO donor. These results indicate the possibility that NO, produced by iNOS in the ileum, is involved in the alteration of ileal P-gp expression and function under STZ-induced diabetic conditions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Diabetes Mellitus, Experimental/metabolism , Nitric Oxide Synthase Type II/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Disease Models, Animal , Guanidines/pharmacology , Humans , Ileum/enzymology , Ileum/metabolism , Male , Mice , Mice, Inbred Strains , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Rhodamine 123/metabolism
7.
Biol Pharm Bull ; 34(3): 436-8, 2011.
Article in English | MEDLINE | ID: mdl-21372399

ABSTRACT

P-glycoprotein (P-gp), a drug efflux transporter, affects the pharmacokinetics of a wide range of substrate drugs. Our previous study clearly revealed that intestinal P-gp expression levels were decreased via an inducible nitric oxide synthase (iNOS)-mediated mechanism in the early phases of diabetes. Here, we focused on changes in ileal P-gp expression and the influences of NOS on the P-gp expression levels in the later phase of diabetic condition using streptozotocin (STZ)-induced diabetic mice. The ileal P-gp expression and activity was analyzed by Western blot analysis and by in situ closed loop method, respectively. In STZ-treated mice, ileal P-gp expression levels and activity significantly decreased on the 9th day after STZ administration. Interestingly, the decrease of P-gp function was recovered to the control level on 15th day in same conditioned mice. In addition, the recovery of P-gp expression levels was completely suppressed by a non-selective NOS inhibitor. These results indicate that the diabetic condition-induced decline of P-gp expression levels was temporary, and both decline- and recovery-process of intestinal P-gp expression levels are mediated by NOS. Furthermore, this study shows the bidirectional effect of NOS on regulation of intestinal P-gp expression.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Diabetes Mellitus, Experimental/metabolism , Ileum/metabolism , Nitric Oxide Synthase/metabolism , Animals , Enzyme Inhibitors/pharmacology , Male , Mice , Mice, Inbred Strains , Streptozocin/pharmacology
8.
J Pharm Pharmacol ; 62(8): 995-1002, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20663033

ABSTRACT

OBJECTIVES: Repeated morphine treatment results in a decreased analgesic effect or the development of analgesic tolerance. However, we reported that some inflammatory chronic pain may inhibit morphine tolerance via kappa opioid receptor (KOR) activation. In this study, we further investigated the role of KOR in the inhibition of morphine tolerance in a chronic pain condition with a focus on the regulation of protein kinase C (PKC) activity. METHODS: Chronic pain was induced by formalin treatment into the dorsal part of the left hind paws of mice. The analgesic effect of morphine was measured by the tail flick method. We analysed the protein expression of PKC and its activity, and G-protein activity of mu opioid receptor (MOR) under repeated morphine treatment with or without formalin treatment. KEY FINDINGS: We found that conventional subtypes of PKC (cPKC) were up-regulated by repeated morphine treatment. Also, antisense oligonucleotide (AS-ODN) targeting cPKC completely suppressed the development of morphine tolerance. The disappearance of the repeated morphine-induced up-regulation of cPKC was completely reversed by treatment with AS-ODN targeting KOR. In addition, AS-ODN targeting KOR significantly reversed the chronic pain-induced down-regulation of PKC activity or up-regulation of MOR [(35)S]GTPgammaS binding activity after repeated morphine treatment. CONCLUSIONS: These results indicate that KOR plays an important role in the inhibition of repeated morphine-induced cPKC up-regulation under chronic pain condition. Furthermore, this may result in the increase of MOR activity and in the inhibition of morphine tolerance under chronic pain condition.


Subject(s)
Analgesics, Opioid/pharmacology , Brain/drug effects , Drug Tolerance , Morphine/pharmacology , Pain Threshold/drug effects , Pain/prevention & control , Protein Kinase C/metabolism , Receptors, Opioid, kappa/drug effects , Animals , Brain/enzymology , Chronic Disease , Disease Models, Animal , Down-Regulation , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Enzyme Activation , Formaldehyde , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Male , Mice , Naphthalenes/pharmacology , Oligonucleotides, Antisense/metabolism , Pain/chemically induced , Pain/enzymology , Pain Measurement , Protein Kinase C/genetics , Protein Kinase Inhibitors/pharmacology , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism , Time Factors
9.
Life Sci ; 86(11-12): 402-9, 2010 Mar 13.
Article in English | MEDLINE | ID: mdl-20097209

ABSTRACT

AIMS: P-glycoprotein (P-gp), one of the important drug-efflux pumps, is known to be affected by pathological conditions such as inflammation or infection. Recently, it is reported that high glucose or hyperglycemia can alternate P-gp expression levels at the blood-brain barrier or in the kidney, although the details are still unknown. Here, we analyzed the alteration of intestinal P-gp expression and function in the development of diabetes and elucidated the mechanisms. MAIN METHODS: Type 1 diabetes was induced in male ddY mice by an i.p. injection of streptozotocin (STZ) (230 mg/kg). We analyzed ileal P-gp expression and function using Western blot analysis and an in situ closed loop method, respectively. KEY FINDINGS: A significant reduction of P-gp expression level in ileum was found 9 days after STZ administration. In contrast, a remarkable decrease in P-gp function was observed on the 3rd and 9th days. Interestingly, nitric oxide synthase (NOS) activity in ilea was significantly increased on the 9th day. The decrease of P-gp expression levels observed on the 9th day was completely suppressed by L-N(G)-nitroarginine methyl ester (L-NAME), a broad range NOS inhibitor, or aminoguanidine, a specific inducible NOS (iNOS) inhibitor. SIGNIFICANCE: These results indicate the possibility that nitric oxide (NO), produced by iNOS in the ileum, is involved in the reduction of ileal P-gp expression under STZ-induced diabetic conditions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Diabetes Mellitus, Experimental/metabolism , Intestinal Mucosa/metabolism , Nitric Oxide Synthase Type II/physiology , Animals , Blood Glucose/metabolism , Blotting, Western , Body Weight/drug effects , Cytoplasm/enzymology , Cytoplasm/metabolism , Diabetes Mellitus, Experimental/enzymology , Drinking , Eating , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Insulin/blood , Intestinal Mucosa/enzymology , Intestines/enzymology , Male , Membranes/enzymology , Membranes/metabolism , Mice , Nitric Oxide Donors/pharmacology , Urodynamics/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...