1
|
Sullivan RM, Lucas CG, Sponchiado M, Eitel EK, Spate LD, Lucy MC, Smith MF, Wells KD, Prather RS, Geisert RD. Conceptus estrogen and prostaglandins provide the maternal recognition of pregnancy signal to prevent luteolysis during early pregnancy in the pig†. Biol Reprod 2024; 111:890-905. [PMID: 38904948 DOI: 10.1093/biolre/ioae104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/13/2024] [Accepted: 06/20/2024] [Indexed: 06/22/2024] Open
Abstract
Conceptus estrogens and prostaglandins have long been considered the primary signals for maternal recognition of pregnancy (MRP) in the pig. However, loss-of-function studies targeting conceptus aromatase genes (CYP19A1 and CYP19A2) and prostaglandin-endoperoxide synthase 2 (PTGS2) indicated that conceptuses can not only signal MRP without estrogens or prostaglandins but can maintain early pregnancy. However, complete loss of estrogen production leads to abortion after day 25 of gestation. Although neither conceptus estrogens nor prostaglandins had a significant effect on early maintenance of corpora lutea (CL) function alone, the two conceptus factors have a biological relationship. To investigate the role that both conceptus estrogens and prostaglandins have on MRP and maintenance of pregnancy, a triple loss-of function model (TKO) was generated for conceptus CYP19A1, CYP19A2, and PTGS2. In addition, a conceptus CYP19A2-/- model (A2KO) was established to determine the role of placental estrogen during later pregnancy. Estrogen and prostaglandin synthesis were greatly reduced in TKO concept uses which resulted in a failure to inhibit luteolysis after day 15 of pregnancy despite the presence of conceptuses in the uterine lumen. However, A2KO placentae not only maintained functional CL but were able to maintain pregnancy to day 32 of gestation. Despite the loss of placental CYP19A2 expression, the allantois fluid content of estrogen was not affected as the placenta compensated by expressing CYP19A1 and CYP19A3, which are normally absent in controls. Results suggest conceptuses can signal MRP through production of conceptus PGE or stimulating PGE synthesis from the endometrium through conceptus estrogen. Failure of conceptuses to produce both factors results in failure of MRP and loss of pregnancy.
Collapse
Affiliation(s)
- Riley M Sullivan
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Caroline G Lucas
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | | | - Emily K Eitel
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Lee D Spate
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Matthew C Lucy
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Michael F Smith
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Kevin D Wells
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Randall S Prather
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Rodney D Geisert
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| |
Collapse
|
2
|
Gomes EDT, Passos GR, Antunes NDJ, de Oliveira MG, de Souza VB, Schenka AA, da Costa JL, Antunes E, Mónica FZ. The multidrug resistance protein 4 is expressed and functionally active in isolated bladder from pig. Am J Physiol Regul Integr Comp Physiol 2024; 327:R291-R303. [PMID: 38881411 DOI: 10.1152/ajpregu.00238.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
Multidrug resistance proteins type 4 (MRP4) and 5 (MRP5) play pivotal roles in the transport of cyclic nucleotides in various tissues. However, their specific functions within the lower urinary tract remain relatively unexplored. This study aimed to investigate the effect of pharmacological inhibition of MRPs on cyclic nucleotide signaling in isolated pig bladder. The relaxation responses of the bladder were assessed in the presence of the MRP inhibitor, MK571. The temporal changes in intra- and extracellular levels of cAMP and cGMP in stimulated tissues were determined by mass spectrometry. The gene (ABCC4) and protein (MRP4) expression were also determined. MK571 administration resulted in a modest relaxation effect of approximately 26% in carbachol-precontracted bladders. The relaxation induced by phosphodiesterase inhibitors such as cilostazol, tadalafil, and sildenafil was significantly potentiated in the presence of MK571. In contrast, no significant potentiation was observed in the relaxation induced by substances elevating cAMP levels or stimulators of soluble guanylate cyclase. Following forskolin stimulation, both intracellular and extracellular cAMP concentrations increased by approximately 15.8-fold and 12-fold, respectively. Similarly, stimulation with tadalafil + BAY 41-2272 resulted in roughly 8.2-fold and 3.4-fold increases in intracellular and extracellular cGMP concentrations, respectively. The presence of MK571 reduced only the extracellular levels of cGMP. This study reveals the presence and function of MRP4 transporters within the porcine bladder and paves the way for future research exploring the role of this transporter in both underactive and overactive bladder disorders.NEW & NOTEWORTHY This study investigates the impact of pharmacological inhibition of MRP4 and MRP5 transporters on cyclic nucleotide signaling in isolated pig bladders. MK571 administration led to modest relaxation, with enhanced effects observed in the presence of phosphodiesterase inhibitors. However, substances elevating cAMP levels remained unaffected. MK571 selectively reduced extracellular cGMP levels. These findings shed light on the role of MRP4 transporters in the porcine bladder, opening avenues for further research into bladder disorders.
Collapse
Affiliation(s)
- Erick de Toledo Gomes
- Section of Pharmacology, Department of Translational Medicine, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Gabriela Reolon Passos
- Section of Pharmacology, Department of Translational Medicine, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Mariana Gonçalves de Oliveira
- Section of Pharmacology, Department of Translational Medicine, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Valeria Barbosa de Souza
- Section of Pharmacology, Department of Translational Medicine, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - André Almeida Schenka
- Section of Pharmacology, Department of Translational Medicine, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - José Luiz da Costa
- Campinas Poison Control Center, University of Campinas, Campinas, Brazil
| | - Edson Antunes
- Section of Pharmacology, Department of Translational Medicine, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Fabiola Zakia Mónica
- Section of Pharmacology, Department of Translational Medicine, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
3
|
Bazer FW, Johnson GA. Early Embryonic Development in Agriculturally Important Species. Animals (Basel) 2024; 14:1882. [PMID: 38997994 PMCID: PMC11240814 DOI: 10.3390/ani14131882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
The fertilization of oocytes ovulated by pigs, sheep, cows, and horses is not considered a limiting factor in successful establishment of pregnancy. Pig, sheep, and cow embryos undergo cleavage to the blastocyst stage, hatch from the zona pellucida, and undergo central-type implantation. Hatched blastocysts of pigs, sheep, and cows transition from tubular to long filamentous forms to establish surface area for exchange of nutrients and gases with the uterus. The equine blastocyst, surrounded by external membranes, does not elongate but migrates throughout the uterine lumen before attaching to the uterine luminal epithelium (LE) to begin implantation. Pregnancy recognition signaling in pigs requires the trophectoderm to express interleukin 1 beta, estrogens, prostaglandin E2, and interferon gamma. Sheep and cow conceptus trophectoderm expresses interferon tau that induces interferon regulatory factor 2 that inhibits transcription of estrogen and oxytocin receptors by uterine epithelia. This prevents oxytocin-induced luteolytic pulses of prostaglandin F2-alpha from regressing the corpora lutea, as well as ensuring the secretion of progesterone required for maintenance of pregnancy. The pregnancy recognition signal produced by equine blastocysts is not known. Implantation in these species requires interactions between extracellular matrix (ECM) proteins and integrins as the conceptus undergoes apposition and firm attachment to the uterine LE. This review provides details with respect to early embryonic development and the transition from spherical to filamentous conceptuses in pigs, sheep, and cows, as well as pre-implantation development of equine blastocysts and implantation of the conceptuses.
Collapse
Affiliation(s)
- Fuller W. Bazer
- Department of Animal Science, Texas A&M University, College Station, TX 77843-2471, USA
| | - Gregory A. Johnson
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-2471, USA;
| |
Collapse
|
4
|
Geisert RD, Bazer FW, Lucas CG, Pfeiffer CA, Meyer AE, Sullivan R, Johns DN, Sponchiado M, Prather RS. Maternal recognition of pregnancy in the pig: A servomechanism involving sex steroids, cytokines and prostaglandins. Anim Reprod Sci 2024; 264:107452. [PMID: 38522133 DOI: 10.1016/j.anireprosci.2024.107452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
Maternal recognition of pregnancy (MRP) is a term utilized in mammals to describe pathways in which the conceptus alters the endometrial environment to prevent regression of corpora lutea to ensure continued production of progesterone (P4) required for establishment and maintenance of pregnancy. For nearly 40 years after publication of the endocrine/exocrine theory, conceptus estrogen (E2) was considered the primary maternal recognition signal in the pig. Conceptus production of prostaglandin E2 (PGE2) was also considered to be a major factor in preventing luteolysis. An addition to E2 and PGE2, pig conceptuses produce interleukin 1B2 (IL1B2) and interferons (IFN) delta (IFND) and gamma (IFNG). The present review provides brief history of the discovery of E2, PGs and IFNS which led to research investigating the role of these conceptus secreted factors in establishing and maintaining pregnancy in the pig. The recent utilization of gene editing technology allowed a more direct approach to investigate the in vivo roles of IL1B2, E2, PGE2, AND IFNG for establishment of pregnancy. These studies revealed unknown functions for IFNG and ILB2 in addition to PGE2 and E2. Thus, pregnancy recognition signal is via a servomechanism in requiring sequential effects of P4, E2, IL1B2, PGE2 and IFNG. Results indicate that the original established dogma for the role of conceptus E2 and PGs in MRP is a far too simplified model that involves the interplay of numerous mechanisms for inhibiting luteolysis, inducing critical elongation of the conceptuses and resolution of inflammation in pigs.
Collapse
Affiliation(s)
- Rodney D Geisert
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA.
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| | - Caroline G Lucas
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Caroline A Pfeiffer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Ashley E Meyer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Riley Sullivan
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Destiny N Johns
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Mariana Sponchiado
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Randall S Prather
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
5
|
Pourmal S, Green E, Bajaj R, Chemmama IE, Knudsen GM, Gupta M, Sali A, Cheng Y, Craik CS, Kroetz DL, Stroud RM. Structural basis of prostaglandin efflux by MRP4. Nat Struct Mol Biol 2024; 31:621-632. [PMID: 38216659 PMCID: PMC11145372 DOI: 10.1038/s41594-023-01176-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 11/07/2023] [Indexed: 01/14/2024]
Abstract
Multidrug resistance protein 4 (MRP4) is a broadly expressed ATP-binding cassette transporter that is unique among the MRP subfamily for transporting prostanoids, a group of signaling molecules derived from unsaturated fatty acids. To better understand the basis of the substrate selectivity of MRP4, we used cryogenic-electron microscopy to determine six structures of nanodisc-reconstituted MRP4 at various stages throughout its transport cycle. Substrate-bound structures of MRP4 in complex with PGE1, PGE2 and the sulfonated-sterol DHEA-S reveal a common binding site that accommodates a diverse set of organic anions and suggest an allosteric mechanism for substrate-induced enhancement of MRP4 ATPase activity. Our structure of a catalytically compromised MRP4 mutant bound to ATP-Mg2+ is outward-occluded, a conformation previously unobserved in the MRP subfamily and consistent with an alternating-access transport mechanism. Our study provides insights into the endogenous function of this versatile efflux transporter and establishes a basis for MRP4-targeted drug design.
Collapse
Affiliation(s)
- Sergei Pourmal
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Program in Chemistry and Chemical Biology, University of California, San Francisco, CA, USA
- Genentech, South San Francisco, CA, USA
| | - Evan Green
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Biophysics Graduate Program, University of California, San Francisco, CA, USA
- Exelixis, Alameda, CA, USA
| | - Ruchika Bajaj
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Ilan E Chemmama
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
- Brightseed, South San Francisco, CA, USA
| | - Giselle M Knudsen
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
| | - Meghna Gupta
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| | - Charles S Craik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Deanna L Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA.
| | - Robert M Stroud
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.
| |
Collapse
|
6
|
Chen Y, Wang L, Hou WT, Zha Z, Xu K, Zhou CZ, Li Q, Chen Y. Structural insights into human ABCC4-mediated transport of platelet agonist and antagonist. NATURE CARDIOVASCULAR RESEARCH 2023; 2:693-701. [PMID: 39195918 DOI: 10.1038/s44161-023-00289-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/19/2023] [Indexed: 08/29/2024]
Abstract
Human platelets contribute to hemostasis and thrombosis, the imbalance of which can cause cardiovascular diseases. The activation and accumulation of platelets can be induced by agonists or inhibited by antagonists. Thus, the human ABC transporter ABCC4, which pumps out platelet agonists and antagonists, might become a promising target for preventing cardiovascular diseases. Here we define five structures of human ABCC4: the apo and three complexed forms in the inward-facing conformation, in addition to an outward-facing occluded conformation upon ATP binding. Combined with biochemical assays, we structurally prove that U46619, a synthetic analog of the unstable agonist TXA2, and the antagonist aspirin are substrates of ABCC4. In addition, we found that the platelet antagonist dipyridamole is a strong competitive inhibitor against ABCC4. These complex structures also enable us to identify a transmembrane pocket in ABCC4 that provides a defined space for the rational design of specific platelet antagonists.
Collapse
Affiliation(s)
- Yu Chen
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Liang Wang
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Wen-Tao Hou
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Zhihui Zha
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Kang Xu
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Cong-Zhao Zhou
- School of Life Sciences, University of Science and Technology of China, Hefei, China.
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| | - Qiong Li
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- School of Life Sciences, University of Science and Technology of China, Hefei, China.
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| | - Yuxing Chen
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- School of Life Sciences, University of Science and Technology of China, Hefei, China.
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
7
|
Yerushalmi GM, Shuraki B, Yung Y, Maman E, Baum M, Hennebold JD, Adashi EY, Hourvitz A. ABCC4 is a PGE2 efflux transporter in the ovarian follicle: A mediator of ovulation and a potential non-hormonal contraceptive target. FASEB J 2023; 37:e22858. [PMID: 36943419 DOI: 10.1096/fj.202101931rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 01/12/2023] [Accepted: 02/22/2023] [Indexed: 03/23/2023]
Abstract
The role of prostaglandins (PGs) in the ovulatory process is known. However, the role of the ATP binding cassette subfamily C member 4 (ABCC4), transmembrane PG carrier protein, in ovulation remains unknown. We report herein that ABCC4 expression is significantly upregulated in preovulatory human granulosa cells (GCs). We found that PGE2 efflux in cultured human GCs is mediated by ABCC4 thus regulating its extracellular concentration. The ABCC4 inhibitor probenecid demonstrated effective blocking of ovulation and affects key ovulatory genes in female mice in vivo. We postulate that the reduction in PGE2 efflux caused by the inhibition of ABCC4 activity in GCs decreases the extracellular concentration of PGE2 and its ovulatory effect. Treatment of female mice with low dose of probenecid as well as with the PTGS inhibitor indomethacin or Meloxicam synergistically blocks ovulation. These results support the hypothesis that ABCC4 has an important role in ovulation and might be a potential target for non-hormonal contraception, especially in combination with PGE2 synthesis inhibitors. These findings may fill the gap in understanding the role of ABCC4 in PGE2 signaling, enhance the understanding of ovulatory disorders, and facilitate the treatment and control of fertility.
Collapse
Affiliation(s)
- Gil M Yerushalmi
- Reproduction Laboratory and IVF Unit, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv), Tel Hashomer, Israel
- IVF Unit, Department of Obstetrics and Gynecology, The Yitzhak Shamir Medical Center (formerly Assaf Harofeh Medical Center) (affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv), Zerifin, Israel
| | - Batel Shuraki
- Reproduction Laboratory and IVF Unit, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv), Tel Hashomer, Israel
| | - Yuval Yung
- Reproduction Laboratory and IVF Unit, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv), Tel Hashomer, Israel
| | - Ettie Maman
- Reproduction Laboratory and IVF Unit, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv), Tel Hashomer, Israel
| | - Micha Baum
- Reproduction Laboratory and IVF Unit, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv), Tel Hashomer, Israel
| | - Jon D Hennebold
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Eli Y Adashi
- Department of Medical Science and Obstetrics and Gynecology, the Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Department of Obstetrics and Gynecology, the Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Ariel Hourvitz
- Reproduction Laboratory and IVF Unit, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv), Tel Hashomer, Israel
- IVF Unit, Department of Obstetrics and Gynecology, The Yitzhak Shamir Medical Center (formerly Assaf Harofeh Medical Center) (affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv), Zerifin, Israel
| |
Collapse
|
8
|
Ghanem CI, Manautou JE. Role and Regulation of Hepatobiliary ATP-Binding Cassette Transporters during Chemical-Induced Liver Injury. Drug Metab Dispos 2022; 50:1376-1388. [PMID: 35914951 PMCID: PMC9513844 DOI: 10.1124/dmd.121.000450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/20/2022] [Indexed: 11/22/2022] Open
Abstract
Severity of drug-induced liver injury (DILI) ranges from mild, asymptomatic, and transient elevations in liver function tests to irreversible liver damage, often needing transplantation. Traditionally, DILI is classified mechanistically as high-frequency intrinsic DILI, commonly dose dependent or DILI that rarely occurs and is idiosyncratic in nature. This latter form is not dose dependent and has a pattern of histopathological manifestation that is not always uniform. Currently, a third type of DILI called indirect hepatotoxicity has been described that is associated with the pharmacological action of the drug. Historically, DILI was primarily linked to drug metabolism events; however, the impact of transporter-mediated rates of drug uptake and excretion has gained greater prominence in DILI research. This review provides a comprehensive view of the major findings from studies examining the contribution of hepatic ATP-binding cassette transporters as key contributors to DILI and how changes in their expression and function influence the development, severity, and overall toxicity outcome. SIGNIFICANCE STATEMENT: Drug-induced liver injury (DILI) continues to be a focal point in drug development research. ATP-binding cassette (ABC) transporters have emerged as important determinants of drug detoxification, disposition, and safety. This review article provides a comprehensive analysis of the literature addressing: (a) the role of hepatic ABC transporters in DILI, (b) the influence of genetic mutations in ABC transporters on DILI, and (c) new areas of research emphasis, such as the influence of the gut microbiota and epigenetic regulation, on ABC transporters.
Collapse
Affiliation(s)
- Carolina I Ghanem
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET) (C.I.G.) and Cátedra de Fisiopatología (C.I.G.), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina; and Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (J.E.M.)
| | - Jose E Manautou
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET) (C.I.G.) and Cátedra de Fisiopatología (C.I.G.), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina; and Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (J.E.M.)
| |
Collapse
|
9
|
Wang JQ, Wu ZX, Yang Y, Teng QX, Li YD, Lei ZN, Jani KA, Kaushal N, Chen ZS. ATP-binding cassette (ABC) transporters in cancer: A review of recent updates. J Evid Based Med 2021; 14:232-256. [PMID: 34388310 DOI: 10.1111/jebm.12434] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023]
Abstract
The ATP-binding cassette (ABC) transporter superfamily is one of the largest membrane protein families existing in wide spectrum of organisms from prokaryotes to human. ABC transporters are also known as efflux pumps because they mediate the cross-membrane transportation of various endo- and xenobiotic molecules energized by ATP hydrolysis. Therefore, ABC transporters have been considered closely to multidrug resistance (MDR) in cancer, where the efflux of structurally distinct chemotherapeutic drugs causes reduced itherapeutic efficacy. Besides, ABC transporters also play other critical biological roles in cancer such as signal transduction. During the past decades, extensive efforts have been made in understanding the structure-function relationship, transportation profile of ABC transporters, as well as the possibility to overcome MDR via targeting these transporters. In this review, we discuss the most recent knowledge regarding ABC transporters and cancer drug resistance in order to provide insights for the development of more effective therapies.
Collapse
Affiliation(s)
- Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Yi-Dong Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
- School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Khushboo A Jani
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Neeraj Kaushal
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| |
Collapse
|
10
|
Arendt M, Ambrosen A, Fall T, Kierczak M, Tengvall K, Meadows JRS, Karlsson Å, Lagerstedt AS, Bergström T, Andersson G, Lindblad-Toh K, Hagman R. The ABCC4 gene is associated with pyometra in golden retriever dogs. Sci Rep 2021; 11:16647. [PMID: 34404837 PMCID: PMC8370986 DOI: 10.1038/s41598-021-95936-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/29/2021] [Indexed: 11/24/2022] Open
Abstract
Pyometra is one of the most common diseases in female dogs, presenting as purulent inflammation and bacterial infection of the uterus. On average 20% of intact female dogs are affected before 10 years of age, a proportion that varies greatly between breeds (3–66%). The clear breed predisposition suggests that genetic risk factors are involved in disease development. To identify genetic risk factors associated with the disease, we performed a genome-wide association study (GWAS) in golden retrievers, a breed with increased risk of developing pyometra (risk ratio: 3.3). We applied a mixed model approach comparing 98 cases, and 96 healthy controls and identified an associated locus on chromosome 22 (p = 1.2 × 10–6, passing Bonferroni corrected significance). This locus contained five significantly associated SNPs positioned within introns of the ATP-binding cassette transporter 4 (ABCC4) gene. This gene encodes a transmembrane transporter that is important for prostaglandin transport. Next generation sequencing and genotyping of cases and controls subsequently identified four missense SNPs within the ABCC4 gene. One missense SNP at chr22:45,893,198 (p.Met787Val) showed complete linkage disequilibrium with the associated GWAS SNPs suggesting a potential role in disease development. Another locus on chromosome 18 overlapping the TESMIN gene, is also potentially implicated in the development of the disease.
Collapse
Affiliation(s)
- Maja Arendt
- Faculty of Health and Medical Sciences, Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark. .,Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
| | - Aime Ambrosen
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Marcin Kierczak
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Katarina Tengvall
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jennifer R S Meadows
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Åsa Karlsson
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Anne-Sofie Lagerstedt
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Tomas Bergström
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Göran Andersson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Kerstin Lindblad-Toh
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ragnvi Hagman
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| |
Collapse
|
11
|
Liu W, Liu Y. Roles of Multidrug Resistance Protein 4 in Microbial Infections and Inflammatory Diseases. MICROBIAL DRUG RESISTANCE (LARCHMONT, N.Y.) 2021; 27:1535-1545. [PMID: 33999661 DOI: 10.1089/mdr.2020.0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Numerous studies have reported the emergence of antimicrobial resistance during the treatment of common infections. Multidrug resistance (MDR) leads to failure of antimicrobial treatment, prolonged illness, and increased morbidity and mortality. Overexpression of multidrug resistance proteins (MRPs) as drug efflux pumps are one of the main contributions of MDR, especially multidrug resistance protein 4 (MRP4/ABCC4) in the development of antimicrobial resistance. The molecular mechanism of antimicrobial resistance is still under investigation. Various intervention strategies have been developed for overcoming MDR, but the effect is limited. Suppression of MRP4 may be an attractive therapeutic approach for addressing drug resistance. However, there are few reports on the involvement of MRP4 in antimicrobial resistance and inflammatory diseases. In this review, we introduced the function and regulation of MRP4, and then summarized the roles of MRP4 in microbial infections and inflammatory diseases as well as polymorphisms in the gene encoding this transporter. Further studies should be conducted on drug therapy targeting MRP4 to improve the efficacy of antimicrobial therapy. This review can provide useful information on MRP4 for overcoming antimicrobial resistance and anti-inflammatory therapy.
Collapse
Affiliation(s)
- Wei Liu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yutian Liu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Nguyen JP, Kim Y, Cao Q, Hirota JA. Interactions between ABCC4/MRP4 and ABCC7/CFTR in human airway epithelial cells in lung health and disease. Int J Biochem Cell Biol 2021; 133:105936. [PMID: 33529712 DOI: 10.1016/j.biocel.2021.105936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/13/2020] [Accepted: 01/07/2021] [Indexed: 12/26/2022]
Abstract
ATP binding cassette (ABC) transporters are present in all three domains of life - Archaea, Bacteria, and Eukarya. The conserved nature is a testament to the importance of these transporters in regulating endogenous and exogenous substrates required for life to exist. In humans, 49 ABC transporters have been identified to date with broad expression in different lung cell types with multiple transporter family members contributing to lung health and disease. The ABC transporter most commonly known to be linked to lung pathology is ABCC7, also known as cystic fibrosis transmembrane conductance regulator - CFTR. Closely related to the CFTR genomic sequence is ABCC4/multi-drug resistance protein-4. Genomic proximity is shared with physical proximity, with ABCC4 and CFTR physically coupled in cell membrane microenvironments of epithelial cells to orchestrate functional consequences of cyclic-adenosine monophosphate (cAMP)-dependent second messenger signaling and extracellular transport of endogenous and exogenous substrates. The present concise review summarizes the emerging data defining a role of the (ABCC7/CFTR)-ABCC4 macromolecular complex in human airway epithelial cells as a physiologically important pathway capable of impacting endogenous and exogenous mediator transport and ion transport in both lung health and disease.
Collapse
Affiliation(s)
- Jenny P Nguyen
- Department of Medicine, McMaster University, Canada; Firestone Institute for Respiratory Health, St. Joseph's Hospital, Canada
| | - Yechan Kim
- Department of Medicine, McMaster University, Canada; Firestone Institute for Respiratory Health, St. Joseph's Hospital, Canada
| | - Quynh Cao
- Department of Medicine, McMaster University, Canada; Firestone Institute for Respiratory Health, St. Joseph's Hospital, Canada
| | - Jeremy A Hirota
- Department of Medicine, McMaster University, Canada; Firestone Institute for Respiratory Health, St. Joseph's Hospital, Canada; McMaster Immunology Research Centre, McMaster University, Canada; Department of Biology, University of Waterloo, Canada; Department of Medicine, University of British Columbia, Canada.
| |
Collapse
|
13
|
Roles of ABCC1 and ABCC4 in Proliferation and Migration of Breast Cancer Cell Lines. Int J Mol Sci 2020; 21:ijms21207664. [PMID: 33081264 PMCID: PMC7589126 DOI: 10.3390/ijms21207664] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
ABCC1 and ABCC4 utilize energy from ATP hydrolysis to transport many different molecules, including drugs, out of the cell and, as such, have been implicated in causing drug resistance. However recently, because of their ability to transport signaling molecules and inflammatory mediators, it has been proposed that ABCC1 and ABCC4 may play a role in the hallmarks of cancer development and progression, independent of their drug efflux capabilities. Breast cancer is the most common cancer affecting women. In this study, the aim was to investigate whether ABCC1 or ABCC4 play a role in the proliferation or migration of breast cancer cell lines MCF-7 (luminal-type, receptor-positive) and MDA-MB-231 (basal-type, triple-negative). The effects of small molecule inhibitors or siRNA-mediated knockdown of ABCC1 or ABCCC4 were measured. Colony formation assays were used to assess the clonogenic capacity, MTT assays to measure the proliferation, and scratch assays and Transwell assays to monitor the cellular migration. The results showed a role for ABCC1 in cellular proliferation, whilst ABCC4 appeared to be more important for cellular migration. ELISA studies implicated cAMP and/or sphingosine-1-phosphate efflux in the mechanism by which these transporters mediate their effects. However, this needs to be investigated further, as it is key to understand the mechanisms before they can be considered as targets for treatment.
Collapse
|
14
|
Tanaka N, Kawai J, Hirasawa N, Mano N, Yamaguchi H. ATP-Binding Cassette Transporter C4 is a Prostaglandin D2 Exporter in HMC-1 cells. Prostaglandins Leukot Essent Fatty Acids 2020; 159:102139. [PMID: 32544819 DOI: 10.1016/j.plefa.2020.102139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/11/2020] [Accepted: 05/25/2020] [Indexed: 01/07/2023]
Abstract
ATP-binding cassette transporter C4 (ABCC4) is associated with multidrug resistance and the regulation of cell signalling. Some prostaglandins (PGs), including: PGE2, PGF2α, PGE3, and PGF3α are known substrates of ABCC4, and are released from some types of cells to exert their biological effects. In the present study, we demonstrate that PGD2 is a novel substrate of ABCC4 using a transport assay based on inside-out membrane vesicles prepared from ABCC4-overexpressing cells. Then, we used two types of cell lines with confirmed ABCC4 mRNA and PGD2 release capacity (human mast cell lines HMC-1 cells and human rhabdomyosarcoma cell lines TE671 cells) to evaluate the contribution of ABCC4. The extracellular levels of PGD2 were unchanged following addition of a selective ABCC4 inhibitor in TE671 cells. Pharmacological inhibition and knockdown of ABCC4 significantly reduced the extracellular levels of PGD2 by at least 53% in HMC-1 cells. Moreover, the extracellular levels of PGD2 decreased by at least 20% using the selective ABCC4 inhibitor in the other mast cell line RBL-2H3 cells. Therefore, our results suggest that ABCC4 functions as a PGD2 exporter in HMC-1 cells.
Collapse
Affiliation(s)
- Nobuaki Tanaka
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Junya Kawai
- Mushroom Research Laboratory, Hokuto Corporation, 800-8, Shimokomazawa, Nagano, 381-0008, Japan; Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Noriyasu Hirasawa
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Nariyasu Mano
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, 980-8574, Japan
| | - Hiroaki Yamaguchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, 980-8574, Japan; Department of Pharmacy, Yamagata University Hospital, Yamagata, 990-9585, Japan.
| |
Collapse
|
15
|
Jones RS, Parker MD, Morris ME. Monocarboxylate Transporter 6-Mediated Interactions with Prostaglandin F 2α: In Vitro and In Vivo Evidence Utilizing a Knockout Mouse Model. Pharmaceutics 2020; 12:pharmaceutics12030201. [PMID: 32110957 PMCID: PMC7150767 DOI: 10.3390/pharmaceutics12030201] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/12/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023] Open
Abstract
Monocarboxylate transporter 6 (MCT6; SLC16A5) is a recently studied drug transporter that currently has no annotated endogenous function. Currently, only a handful of compounds have been characterized as substrates for MCT6 (e.g., bumetanide, nateglinide, probenecid, and prostaglandin F2α (PGF2α)). The objective of our research was to characterize the MCT6-specific transporter kinetic parameters and MCT6-specific in vitro and in vivo interactions of PGF2α. Murine and human MCT6-mediated transport of PGF2α was assessed in MCT6-transfected oocytes. Additionally, endogenous PGF2α and a primary PGF2α metabolite (PGFM) were measured in plasma and urine in Mct6 knockout (Mct6−/−) and wild-type (Mct6+/+) mice. Results demonstrated that the affinity was approximately 40.1 and 246 µM respectively, for mouse and human, at pH 7.4. In vivo, plasma PGF2α concentrations in Mct6−/− mice were significantly decreased, compared to Mct6+/+ mice (3.3-fold). Mct6-/- mice demonstrated a significant increase in urinary PGF2α concentrations (1.7-fold). A similar trend was observed with plasma PGFM concentrations. However, overnight fasting resulted in significantly increased plasma PGF2α concentrations, suggesting a diet-dependent role of Mct6 regulation on the homeostasis of systemic PGF2α. Overall, these results are the first to suggest the potential regulatory role of MCT6 in PGF2α homeostasis, and potentially other PGs, in distribution and metabolism.
Collapse
Affiliation(s)
- Robert S. Jones
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA;
- Current Address Is Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Mark D. Parker
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA;
| | - Marilyn E. Morris
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA;
- Correspondence: ; Tel.: +1-(716)-645-4839
| |
Collapse
|
16
|
Tanaka N, Yamaguchi H, Mano N. Involvement of H +-gradient dependent transporter in PGE 2 release from A549 cells. Prostaglandins Leukot Essent Fatty Acids 2019; 149:30-36. [PMID: 31421525 DOI: 10.1016/j.plefa.2019.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/18/2019] [Accepted: 08/07/2019] [Indexed: 11/24/2022]
Abstract
The purpose of this study was to identify the transporter involved in the release of prostaglandin E2 (PGE2). In the present study, transport assays were conducted using membrane vesicles prepared from human lung adenocarcinoma A549 cells, thus enabling identification of the novel exporter present in A549 cells. PGE2 transport into A549 vesicles was higher in the presence of a proton (H+)-gradient, thus suggesting the involvement of PGE2H+ symporter in PGE2 transport. Results from our experiments showed enhanced PGE2 release in A549 cells in the presence of H+-gradient ([H+]extracellular < [H+]intracellular). Moreover, in vesicular transport assays, H+-gradient-dependent transport of PGE2 did not show saturation up to 500 μM PGE2, and 10 mM aromatic monocarboxylic acids (acetylsalicylic acid, salicylic acid, and p-nitrobenzoic acid) significantly inhibited PGE2 transport by 62-70%. These results suggest, the involvement of monocarboxylate transporters in the H+-gradient-dependent PGE2 export.
Collapse
Affiliation(s)
- Nobuaki Tanaka
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Hiroaki Yamaguchi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, 980-8574, Japa; Yamagata University Graduate School of Medicine/Department of Pharmacy, Yamagata University Hospital, 2-2-2, Iida-nishi, Yamagata, 990-9585, Japan.
| | - Nariyasu Mano
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, 980-8574, Japa
| |
Collapse
|
17
|
Yaneff A, Sahores A, Gómez N, Carozzo A, Shayo C, Davio C. MRP4/ABCC4 As a New Therapeutic Target: Meta-Analysis to Determine cAMP Binding Sites as a Tool for Drug Design. Curr Med Chem 2019; 26:1270-1307. [PMID: 29284392 DOI: 10.2174/0929867325666171229133259] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 12/01/2017] [Accepted: 12/14/2017] [Indexed: 02/06/2023]
Abstract
MRP4 transports multiple endogenous and exogenous substances and is critical not only for detoxification but also in the homeostasis of several signaling molecules. Its dysregulation has been reported in numerous pathological disorders, thus MRP4 appears as an attractive therapeutic target. However, the efficacy of MRP4 inhibitors is still controversial. The design of specific pharmacological agents with the ability to selectively modulate the activity of this transporter or modify its affinity to certain substrates represents a challenge in current medicine and chemical biology. The first step in the long process of drug rational design is to identify the therapeutic target and characterize the mechanism by which it affects the given pathology. In order to develop a pharmacological agent with high specific activity, the second step is to systematically study the structure of the target and identify all the possible binding sites. Using available homology models and mutagenesis assays, in this review we recapitulate the up-to-date knowledge about MRP structure and aligned amino acid sequences to identify the candidate MRP4 residues where cyclic nucleotides bind. We have also listed the most relevant MRP inhibitors studied to date, considering drug safety and specificity for MRP4 in particular. This meta-analysis platform may serve as a basis for the future development of inhibitors of MRP4 cAMP specific transport.
Collapse
Affiliation(s)
- Agustín Yaneff
- Instituto de Investigaciones Farmacologicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana Sahores
- Instituto de Investigaciones Farmacologicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natalia Gómez
- Instituto de Investigaciones Farmacologicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro Carozzo
- Instituto de Investigaciones Farmacologicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carina Shayo
- Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Carlos Davio
- Instituto de Investigaciones Farmacologicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
18
|
Abstract
INTRODUCTION In men, lower urinary tract symptoms (LUTS) are primarily attributed to benign prostatic hyperplasia (BPH). Therapeutic options are targeted to relax prostate smooth muscle and/or reduce prostate enlargement. Areas covered: This article reviews the major preclinical and clinical data on PDE5 inhibitors with a specific focus on tadalafil. It includes details of the role of the nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) - PDE5 pathway in the LUT organs (bladder and prostate) in addition to the available data on tadalafil in patients with LUTS secondary to BPH with or without erectile dysfunction (ED). Expert opinion: Preclinical and clinical data have clearly demonstrated that PDE5 inhibitors induce bladder and prostate relaxation, which contributes to the improvement seen in storage symptoms in both animal models of bladder and prostate hypercontractility. Tadalafil is effective both as a monotherapy and add-on therapy in patients with LUTS secondary to BPH. Furthermore, as LUTS-BPH and ED are urological disorders that commonly coexist in aging men, tadalafil is more advantageous than α1-adrenoceptors and should be used as the first option. Tadalafil is a safe and tolerable therapy and unlike α1- adrenoceptors and 5-alpha reductase inhibitors, which can cause sexual dysfunctions, tadalafil improves sexual function.
Collapse
Affiliation(s)
- Fabiola Zakia Mónica
- a Department of Pharmacology, Faculty of Medical Sciences , University of Campinas , Campinas , Sao Paulo , Brazil
| | - Gilberto De Nucci
- a Department of Pharmacology, Faculty of Medical Sciences , University of Campinas , Campinas , Sao Paulo , Brazil
| |
Collapse
|
19
|
Yamaguchi H, Mano N. Analysis of membrane transport mechanisms of endogenous substrates using chromatographic techniques. Biomed Chromatogr 2019; 33:e4495. [PMID: 30661254 DOI: 10.1002/bmc.4495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 02/06/2023]
Abstract
Membrane transporters are expressed in various bodily tissues and play essential roles in the homeostasis of endogenous substances and the absortion, distribution and/or excretion of xenobiotics. For transporter assays, radioisotope-labeled compounds have been mainly used. However, commercially available radioisotope-labeled compounds are limited in number and relatively expensive. Chromatographic analyses such as high-performance liquid chromatography with ultraviolet absorptiometry and liquid chromatography with tandem mass spectrometry have also been applied for transport assays. To elucidate the transport properties of endogenous substrates, although there is no difficulty in performing assays using radioisotope-labeled probes, the endogenous background and the metabolism of the compound after its translocation across cell membranes must be considered when the intact compound is assayed. In this review, the current state of knowledge about the transport of endogenous substrates via membrane transporters as determined by chromatographic techniques is summarized. Chromatographic techniques have contributed to our understanding of the transport of endogenous substances including amino acids, catecholamines, bile acids, prostanoids and uremic toxins via membrane transporters.
Collapse
Affiliation(s)
- Hiroaki Yamaguchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
20
|
Bertollotto GM, de Oliveira MG, Alexandre EC, Calmasini FB, Passos GR, Antunes E, Mónica FZ. Inhibition of Multidrug Resistance Proteins by MK 571 Enhances Bladder, Prostate, and Urethra Relaxation through cAMP or cGMP Accumulation. J Pharmacol Exp Ther 2018; 367:138-146. [PMID: 30108158 DOI: 10.1124/jpet.118.250076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/03/2018] [Indexed: 12/28/2022] Open
Abstract
The biologic effect of cAMP and cGMP is terminated by phosphodiesterases and multidrug resistance proteins MRP4 and MRP5, which pump cyclic nucleotides out of the cell. Therefore, this study aimed to characterize the role of MRP inhibitor, MK 571 (3-[[[3-[(1E)-2-(7-chloro-2-quinolinyl)ethenyl]phenyl][[3-(dimethylamino)-3-oxopropyl]thio]methyl]thio]propanoic acid), in the bladder, prostate, and urethra of male mice by means of functional assays, protein expression, and cyclic nucleotide quantification. The cumulative addition of MK 571 (1-30 µM) produced only small relaxation responses (approximately 25%) in all studied tissues. In the bladder, isoprenaline/fenoterol and forskolin concentration-dependently relaxed and MK 571 (20 µM) increased the maximal response values by 37% and 24%, respectively. When MK 571 was coincubated with fenoterol or forskolin, intracellular levels of cAMP and protein expression of phospho-vasodilator-stimulated phosphoprotein (p-VASP) Ser157 were significantly greater compared with bladders stimulated with fenoterol or forskolin alone. In the prostate and urethra, sodium nitroprusside concentration-dependently relaxed and MK 571 (20 µM) significantly increased relaxation responses by 70% and 56%, respectively, accompanied by greater intracellular levels of cGMP and protein expression of p-VASP Ser239 in the prostate. Tadalafil and BAY 41-2272 (5-cyclopropyl-2-[1-[(2-fluorophenyl)methyl]-1H-pyrazolo[3,4-b]pyridin-3-yl]-4-pyrimidinamine) also relaxed the prostate and urethra, respectively, and MK 571 markedly enhanced this response. The stable analog of cGMP (8-Br-cGMP) induced concentration-dependent relaxation responses in the prostate and urethra, and MK 571 significantly increased the relaxation response. In conclusion, to our knowledge, this is the first study to show that efflux transporters are physiologically active in the bladder, prostate, and urethra to control intracellular levels of cAMP or cGMP.
Collapse
Affiliation(s)
- Gabriela Maria Bertollotto
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | | | - Eduardo Costa Alexandre
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Fabiano Beraldi Calmasini
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Gabriela Reolon Passos
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Edson Antunes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Fabiola Zakia Mónica
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
21
|
Liu YT, Liu W, Zhu GY, Wang FL, Chen Q. Involvement of multidrug resistance protein 4 in the hepatocyte efflux of lamivudine and entecavir. Mol Med Rep 2018; 17:7113-7121. [PMID: 29568871 PMCID: PMC5928661 DOI: 10.3892/mmr.2018.8779] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 05/19/2017] [Indexed: 12/12/2022] Open
Abstract
Multidrug resistance protein 4 (MRP4) is capable of transporting acyclic nucleotide phosphonates, but little is known about its role in lamivudine (LAM) and entecavir (ETV) transport. In the present study, the involvement of MRP4 in the transport of LAM and ETV was investigated through in vitro experiments. The cytotoxicity of three antiviral drugs and their activities against HBV as characterized in HepG2.4D14 [wild‑type hepatitis B virus (HBV)] and HepG2.A64 (ETV‑resistant HBV) cells. LAM, ETV and tenofovir (TFV) demonstrated a 50% effective concentration against HBV of 4.14±0.03, 0.13±0.02 and 3.24±0.01 µM in HepG2.4D14 cells and of 5.94±0.20, 6.28±0.07 and 11.43±0.09 µM in HepG2.A64 cells, respectively. After administering 3-([(3-(2-[7-chloro-2-quinolinyl]ethyl)phenyl]-[(3-dimethylamino-3-oxoporphyl)-thio)-methyl]-thio) propanoic acid (MK571), the intracellular concentrations of all three drugs were much lower than the extracellular drug concentrations in these two cell types, whereas the intracellular drug concentrations in wild‑type cells were higher than those in ETV‑resistant cells. Furthermore, the intracellular levels of LAM, ETV and TFV were enhanced and the extracellular concentrations were reduced by addition of MK571. Thus, MRP4 is mainly responsible for the efflux of LAM and ETV in hepatocyte cultures. These results may contribute to enhancing antiviral efficacy.
Collapse
Affiliation(s)
- Yu-Tian Liu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wei Liu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Gang-Yan Zhu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fu-Liang Wang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qian Chen
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
22
|
Ahmad AA, Randall MD, Roberts RE. Sex differences in the role of phospholipase A 2 -dependent arachidonic acid pathway in the perivascular adipose tissue function in pigs. J Physiol 2017; 595:6623-6634. [PMID: 28877347 DOI: 10.1113/jp274831] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/21/2017] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS The fat surrounding blood vessels (perivascular adipose tissue or PVAT) releases vasoactive compounds that regulate vascular smooth muscle tone. There are sex differences in the regulation of vascular tone, but, to date, no study has investigated whether there are sex differences in the regulation of blood vessel tone by PVAT. This study has identified that the cyclooxygenase products thromboxane and PGF2α are released from coronary artery PVAT from pigs. Thromboxane appears to mediate the PVAT-induced contraction in arteries from females, whereas PGF2α appears to mediate the contraction in arteries from males. These sex differences in the role of these prostanoids in the PVAT-induced contraction can be explained by a greater release of thromboxane from PVAT from female animals and greater sensitivity to PGF2α in the porcine coronary artery from males. ABSTRACT Previous studies have demonstrated that perivascular adipose tissue (PVAT) causes vasoconstriction. In this present study, we determined the role of cyclooxygenase-derived prostanoids in this contractile response and determined whether there were any sex differences in the regulation of vascular tone by PVAT. Contractions in isolated segments of coronary arteries were determined using isolated tissue baths and isometric tension recording. Segments were initially cleaned of PVAT, which was then re-added to the tissue bath and changes in tone measured over 1 h. Levels of PGF2α and thromboxane B2 (TXB2 ) were quantified by ELISA, and PGF2α (FP) and thromboxane A2 (TP) receptor expression determined by Western blotting. In arteries from both male and female pigs, re-addition of PVAT caused a contraction, which was partially inhibited by the cyclooxygenase inhibitors indomethacin and flurbiprofen. The FP receptor antagonist AL8810 attenuated the PVAT-induced contraction in arteries from males, whereas the TP receptor antagonist GR32191B inhibited the PVAT-induced contraction in arteries from females. Although there was no difference in PGF2α levels in PVAT between females and males, PGF2α produced a larger contraction in arteries from males, correlating with a higher FP receptor expression. In contrast, release of TXB2 from PVAT from females was greater than from males, but there was no difference in the contraction by the TXA2 agonist U46619, or TP receptor expression in arteries from different sexes. These findings demonstrate clear sex differences in PVAT function in which PGF2α and TXA2 antagonists can inhibit the PVAT-induced vasoconstriction in male and female PCAs, respectively.
Collapse
Affiliation(s)
- Abdulla A Ahmad
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Michael D Randall
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Richard E Roberts
- Cell Signalling and Pharmacology Research Group, School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| |
Collapse
|
23
|
Bloise E, Ortiga-Carvalho TM, Reis FM, Lye SJ, Gibb W, Matthews SG. ATP-binding cassette transporters in reproduction: a new frontier. Hum Reprod Update 2015; 22:164-81. [PMID: 26545808 DOI: 10.1093/humupd/dmv049] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/19/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The transmembrane ATP-binding cassette (ABC) transporters actively efflux an array of clinically relevant compounds across biological barriers, and modulate biodistribution of many physiological and pharmacological factors. To date, over 48 ABC transporters have been identified and shown to be directly and indirectly involved in peri-implantation events and fetal/placental development. They efflux cholesterol, steroid hormones, vitamins, cytokines, chemokines, prostaglandins, diverse xenobiotics and environmental toxins, playing a critical role in regulating drug disposition, immunological responses and lipid trafficking, as well as preventing fetal accumulation of drugs and environmental toxins. METHODS This review examines ABC transporters as important mediators of placental barrier functions and key reproductive processes. Expression, localization and function of all identified ABC transporters were systematically reviewed using PubMed and Google Scholar websites to identify relevant studies examining ABC transporters in reproductive tissues in physiological and pathophysiological states. Only reports written in English were incorporated with no restriction on year of publication. While a major focus has been placed on the human, extensive evidence from animal studies is utilized to describe current understanding of the regulation and function of ABC transporters relevant to human reproduction. RESULTS ABC transporters are modulators of steroidogenesis, fertilization, implantation, nutrient transport and immunological responses, and function as 'gatekeepers' at various barrier sites (i.e. blood-testes barrier and placenta) against potentially harmful xenobiotic factors, including drugs and environmental toxins. These roles appear to be species dependent and change as a function of gestation and development. The best-described ABC transporters in reproductive tissues (primarily in the placenta) are the multidrug transporters p-glycoprotein and breast cancer-related protein, the multidrug resistance proteins 1 through 5 and the cholesterol transporters ABCA1 and ABCG1. CONCLUSIONS The ABC transporters have various roles across multiple reproductive tissues. Knowledge of efflux direction, tissue distribution, substrate specificity and regulation of the ABC transporters in the placenta and other reproductive tissues is rapidly expanding. This will allow better understanding of the disposition of specific substrates within reproductive tissues, and facilitate development of novel treatments for reproductive disorders as well as improved approaches to protecting the developing fetus.
Collapse
Affiliation(s)
- E Bloise
- Laboratory of Translational Endocrinology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - T M Ortiga-Carvalho
- Laboratory of Translational Endocrinology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - F M Reis
- Division of Human Reproduction, Department of Obstetrics and Gynecology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - S J Lye
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, Canada M5S 1A8 Department Obstetrics & Gynecology, University of Toronto, Toronto, ON, Canada Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - W Gibb
- Department of Obstetrics & Gynecology, University of Ottawa, Ottawa, ON, Canada Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - S G Matthews
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, Canada M5S 1A8 Department Obstetrics & Gynecology, University of Toronto, Toronto, ON, Canada Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
24
|
Research Progress on the Role of ABC Transporters in the Drug Resistance Mechanism of Intractable Epilepsy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:194541. [PMID: 26491660 PMCID: PMC4600483 DOI: 10.1155/2015/194541] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 03/04/2015] [Accepted: 03/05/2015] [Indexed: 12/17/2022]
Abstract
The pathogenesis of intractable epilepsy is not fully clear. In recent years, both animal and clinical trials have shown that the expression of ATP-binding cassette (ABC) transporters is increased in patients with intractable epilepsy; additionally, epileptic seizures can lead to an increase in the number of sites that express ABC transporters. These findings suggest that ABC transporters play an important role in the drug resistance mechanism of epilepsy. ABC transporters can perform the funcions of a drug efflux pump, which can reduce the effective drug concentration at epilepsy lesions by reducing the permeability of the blood brain barrier to antiepileptic drugs, thus causing resistance to antiepileptic drugs. Given the important role of ABC transporters in refractory epilepsy drug resistance, antiepileptic drugs that are not substrates of ABC transporters were used to obtain ABC transporter inhibitors with strong specificity, high safety, and few side effects, making them suitable for long-term use; therefore, these drugs can be used for future clinical treatment of intractable epilepsy.
Collapse
|
25
|
Wen J, Luo J, Huang W, Tang J, Zhou H, Zhang W. The Pharmacological and Physiological Role of Multidrug-Resistant Protein 4. J Pharmacol Exp Ther 2015; 354:358-375. [PMID: 26148856 DOI: 10.1124/jpet.115.225656] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 06/30/2015] [Indexed: 12/11/2022] Open
Abstract
Multidrug-resistant protein 4 (MRP4), a member of the C subfamily of ATP-binding cassette transporters, is distributed in a variety of tissues and a number of cancers. As a drug transporter, MRP4 is responsible for the pharmacokinetics and pharmacodynamics of numerous drugs, especially antiviral drugs, antitumor drugs, and diuretics. In this regard, the functional role of MRP4 is affected by a number of factors, such as genetic mutations; tissue-specific transcriptional regulations; post-transcriptional regulations, including miRNAs and membrane internalization; and substrate competition. Unlike other C family members, MRP4 is in a pivotal position to transport cellular signaling molecules, through which it is tightly connected to the living activity and physiologic processes of cells and bodies. In the context of several cancers in which MRP4 is overexpressed, MRP4 inhibition shows striking effects against cancer progression and drug resistance. In this review, we describe the role of MRP4 more specifically in both healthy conditions and disease states, with an emphasis on its potential as a drug target.
Collapse
Affiliation(s)
- Jiagen Wen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| | - Jianquan Luo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| | - Jie Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| |
Collapse
|
26
|
Large RJ, Kshatri A, Webb TI, Roy S, Akande A, Bradley E, Sergeant GP, Thornbury KD, McHale NG, Hollywood MA. Effects of the novel BK (KCa 1.1) channel opener GoSlo-SR-5-130 are dependent on the presence of BKβ subunits. Br J Pharmacol 2015; 172:2544-56. [PMID: 25598230 DOI: 10.1111/bph.13085] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 12/12/2014] [Accepted: 01/02/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND PURPOSE GoSlo-SR compounds are efficacious BK (KCa 1.1) channel openers, but little is known about their mechanism of action or effect on bladder contractility. We examined the effects of two closely related compounds on BK currents and bladder contractions. EXPERIMENTAL APPROACH A combination of electrophysiology, molecular biology and synthetic chemistry was used to examine the effects of two novel channel agonists on BK channels from bladder smooth muscle cells and in HEK cells expressing BKα alone or in combination with either β1 or β4 subunits. KEY RESULTS GoSlo-SR-5-6 shifted the voltage required for half maximal activation (V1/2 ) of BK channels approximately -100 mV, irrespective of the presence of regulatory β subunits. The deaminated derivative, GoSlo-SR-5-130, also shifted the activation V1/2 in smooth muscle cells by approximately -100 mV; however, this was reduced by ∼80% in HEK cells expressing only BKα subunits. When β1 or β4 subunits were co-expressed with BKα, efficacy was restored. GoSlo-SR-5-130 caused a concentration-dependent reduction in spontaneous bladder contraction amplitude and this was abolished by iberiotoxin, consistent with an effect on BK channels. CONCLUSIONS AND IMPLICATIONS GoSlo-SR-5-130 required β1 or β4 subunits to mediate its full effects, whereas GoSlo-SR-5-6 worked equally well in the absence or presence of β subunits. GoSlo-SR-5-130 inhibited spontaneous bladder contractions by activating BK channels. The novel BK channel opener, GoSlo-SR-5-130, is approximately fivefold more efficacious on BK channels with regulatory β subunits and may be a useful scaffold in the development of drugs to treat diseases such as overactive bladder.
Collapse
Affiliation(s)
- R J Large
- The Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Yu DMT, Huynh T, Truong AM, Haber M, Norris MD. ABC transporters and neuroblastoma. Adv Cancer Res 2015; 125:139-70. [PMID: 25640269 DOI: 10.1016/bs.acr.2014.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Neuroblastoma is the most common cancer of infancy and accounts for 15% of all pediatric oncology deaths. Survival rates of high-risk neuroblastoma remain less than 50%, with amplification of the MYCN oncogene the most important aberration associated with poor outcome. Direct transcriptional targets of MYCN include a number of ATP-binding cassette (ABC) transporters, of which ABCC1 (MRP1), ABCC3 (MRP3), and ABCC4 (MRP4) are the best characterized. These three transporter genes have been shown to be strongly prognostic of neuroblastoma outcome in primary untreated neuroblastoma. In addition to their ability to efflux a number of chemotherapeutic drugs, evidence suggests that these transporters also contribute to neuroblastoma outcome independent of any role in cytotoxic drug efflux. Endogenous substrates of ABCC1 and ABCC4 that may be potential candidates affecting neuroblastoma biology include molecules such as prostaglandins and leukotrienes. These bioactive lipid mediators have the ability to influence biological processes contributing to cancer initiation and progression, such as angiogenesis, cell signaling, inflammation, proliferation, and migration and invasion. ABCC1 and ABCC4 are thus potential targets for therapeutic suppression in high-risk neuroblastoma, and recently developed small-molecule inhibitors may be an effective strategy in treating aggressive forms of this cancer, as well as other cancers that express high levels of these transporters.
Collapse
Affiliation(s)
- Denise M T Yu
- Lowy Cancer Research Centre, Children's Cancer Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Tony Huynh
- Lowy Cancer Research Centre, Children's Cancer Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Alan M Truong
- Lowy Cancer Research Centre, Children's Cancer Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Michelle Haber
- Lowy Cancer Research Centre, Children's Cancer Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Murray D Norris
- Lowy Cancer Research Centre, Children's Cancer Institute, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
28
|
Ventimiglia MS, Najenson AC, Perazzo JC, Carozzo A, Vatta MS, Davio CA, Bianciotti LG. Blockade of Multidrug Resistance-Associated Proteins Aggravates Acute Pancreatitis and Blunts Atrial Natriuretic Factor's Beneficial Effect in Rats: Role of MRP4 (ABCC4). Mol Med 2015; 21:58-67. [PMID: 25569802 PMCID: PMC4461582 DOI: 10.2119/molmed.2014.00166] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 01/05/2015] [Indexed: 12/23/2022] Open
Abstract
We previously reported that atrial natriuretic factor (ANF) stimulates secretin-evoked cAMP efflux through multidrug resistance-associated protein 4 (MRP4) in the exocrine pancreas. Here we sought to establish in vivo whether this mechanism was involved in acute pancreatitis onset in the rat. Rats pretreated with or without probenecid (MRPs general inhibitor) were infused with secretin alone or with ANF. A set of these animals were given repetitive cerulein injections to induce acute pancreatitis. Plasma amylase and intrapancreatic trypsin activities were measured and histological examination of the pancreas performed. Secretin alone activated trypsinogen but induced no pancreatic histological changes. Blockade by probenecid in secretin-treated rats increased trypsin and also induced vacuolization, a hallmark of acute pancreatitis. ANF prevented the secretin response but in the absence of probenecid. In rats with acute pancreatitis, pretreatment with secretin aggravated the disease, but ANF prevented secretin-induced changes. Blockade of MRPs in rats with acute pancreatitis induced trypsinogen activation and larger cytoplasmic vacuoles as well as larger areas of necrosis and edema that were aggravated by secretin but not prevented by ANF. The temporal resolution of intracellular cAMP levels seems critical in the onset of acute pancreatitis, since secretin-evoked cAMP in a context of MRP inhibition makes the pancreas prone to injury in normal rats and aggravates the onset of acute pancreatitis. Present findings support a protective role for ANF mediated by cAMP extrusion through MRP4 and further suggest that the regulation of MRP4 by ANF would be relevant to maintain pancreatic acinar cell homeostasis.
Collapse
Affiliation(s)
- María Silvia Ventimiglia
- Cátedra de Fisiopatología, Instituto de Inmunología, Genética y Metabolismo (INIGEM-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana Clara Najenson
- Cátedra de Fisiopatología, Instituto de Inmunología, Genética y Metabolismo (INIGEM-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Carlos Perazzo
- Cátedra de Fisiopatología, Instituto de Inmunología, Genética y Metabolismo (INIGEM-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro Carozzo
- Laboratorio de Farmacología de Receptores, Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marcelo S Vatta
- Cátedra de Fisiología-Instituto de Química y Metabolismo del Fármaco (IQUIMEFA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos A Davio
- Laboratorio de Farmacología de Receptores, Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Liliana G Bianciotti
- Cátedra de Fisiopatología, Instituto de Inmunología, Genética y Metabolismo (INIGEM-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
29
|
Tanaka N, Yamaguchi H, Mano N. Transport of eicosapentaenoic acid-derived PGE₃, PGF(3α), and TXB₃ by ABCC4. PLoS One 2014; 9:e109270. [PMID: 25275481 PMCID: PMC4183643 DOI: 10.1371/journal.pone.0109270] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 09/08/2014] [Indexed: 01/17/2023] Open
Abstract
Background Eicosapentaenoic acid-derived prostaglandin (PG) E3, PGF3α, and thromboxane (TX) B3 are bioactive lipid mediators which have anti-cancer and anti-inflammatory effects. To exert their effects, PGE3, PGF3α, and TXB3 must be released to the extracellular space from cells, but the release mechanism has been unclear. We therefore investigated the contribution of ATP-binding cassette transporter C4 (ABCC4), which has been known as a prostanoids efflux transporter, to the release of PGE3, PGF3α, and TXB3. Materials and Methods ATP-dependent transport of PGE3, PGF3α, and TXB3 via ABCC4 was investigated by using inside-out membrane vesicles prepared from ABCC4-overexpressing HEK293 cells. To evaluate the contribution of ABCC4 to the release of PGE3, PGF3α, and TXB3, we measured the extracellular and intracellular levels of PGE3, PGF3α, and TXB3 in A549 cells when we used ABCC4 inhibitors (dipyridamole, MK571, and probenecid) or ABCC4 siRNAs. The quantification of PGE3, PGF3α, and TXB3 was performed by using liquid chromatography-tandem mass spectrometry. Results The apparent Km values for ABCC4-mediated transport were 2.9±0.1 µM for PGE3, 12.1±1.3 µM for PGF3α, and 11.9±1.4 µM for TXB3 and the ATP-dependent accumulation of PGE3, PGF3α, and TXB3 into vesicles was decreased by using typical substrates and inhibitors of ABCC4. ABCC4 inhibitors and ABCC4 knockdown showed the reduction of extracellular/intracellular ratio of PGE3 (40–60% of control) and PGF3α (60–80% of control) in A549 cells. Conclusions Our results suggest that PGE3, PGF3α, and TXB3 are substrates of ABCC4 and ABCC4 partially contributes to the release of PGE3 and PGF3α.
Collapse
Affiliation(s)
- Nobuaki Tanaka
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| | - Hiroaki Yamaguchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
- * E-mail:
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
30
|
Tanaka N, Yamaguchi H, Furugen A, Ogura J, Kobayashi M, Yamada T, Mano N, Iseki K. Quantification of intracellular and extracellular eicosapentaenoic acid-derived 3-series prostanoids by liquid chromatography/electrospray ionization tandem mass spectrometry. Prostaglandins Leukot Essent Fatty Acids 2014; 91:61-71. [PMID: 24996760 DOI: 10.1016/j.plefa.2014.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 03/18/2014] [Accepted: 04/19/2014] [Indexed: 01/31/2023]
Abstract
3-Series prostanoids are bioactive lipid mediators synthesized from eicosapentaenoic acid (EPA). Determination of intracellular and extracellular levels of prostanoids is needed to elucidate the mechanism of action, and we therefore developed a method for quantification of intracellular and extracellular levels of 3-series prostanoids (including prostaglandin E3 (PGE3), PGD3, PGF3α, thromboxane B3 (TXB3), and Δ(17)-6-keto PGF1α) by using liquid chromatography/electrospray ionization tandem mass spectrometry. The separation of prostanoids was performed with a CAPCELL PAK C18 MG II column (2.0mm×150mm, 3µm) with an isocratic flow of acetonitrile/water/acetic acid (40:60:0.1, v/v/v). This method was validated for measurement of both extracellular and intracellular samples with high levels of precision and accuracy. We applied this method to human lung epithelial A549 cells stimulated with calcium ionophore A23187 under the condition of arachidonic acid or EPA treatment and we could measure PGE3 in both intracellular and extracellular samples.
Collapse
Affiliation(s)
- Nobuaki Tanaka
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Hiroaki Yamaguchi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Ayako Furugen
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Jiro Ogura
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Takehiro Yamada
- Department of Pharmacy, Hokkaido University Hospital, Sapporo 060-8648, Japan
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Ken Iseki
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan; Department of Pharmacy, Hokkaido University Hospital, Sapporo 060-8648, Japan.
| |
Collapse
|
31
|
Tachikawa M, Hosoya KI, Terasaki T. Pharmacological significance of prostaglandin E2 and D2 transport at the brain barriers. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 71:337-60. [PMID: 25307222 DOI: 10.1016/bs.apha.2014.06.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Prostaglandin (PG) E2 and PGD2, which are biosynthesized from arachidonic acid generated by enzymatic cleavage of membrane phospholipid in response to various stimuli, play key roles in multiple brain pathophysiological processes, including modulation of synaptic plasticity, neuroinflammation, and sleep promotion. Concentrations of PGE2 and PGD2 in brain interstitial fluid (ISF) and cerebrospinal fluid (CSF) are maintained at appropriate levels for normal brain function by regulatory systems. The blood-brain barrier (BBB) and the blood-CSF barrier (BCSFB) possess ISF/CSF-to-blood efflux transport systems that are the primary cerebral clearance pathways for PGE2 and PGD2. However, regulatory dysfunction at the brain barriers may seriously affect brain function. In a mouse inflammation model, significant reduction of PGE2 efflux transport at the BBB has been observed. Several kinds of cephalosporin antibiotics and nonsteroidal anti-inflammatory drugs inhibit the BBB- and BCSFB-mediated efflux transport of PGE2 and PGD2. Especially, drugs that inhibit multidrug resistance-associated protein 4 (MRP4)-mediated PGE2 transport are capable of reducing PGE2 efflux at the BBB. Thus, it might be important in the treatment of inflammatory and infectious diseases to use drugs that do not inhibit clearance of PGE2 at the brain barriers, in order to avoid unexpected adverse CNS effects. Further, considering that PGD2 in CSF is a natural sleep-promoting factor, changes in the activity of the PGD2 efflux transport system at the BCSFB may modify the PGD2 level in CSF, thus affecting physiological sleep. These findings indicate that the efflux transport systems at the brain barriers play key roles in the pathophysiology and pharmacology of PGE2 and PGD2.
Collapse
Affiliation(s)
- Masanori Tachikawa
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ken-ichi Hosoya
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Tetsuya Terasaki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
32
|
Furugen A, Yamaguchi H, Tanaka N, Shiida N, Ogura J, Kobayashi M, Iseki K. Contribution of multidrug resistance-associated proteins (MRPs) to the release of prostanoids from A549 cells. Prostaglandins Other Lipid Mediat 2013; 106:37-44. [DOI: 10.1016/j.prostaglandins.2013.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 08/07/2013] [Accepted: 08/19/2013] [Indexed: 11/29/2022]
|
33
|
Shirasaka Y, Shichiri M, Kasai T, Ohno Y, Nakanishi T, Hayashi K, Nishiura A, Tamai I. A role of prostaglandin transporter in regulating PGE₂ release from human bronchial epithelial BEAS-2B cells in response to LPS. J Endocrinol 2013; 217:265-74. [PMID: 23528477 DOI: 10.1530/joe-12-0339] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Naturally occurring prostaglandin E₂ (PGE₂) plays a role in inflammatory responses through eicosanoid signaling pathways. PGE₂ is impermeable to cell membranes at physiological pH and needs solute carrier across the membranes; however, it remains unclear how intercellular concentrations of PGE₂ are regulated under the condition of inflammation. We aimed to clarify a role of organic anion-transporting polypeptide 2A1 (OATP2A1/SLCO2A1), also known as prostaglandin transporter (PGT), in PGE₂ release from cells. Human bronchial epithelial BEAS-2B cells were treated with lipopolysaccharide (LPS), and PGT inhibitors were tested to evaluate contribution of PGT to PGE₂ release by assessing its extracellular concentration and characterizing PGT-mediated PGE₂ efflux in Xenopus laevis oocytes. As a result, LPS elevated mRNA expression of a pro-inflammatory cytokine IL6 and extracellular concentration of PGE₂ in human bronchial epithelial BEAS-2B cells. PGT inhibitors tested (e.g. bromocresol green (BCG), bromosulfophthalein (BSP), and PGB₁) significantly inhibited efflux of PGE₂ from oocytes expressing PGT. Similarly, the amount of released PGE2 from the BEAS-2B cells decreased in the presence of BCG and BSP by 45 and 44% respectively while TGBz increased the concentration by 71%, suggesting that PGT mediates the release. In conclusion, these results imply a role of PGT in regulating intra- and extracellular concentrations of PGE₂ in response to cells under inflammatory conditions.
Collapse
Affiliation(s)
- Yoshiyuki Shirasaka
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Pharmacogenetics of chronic pain and its treatment. Mediators Inflamm 2013; 2013:864319. [PMID: 23766564 PMCID: PMC3671679 DOI: 10.1155/2013/864319] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 04/12/2013] [Indexed: 12/20/2022] Open
Abstract
This paper reviews the impact of genetic variability of drug metabolizing enzymes, transporters, receptors, and pathways involved in chronic pain perception on the efficacy and safety of analgesics and other drugs used for chronic pain treatment. Several candidate genes have been identified in the literature, while there is usually only limited clinical evidence substantiating for the penetration of the testing for these candidate biomarkers into the clinical practice. Further, the pain-perception regulation and modulation are still not fully understood, and thus more complex knowledge of genetic and epigenetic background for analgesia will be needed prior to the clinical use of the candidate genetic biomarkers.
Collapse
|
35
|
Seo H, Choi Y, Shim J, Choi Y, Ka H. Regulatory Mechanism for Expression of IL1B Receptors in the Uterine Endometrium and Effects of IL1B on Prostaglandin Synthetic Enzymes During the Implantation Period in Pigs1. Biol Reprod 2012; 87:31. [DOI: 10.1095/biolreprod.112.099051] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
36
|
Abstract
Platelets store signaling molecules (eg, serotonin and ADP) within their granules. Transporters mediate accumulation of these molecules in platelet granules and, on platelet activation, their translocation across the plasma membrane. The balance between transporter-mediated uptake and elimination of signaling molecules and drugs in platelets determines their intracellular concentrations and effects. Several members of the 2 major transporter families, ATP-binding cassette (ABC) transporters and solute carriers (SLCs), have been identified in platelets. An example of an ABC transporter is MRP4 (ABCC4), which facilitates ADP accumulation in dense granules. MRP4 is a versatile transporter, and various additional functions have been proposed, notably lipid mediator release and a role in aspirin resistance. Several other ABC proteins have been detected in platelets with functions in glutathione and lipid homeostasis. The serotonin transporter (SERT, SLC6A4) in the platelet plasma membrane represents a well-characterized example of the SLC family. Moreover, recent experiments indicate expression of OATP2B1 (SLCO2B1), a high affinity transporter for certain statins, in platelets. Changes in transporter localization and expression can affect platelet function and drug sensitivity. This review summarizes available data on the physiologic and pharmacologic role of transporters in platelets.
Collapse
|
37
|
Abstract
The ATP-binding cassette (ABC) transporters are a superfamily of membrane proteins that are best known for their ability to transport a wide variety of exogenous and endogenous substances across membranes against a concentration gradient via ATP hydrolysis. There are seven subfamilies of human ABC transporters, one of the largest being the 'C' subfamily (gene symbol ABCC). Nine ABCC subfamily members, the so-called multidrug resistance proteins (MRPs) 1-9, have been implicated in mediating multidrug resistance in tumor cells to varying degrees as the efflux extrude chemotherapeutic compounds (or their metabolites) from malignant cells. Some of the MRPs are also known to either influence drug disposition in normal tissues or modulate the elimination of drugs (or their metabolites) via hepatobiliary or renal excretory pathways. In addition, the cellular efflux of physiologically important organic anions such as leukotriene C(4) and cAMP is mediated by one or more of the MRPs. Finally, mutations in several MRPs are associated with human genetic disorders. In this minireview, the current biochemical and physiological knowledge of MRP1-MRP9 in cancer chemotherapy and human genetic disease is summarized. The mutations in MRP2/ABCC2 leading to conjugated hyperbilirubinemia (Dubin-Johnson syndrome) and in MRP6/ABCC6 leading to the connective tissue disorder Pseudoxanthoma elasticum are also discussed.
Collapse
Affiliation(s)
- Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, Queens, NY 11439, USA.
| | | |
Collapse
|
38
|
Keppler D. Multidrug resistance proteins (MRPs, ABCCs): importance for pathophysiology and drug therapy. Handb Exp Pharmacol 2011:299-323. [PMID: 21103974 DOI: 10.1007/978-3-642-14541-4_8] [Citation(s) in RCA: 216] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The nine multidrug resistance proteins (MRPs) represent the major part of the 12 members of the MRP/CFTR subfamily belonging to the 48 human ATP-binding cassette (ABC) transporters. Cloning, functional characterization, and cellular localization of most MRP subfamily members have identified them as ATP-dependent efflux pumps with a broad substrate specificity for the transport of endogenous and xenobiotic anionic substances localized in cellular plasma membranes. Prototypic substrates include glutathione conjugates such as leukotriene C(4) for MRP1, MRP2, and MRP4, bilirubin glucuronosides for MRP2 and MRP3, and cyclic AMP and cyclic GMP for MRP4, MRP5, and MRP8. Reduced glutathione (GSH), present in living cells at millimolar concentrations, modifies the substrate specificities of several MRPs, as exemplified by the cotransport of vincristine with GSH by MRP1, or by the cotransport of GSH with bile acids or of GSH with leukotriene B(4) by MRP4.The role of MRP subfamily members in pathophysiology may be illustrated by the MRP-mediated release of proinflammatory and immunomodulatory mediators such as leukotrienes and prostanoids. Pathophysiological consequences of many genetic variants leading to a lack of functional MRP protein in the plasma membrane are observed in the hereditary MRP2 deficiency associated with conjugated hyperbilirubinemia in Dubin-Johnson syndrome, in pseudoxanthoma elasticum due to mutations in the MRP6 (ABCC6) gene, or in the type of human earwax and osmidrosis determined by single nucleotide polymorphisms in the MRP8 (ABCC8) gene. The hepatobiliary and renal elimination of many drugs and their metabolites is mediated by MRP2 in the hepatocyte canalicular membrane and by MRP4 as well as MRP2 in the luminal membrane of kidney proximal tubules. Therefore, inhibition of these efflux pumps affects pharmacokinetics, unless compensated by other ATP-dependent efflux pumps with overlapping substrate specificities.
Collapse
|
39
|
Rius M, Keller D, Brom M, Hummel-Eisenbeiss J, Lyko F, Keppler D. Vectorial transport of nucleoside analogs from the apical to the basolateral membrane in double-transfected cells expressing the human concentrative nucleoside transporter hCNT3 and the export pump ABCC4. Drug Metab Dispos 2010; 38:1054-63. [PMID: 20360301 DOI: 10.1124/dmd.110.032664] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The identification of the transport proteins responsible for the uptake and the efflux of nucleosides and their metabolites enables the characterization of their vectorial transport and a better understanding of their absorption, distribution, and elimination. Human concentrative nucleoside transporters (hCNTs/SLC28A) are known to mediate the transport of natural nucleosides and some nucleoside analogs into cells in a sodium-dependent and unidirectional manner. On the other hand, several human multidrug resistance proteins [human ATP-binding cassette transporter, subfamily C (ABCC)] cause resistance against nucleoside analogs and mediate transport of phosphorylated nucleoside derivatives out of the cells in an ATP-dependent manner. For the integrated analysis of uptake and efflux of these compounds, we established a double-transfected Madin-Darby canine kidney (MDCK) II cell line stably expressing the human uptake transporter hCNT3 in the apical membrane and the human efflux pump ABCC4 in the basolateral membrane. The direction of transport was from the apical to the basolateral compartment, which is in line with the unidirectional transport and the localization of both recombinant proteins in the MDCKII cells. Recombinant hCNT3 mediated the transport of several known nucleoside substrates, and we identified 5-azacytidine as a new substrate for hCNT3. It is of interest that coexpression of both transporters was confirmed in pancreatic adenocarcinomas, which represent an important clinical indication for the therapeutic use of nucleoside analogs. Thus, our results establish a novel cell system for studies on the vectorial transport of nucleosides and their analogs from the apical to the basolateral compartment. The results contribute to a better understanding of the cellular transport characteristics of nucleoside drugs.
Collapse
Affiliation(s)
- Maria Rius
- Division of Tumor Biochemistry, German Cancer Research Center, D-69120 Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
40
|
Phillips RJ, Al-Zamil H, Hunt LP, Fortier MA, López Bernal A. Genes for prostaglandin synthesis, transport and inactivation are differentially expressed in human uterine tissues, and the prostaglandin F synthase AKR1B1 is induced in myometrial cells by inflammatory cytokines. Mol Hum Reprod 2010; 17:1-13. [PMID: 20595240 DOI: 10.1093/molehr/gaq057] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Prostaglandins (PGs) are important factors in the physiology of human parturition and the control of uterine contractility. We have characterized the expression of 15 genes from all stages of the PG pathway in human pregnant and non-pregnant (NP) myometrium and in other uterine tissues at delivery, and the results show patterns indicative of different capacities for PG synthesis and catabolism in each tissue. In placenta, the PG synthase expression profile favours production of PGD₂, PGE₂ and PGF₂, with high levels of PG transporters and catabolic PG dehydrogenase suggesting rapid PG turnover. Choriodecidua is primed for PGE₂, PGF₂ and PGD₂ production and high PG turnover, whereas amnion expresses genes for PGE₂ synthesis with low levels of PG transporters and dehydrogenase. In umbilical cord, PGI₂ synthase is highly expressed. In pregnant myometrium, PGI₂, PGD₂ and PGF₂ synthases are highly expressed, whereas PG dehydrogenase is underexpressed. Myometrium from women with spontaneous or induced labour had higher expression of the PGH₂ synthase PTGS2 than tissue from women not-in-labour. Myometrium from NP women had lower levels of PG synthases and higher levels of PG dehydrogenase than pregnant myometrium. Discriminant function analysis showed that expression of selected genes in myometrium could distinguish groups of women with different modes of labour from each other and from NP women. In cultured myometrial cells, there was a dose-dependent stimulatory effect of interleukin 1β and tumour necrosis factor α on PTGS2, PTGES and AKR1B1 (PGF synthase) expression.
Collapse
Affiliation(s)
- R J Phillips
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Clinical Science at South Bristol (Obstetrics and Gynaecology), University of Bristol, Dorothy Hodgkin Building, Bristol BS1 3NY, UK.
| | | | | | | | | |
Collapse
|
41
|
Omura N, Griffith M, Vincent A, Li A, Hong SM, Walter K, Borges M, Goggins M. Cyclooxygenase-deficient pancreatic cancer cells use exogenous sources of prostaglandins. Mol Cancer Res 2010; 8:821-32. [PMID: 20530583 PMCID: PMC2888921 DOI: 10.1158/1541-7786.mcr-09-0336] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genes that are differentially expressed in pancreatic cancers and under epigenetic regulation are of considerable biological and therapeutic interest. We used global gene expression profiling and epigenetic treatment of pancreatic cell lines including pancreatic cancer cell lines, pancreatic cancer-associated fibroblasts, and cell lines derived from nonneoplastic pancreata. We examined expression and epigenetic alterations of cyclooxygenase-1 (COX-1) and COX-2 in pancreatic cancers and normal pancreas and performed proliferation, knockdown, and coculture experiments to understand the role of stromal sources of prostaglandins for pancreatic cancers. We identify COX-1 as a gene under epigenetic regulation in pancreatic cancers. We find that COX-1 expression is absent in many pancreatic cancer cells and some of these cancers also lack COX-2 expression. Suspecting that such cancers must rely on exogenous sources of prostaglandins, we show that pancreatic cancer stromal cells, such as fibroblasts expressing COX-1 and COX-2, are a likely source of prostaglandins for pancreatic cancer cells deficient in COX. Knocking down the prostaglandin transporter multidrug resistance-associated protein-4 in fibroblasts suppresses the proliferation of cocultured pancreatic cancer cells lacking COX. Pancreatic cancers that lack COX can use exogenous sources of prostaglandins. Blocking multidrug resistance-associated protein-4 may be a useful therapeutic strategy to deplete COX-deficient pancreatic cancers of prostaglandins.
Collapse
Affiliation(s)
- Noriyuki Omura
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - Margaret Griffith
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - Audrey Vincent
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - Ang Li
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - Seung-Mo Hong
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - Kimberly Walter
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - Michael Borges
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
42
|
Klaassen CD, Aleksunes LM. Xenobiotic, bile acid, and cholesterol transporters: function and regulation. Pharmacol Rev 2010; 62:1-96. [PMID: 20103563 PMCID: PMC2835398 DOI: 10.1124/pr.109.002014] [Citation(s) in RCA: 582] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Transporters influence the disposition of chemicals within the body by participating in absorption, distribution, and elimination. Transporters of the solute carrier family (SLC) comprise a variety of proteins, including organic cation transporters (OCT) 1 to 3, organic cation/carnitine transporters (OCTN) 1 to 3, organic anion transporters (OAT) 1 to 7, various organic anion transporting polypeptide isoforms, sodium taurocholate cotransporting polypeptide, apical sodium-dependent bile acid transporter, peptide transporters (PEPT) 1 and 2, concentrative nucleoside transporters (CNT) 1 to 3, equilibrative nucleoside transporter (ENT) 1 to 3, and multidrug and toxin extrusion transporters (MATE) 1 and 2, which mediate the uptake (except MATEs) of organic anions and cations as well as peptides and nucleosides. Efflux transporters of the ATP-binding cassette superfamily, such as ATP-binding cassette transporter A1 (ABCA1), multidrug resistance proteins (MDR) 1 and 2, bile salt export pump, multidrug resistance-associated proteins (MRP) 1 to 9, breast cancer resistance protein, and ATP-binding cassette subfamily G members 5 and 8, are responsible for the unidirectional export of endogenous and exogenous substances. Other efflux transporters [ATPase copper-transporting beta polypeptide (ATP7B) and ATPase class I type 8B member 1 (ATP8B1) as well as organic solute transporters (OST) alpha and beta] also play major roles in the transport of some endogenous chemicals across biological membranes. This review article provides a comprehensive overview of these transporters (both rodent and human) with regard to tissue distribution, subcellular localization, and substrate preferences. Because uptake and efflux transporters are expressed in multiple cell types, the roles of transporters in a variety of tissues, including the liver, kidneys, intestine, brain, heart, placenta, mammary glands, immune cells, and testes are discussed. Attention is also placed upon a variety of regulatory factors that influence transporter expression and function, including transcriptional activation and post-translational modifications as well as subcellular trafficking. Sex differences, ontogeny, and pharmacological and toxicological regulation of transporters are also addressed. Transporters are important transmembrane proteins that mediate the cellular entry and exit of a wide range of substrates throughout the body and thereby play important roles in human physiology, pharmacology, pathology, and toxicology.
Collapse
Affiliation(s)
- Curtis D Klaassen
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160-7417, USA.
| | | |
Collapse
|
43
|
Fletcher JI, Haber M, Henderson MJ, Norris MD. ABC transporters in cancer: more than just drug efflux pumps. Nat Rev Cancer 2010; 10:147-56. [PMID: 20075923 DOI: 10.1038/nrc2789] [Citation(s) in RCA: 797] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Multidrug transporter proteins are best known for their contributions to chemoresistance through the efflux of anticancer drugs from cancer cells. However, a considerable body of evidence also points to their importance in cancer extending beyond drug transport to fundamental roles in tumour biology. Currently, much of the evidence for these additional roles is correlative and definitive studies are needed to confirm causality. We propose that delineating the precise roles of these transporters in tumorigenesis and treatment response will be important for the development of more effective targeted therapies.
Collapse
Affiliation(s)
- Jamie I Fletcher
- Children's Cancer Institute Australia for Medical Research, Lowry Cancer Research Centre, University of New South Wales, P.O. BOX 151, Randwick NSW 2031, Australia
| | | | | | | |
Collapse
|
44
|
Liu YH, Di YM, Zhou ZW, Mo SL, Zhou SF. Multidrug resistance-associated proteins and implications in drug development. Clin Exp Pharmacol Physiol 2009; 37:115-20. [PMID: 19566819 DOI: 10.1111/j.1440-1681.2009.05252.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
1. The multidrug resistance-associated proteins (MRPs) belong to the ATP-binding cassette superfamily (ABCC family) of transporters that are expressed differentially in the liver, kidney, intestine and blood-brain barrier. There are nine human MRPs that transport a structurally diverse array of endo- and xenobiotics as well as their conjugates. 2. Multidrug resistance-associated protein 1 can be distinguished from MRP2 and MRP3 by its higher affinity for leukotriene C(4). Unlike MRP1, MRP2 functions in the extrusion of endogenous organic anions, such as bilirubin glucuronide and certain anticancer agents. In addition to the transport of glutathione and glucuronate conjugates, MRP3 has the additional capability of mediating the transport of monoanionic bile acids. 3. Both MRP4 and MRP5 are able to mediate the transport of cyclic nucleotides and confer resistance to certain antiviral and anticancer nucleotide analogues. Hereditary deficiency of MRP6 results in pseudoxanthoma elasticum. In the body, MRP6 is involved in the transport of glutathione conjugates and the cyclic pentapeptide BQ123. 4. Various MRPs show considerable differences in tissue distribution, substrate specificity and proposed physiological function. These proteins play a role in drug disposition and excretion and thus are implicated in drug toxicity and drug interactions. Increased efflux of natural product anticancer drugs and other anticancer agents mediated by MRPs from cancer cells is associated with tumour resistance. 5. A better understanding of the function and regulating mechanisms of MRPs could help minimize and avoid drug toxicity and unfavourable drug-drug interactions, as well as help overcome drug resistance.
Collapse
Affiliation(s)
- Ya-He Liu
- Discipline of Chinese Medicine, School of Health Sciences, RMIT University, Melbourne, Victoria 3083, Australia
| | | | | | | | | |
Collapse
|
45
|
Radilova H, Libra A, Holasova S, Safarova M, Viskova A, Kunc F, Buncek M. COX-1 is coupled with mPGES-1 and ABCC4 in human cervix cancer cells. Mol Cell Biochem 2009; 330:131-40. [DOI: 10.1007/s11010-009-0126-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2008] [Accepted: 04/16/2009] [Indexed: 12/23/2022]
|
46
|
Nies AT, Schwab M, Keppler D. Interplay of conjugating enzymes with OATP uptake transporters and ABCC/MRP efflux pumps in the elimination of drugs. Expert Opin Drug Metab Toxicol 2008; 4:545-68. [PMID: 18484914 DOI: 10.1517/17425255.4.5.545] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Biliary excretion is a major elimination route of many drugs and their metabolites. Hepatobiliary elimination is a vectorial process involving uptake transporters in the basolateral hepatocyte membrane, possibly Phase I and Phase II metabolizing enzymes, and ATP-dependent efflux pumps in the apical hepatocyte membrane. OBJECTIVES Because many drugs and their metabolites are anions, this review focuses on transporters involved in their hepatocellular uptake (members of the organic anion transporting polypeptide (OATP) family) and biliary elimination (apical conjugate efflux pump ABCC2/MRP2). METHODS The molecular and functional characteristics of the human OATP and ABCC/MRP transporters are presented, including a detailed overview of endogenous and drug substrates. Examples illustrate the interplay of transporters with Phase II conjugating enzymes. Model systems to study the vectorial transport of organic anions are also discussed. RESULTS/CONCLUSIONS OATP uptake transporters, conjugating enzymes, and ABCC2/MRP2 work in concert to enable the hepatobiliary elimination of anionic drugs and their metabolites. It is increasingly important to understand how genetic variants of these transporters and enzymes influence the interindividual variability of drug elimination.
Collapse
Affiliation(s)
- Anne T Nies
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Auerbachstrasse 112, 70376 Stuttgart, Germany.
| | | | | |
Collapse
|
47
|
Ho LL, Kench JG, Handelsman DJ, Scheffer GL, Stricker PD, Grygiel JG, Sutherland RL, Henshall SM, Allen JD, Horvath LG. Androgen regulation of multidrug resistance-associated protein 4 (MRP4/ABCC4) in prostate cancer. Prostate 2008; 68:1421-9. [PMID: 18615486 DOI: 10.1002/pros.20809] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND MRP4/ABCC4 is an ATP-binding cassette transporter expressed in normal prostate. This study aimed to define the pattern of MRP4/ABCC4 expression in normal and malignant prostate tissue and the relationship of MRP4/ABCC4 expression and function in response to androgen signaling. METHODS Eighty-four radical prostatectomy specimens from patients with localized prostate cancer (PC) (22 neoadjuvant androgen ablation, AA, 62 no AA), 42 non-cancer and 16 advanced PCs were assessed for MRP4/ABCC4 mRNA/protein expression. The effect of DHT and bicalutamide on LNCaP cells was assessed by immunoblotting. HEK293 cells (+/-MRP4/ABCC4) were assessed for the ability to efflux androgens and anti-androgens. RESULTS MRP4/ABCC4 mRNA/protein levels were higher in localized PC compared to non-cancer (P = 0.006). MRP4/ABCC4 levels were significantly decreased in PCs treated with AA compared to cancers exposed to normal testosterone levels (P < 0.0001). MRP4/ABCC4 expression in normal human tissues was limited to the prostate and the renal tubules. MRP4/ABCC4 protein levels increased in LNCaP cells after DHT which was partially blocked by bicalutamide. However, DHT did not alter the activation of the MRP4/ABCC4 promotor in luciferase reporter assays and testosterone, DHT, flutamide and hydroxy-flutamide were not substrates for MRP4/ABCC4. DISCUSSION Elevated MRP4/ABCC4 expression is found in malignant compared to benign prostate tissue while lower MRP4/ABCC4 expression is seen after AA. Furthermore, MRP4/ABCC4 is upregulated by androgen and downregulated by anti-androgen treatment in vitro potentially through an indirect mode of action. These data strongly suggest that MRP4/ABCC4 is an androgen-regulated gene important in the progression to PC and may be a potential drug target.
Collapse
Affiliation(s)
- Lye Lin Ho
- Centenary Institute of Cancer Medicine and Cell Biology, Newtown, NSW 2042, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Aleksunes LM, Augustine LM, Scheffer GL, Cherrington NJ, Manautou JE. Renal xenobiotic transporters are differentially expressed in mice following cisplatin treatment. Toxicology 2008; 250:82-8. [PMID: 18640236 PMCID: PMC2570650 DOI: 10.1016/j.tox.2008.06.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Revised: 06/02/2008] [Accepted: 06/03/2008] [Indexed: 11/19/2022]
Abstract
The goal of this study was to identify alterations in mRNA and protein expression of various xenobiotic transport proteins in mouse kidney during cisplatin-induced acute renal failure. For this purpose, male C57BL/6J mice received a single dose of cisplatin (18 mg/kg, i.p.) or vehicle. Four days later, tissues were collected for assessment of plasma BUN, histopathological analysis of renal lesions, and mRNA and Western blot analysis of renal transporters including organic anion and cation transporters (Oat, Oct), organic anion transporting polypeptides (Oatp), multidrug resistance-associated proteins (Mrp), multidrug resistance proteins (Mdr), breast cancer resistance protein (Bcrp) and multidrug and toxin extrusion proteins (Mate). Cisplatin treatment caused necrosis of renal proximal tubules along with elevated plasma BUN and renal kidney injury molecule-1 mRNA expression. Cisplatin-induced renal injury increased mRNA and protein levels of the efflux transporters Mrp2, Mrp4, Mrp5, Mdr1a and Mdr1b. Uptake transporters Oatp2a1 and Oatp2b1 mRNA were also up-regulated following cisplatin. By contrast, expression of Oat1, Oat2, Oct2 and Oatp1a1 mRNA was reduced in cisplatin-treated mice. Expression of several uptake and efflux transporters was unchanged in cisplatin-treated mice. Apical staining of Mrp2 and Mrp4 proteins was enhanced in proximal tubules from cisplatin-treated mice. Collectively, these expression patterns suggest coordinated regulation of uptake and efflux pathways during cisplatin-induced renal injury. Reduced expression of basolateral and apical uptake transporters along with enhanced transcription of export transporters likely represents an adaptation to lower intracellular accumulation of chemicals, prevent their reabsorption and enhance urinary clearance.
Collapse
Affiliation(s)
- Lauren M Aleksunes
- University of Connecticut, Dept of Pharmaceutical Sciences, 69 North Eagleville Rd. U-3092, Storrs, CT, 06269-3092
| | - Lisa M Augustine
- University of Arizona, Dept of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 E. Mabel, Tucson, AZ 85721
| | - George L Scheffer
- VU Medical Center, Dept of Pathology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Nathan J Cherrington
- University of Arizona, Dept of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 E. Mabel, Tucson, AZ 85721
| | - José E Manautou
- University of Connecticut, Dept of Pharmaceutical Sciences, 69 North Eagleville Rd. U-3092, Storrs, CT, 06269-3092
| |
Collapse
|
49
|
Aleksunes LM, Cui Y, Klaassen CD. Prominent expression of xenobiotic efflux transporters in mouse extraembryonic fetal membranes compared with placenta. Drug Metab Dispos 2008; 36:1960-70. [PMID: 18566041 PMCID: PMC2574899 DOI: 10.1124/dmd.108.021337] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Fetal exposure to xenobiotics can be restricted by transporters at the interface between maternal and fetal circulation. Previous work identified transporters in the placenta; however, less is known about the presence of these transporters in the fetal membranes (i.e., yolk sac and amniotic membranes). The purpose of this study was to quantify mRNA and protein expression of xenobiotic transporters in mouse placenta and fetal membranes during mid to late gestation. Concepti (placenta and fetal membranes, gestation day 11) or placenta and fetal membranes (gestation days 14 and 17) were collected from pregnant mice and analyzed for expression of multidrug resistance-associated proteins (Mrps), multidrug resistance proteins (Mdrs), multidrug and toxin extrusion proteins (Mates), breast cancer resistance protein (Bcrp), and organic anion-transporting polypeptides (Oatps). Maternal liver and kidneys were also collected at day 14 for mRNA and immunohistochemical analysis. mRNA expression of Mrp, Mdr, Bcrp, Mate-1, and Oatp isoforms was detected at day 11. The uptake carriers Oatp2a1, 3a1, 4a1, and 5a1 showed placenta-predominant expression. At days 14 and 17, fetal membranes expressed higher mRNA levels of the efflux transporters Mrp2 (7-fold), Mrp4 (5-fold), Mrp5 (3-fold), Mrp6 (12-fold), Bcrp (2-fold), and Mate-1 (7-fold) than placenta. Western blot analysis of Mrp2, Mrp4, Mrp6, and Bcrp confirmed higher expression in fetal membranes. Immunostaining revealed apical (Mrp2 and Bcrp) and basolateral (Mrp4, 5, and 6) cellular localization in epithelial cells of the yolk sac. In conclusion, xenobiotic transporters in the fetal membranes may provide an additional route to protect the fetus against endogenous chemicals and xenobiotics.
Collapse
Affiliation(s)
- Lauren M Aleksunes
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160-7417, USA
| | | | | |
Collapse
|
50
|
Campion SN, Johnson R, Aleksunes LM, Goedken MJ, van Rooijen N, Scheffer GL, Cherrington NJ, Manautou JE. Hepatic Mrp4 induction following acetaminophen exposure is dependent on Kupffer cell function. Am J Physiol Gastrointest Liver Physiol 2008; 295:G294-304. [PMID: 18556419 PMCID: PMC2519859 DOI: 10.1152/ajpgi.00541.2007] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
During acetaminophen (APAP) hepatotoxicity, increased expression of multidrug resistance-associated proteins 2, 3, and 4 (Mrp2-4) occurs. Mrp4 is the most significantly upregulated transporter in mouse liver following APAP treatment. Although the expression profiles of liver transporters following APAP hepatotoxicity are well characterized, the regulatory mechanisms contributing to these changes remain unknown. We hypothesized that Kupffer cell-derived mediators participate in the regulation of hepatic transporters during APAP toxicity. To investigate this, C57BL/6J mice were pretreated with clodronate liposomes (0.1 ml iv) to deplete Kupffer cells and then challenged with APAP (500 mg/kg ip). Liver injury was assessed by plasma alanine aminotransferase and hepatic transporter protein expression was determined by Western blot and immunohistochemistry. Depletion of Kupffer cells by liposomal clodronate increased susceptibility to APAP hepatotoxicity. Although increased expression of several efflux transporters was observed after APAP exposure, only Mrp4 was found to be differentially regulated following Kupffer cell depletion. At 48 and 72 h after APAP dosing, Mrp4 levels were increased by 10- and 33-fold, respectively, in mice receiving empty liposomes. Immunohistochemistry revealed Mrp4 staining confined to centrilobular hepatocytes. Remarkably, Kupffer cell depletion completely prevented Mrp4 induction by APAP. Elevated plasma levels of TNF-alpha and IL-1beta were also prevented by Kupffer cell depletion. These findings show that Kupffer cells protect the liver from APAP toxicity and that Kupffer cell mediators released in response to APAP are likely responsible for the induction of Mrp4.
Collapse
Affiliation(s)
- Sarah N. Campion
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut; Department of Pathology, Schering Plough Research Institute, Lafayette, New Jersey; Departments of Molecular Cell Biology and Pathology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands; and Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona
| | - Rachel Johnson
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut; Department of Pathology, Schering Plough Research Institute, Lafayette, New Jersey; Departments of Molecular Cell Biology and Pathology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands; and Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona
| | - Lauren M. Aleksunes
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut; Department of Pathology, Schering Plough Research Institute, Lafayette, New Jersey; Departments of Molecular Cell Biology and Pathology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands; and Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona
| | - Michael J. Goedken
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut; Department of Pathology, Schering Plough Research Institute, Lafayette, New Jersey; Departments of Molecular Cell Biology and Pathology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands; and Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona
| | - Nico van Rooijen
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut; Department of Pathology, Schering Plough Research Institute, Lafayette, New Jersey; Departments of Molecular Cell Biology and Pathology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands; and Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona
| | - George L. Scheffer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut; Department of Pathology, Schering Plough Research Institute, Lafayette, New Jersey; Departments of Molecular Cell Biology and Pathology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands; and Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona
| | - Nathan J. Cherrington
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut; Department of Pathology, Schering Plough Research Institute, Lafayette, New Jersey; Departments of Molecular Cell Biology and Pathology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands; and Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona
| | - José E. Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut; Department of Pathology, Schering Plough Research Institute, Lafayette, New Jersey; Departments of Molecular Cell Biology and Pathology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands; and Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona
| |
Collapse
|