1
|
Carstens G, Verbeek MM, Rohlwink UK, Figaji AA, te Brake L, van Laarhoven A. Metabolite transport across central nervous system barriers. J Cereb Blood Flow Metab 2024; 44:1063-1077. [PMID: 38546534 PMCID: PMC11179608 DOI: 10.1177/0271678x241241908] [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: 07/31/2023] [Revised: 02/02/2024] [Accepted: 02/27/2024] [Indexed: 06/13/2024]
Abstract
Metabolomic analysis of cerebrospinal fluid (CSF) is used to improve diagnostics and pathophysiological understanding of neurological diseases. Alterations in CSF metabolite levels can partly be attributed to changes in brain metabolism, but relevant transport processes influencing CSF metabolite concentrations should be considered. The entry of molecules including metabolites into the central nervous system (CNS), is tightly controlled by the blood-brain, blood-CSF, and blood-spinal cord barriers, where aquaporins and membrane-bound carrier proteins regulate influx and efflux via passive and active transport processes. This report therefore provides reference for future CSF metabolomic work, by providing a detailed summary of the current knowledge on the location and function of the involved transporters and routing of metabolites from blood to CSF and from CSF to blood.
Collapse
Affiliation(s)
- Gesa Carstens
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Marcel M Verbeek
- Departments of Neurology and Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Netherlands
| | - Ursula K Rohlwink
- Division of Neurosurgery, Department of Surgery, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Anthony A Figaji
- Division of Neurosurgery, Department of Surgery, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Lindsey te Brake
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arjan van Laarhoven
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
2
|
Bastedo WE, Scott RW, Arostegui M, Underhill TM. Single-cell analysis of mesenchymal cells in permeable neural vasculature reveals novel diverse subpopulations of fibroblasts. Fluids Barriers CNS 2024; 21:31. [PMID: 38575991 PMCID: PMC10996213 DOI: 10.1186/s12987-024-00535-7] [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: 07/31/2023] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND In the choroid plexus and pituitary gland, vasculature is known to have a permeable, fenestrated phenotype which allows for the free passage of molecules in contrast to the blood brain barrier observed in the rest of the CNS. The endothelium of these compartments, along with secretory, neural-lineage cells (choroid epithelium and pituitary endocrine cells) have been studied in detail, but less attention has been given to the perivascular mesenchymal cells of these compartments. METHODS The Hic1CreERT2 Rosa26LSL-TdTomato mouse model was used in conjunction with a PdgfraH2B-EGFP mouse model to examine mesenchymal cells, which can be subdivided into Pdgfra+ fibroblasts and Pdgfra- pericytes within the choroid plexus (CP) and pituitary gland (PG), by histological, immunofluorescence staining and single-cell RNA-sequencing analyses. RESULTS We found that both CP and PG possess substantial populations of distinct Hic1+ mesenchymal cells, including an abundance of Pdgfra+ fibroblasts. Within the pituitary, we identified distinct subpopulations of Hic1+ fibroblasts in the glandular anterior pituitary and the neurosecretory posterior pituitary. We also identified multiple distinct markers of CP, PG, and the meningeal mesenchymal compartment, including alkaline phosphatase, indole-n-methyltransferase and CD34. CONCLUSIONS Novel, distinct subpopulations of mesenchymal cells can be found in permeable vascular interfaces, including the CP, PG, and meninges, and make distinct contributions to both organs through the production of structural proteins, enzymes, transporters, and trophic molecules.
Collapse
Affiliation(s)
- William E Bastedo
- Department of Cellular and Physiological Sciences, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - R Wilder Scott
- Department of Cellular and Physiological Sciences, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- School of Biomedical Engineering and the Biomedical Research Centre, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Martin Arostegui
- Department of Cellular and Physiological Sciences, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - T Michael Underhill
- Department of Cellular and Physiological Sciences, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
- School of Biomedical Engineering and the Biomedical Research Centre, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
3
|
Honan LE, Fraser-Spears R, Daws LC. Organic cation transporters in psychiatric and substance use disorders. Pharmacol Ther 2024; 253:108574. [PMID: 38072333 PMCID: PMC11052553 DOI: 10.1016/j.pharmthera.2023.108574] [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: 10/06/2023] [Revised: 11/01/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Psychiatric and substance use disorders inflict major public health burdens worldwide. Their widespread burden is compounded by a dearth of effective treatments, underscoring a dire need to uncover novel therapeutic targets. In this review, we summarize the literature implicating organic cation transporters (OCTs), including three subtypes of OCTs (OCT1, OCT2, and OCT3) and the plasma membrane monoamine transporter (PMAT), in the neurobiology of psychiatric and substance use disorders with an emphasis on mood and anxiety disorders, alcohol use disorder, and psychostimulant use disorder. OCTs transport monoamines with a low affinity but high capacity, situating them to play a central role in regulating monoamine homeostasis. Preclinical evidence discussed here suggests that OCTs may serve as promising targets for treatment of psychiatric and substance use disorders and encourage future research into their therapeutic potential.
Collapse
Affiliation(s)
- Lauren E Honan
- The University of Texas Health Science Center at San Antonio, Department of Cellular & Integrative Physiology, USA
| | - Rheaclare Fraser-Spears
- University of the Incarnate Word, Feik School of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Lynette C Daws
- The University of Texas Health Science Center at San Antonio, Department of Cellular & Integrative Physiology, USA; The University of Texas Health Science Center at San Antonio, Department of Pharmacology, USA.
| |
Collapse
|
4
|
Rios ACH, Nasner SLC, Londoño-Gil M, Gonzalez-Herrera LG, Lopez-Herrera A, Flórez JCR. Genome-wide association study for reproduction traits in Colombian Creole Blanco Orejinegro cattle. Trop Anim Health Prod 2023; 55:429. [PMID: 38044379 DOI: 10.1007/s11250-023-03847-2] [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: 06/20/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
The profitability of the beef cattle production system relies heavily on reproductive traits. Unfortunately, certain traits, such as age at first calving (AFC), calving interval (CI), and gestation length (GL), can pose challenges in traditional breeding programs because of their low heritability (0.01-0.12) and sex-limited characteristics. Another important aspect is the conservation of the genetic resources of animals adapted to the Colombian regions, which implies the preservation and rational use of the creole breeds in the country market. Therefore, this study aimed to identify genomic regions in the creole cattle breed Blanco Orejinegro (BON) that influence the reproductive traits in females. The dataset comprised 439 animals and 118,116 single-nucleotide polymorphisms' (SNPs) markers. The GS3 program was used to identify the SNP effects employing the BAYES Cπ methodology. The number of SNPs with effect for AFC was 25, 1527 for CI, and 23 for GL. Some of the genes found associated with reproductive and growth traits as well as immune response and environmental adaptation ECE1, EPH, EPHB2, SMARCAL1, IGFBP5, IGFBP2, FCGRT, EGFR, MUL1, PINK1, STPG1, CNGB1, TGFB1, OXTR, IL22RA1, MYOM3, OXTR, CNR2, HIVEP3, CTPS1, CXCL8, FCGRT, MREG, TMEM169, PECR, and MC1R. Our results evidenced a high contribution of the genetic architecture of the Colombian creole cattle breed Blanco Orejinegro that may impact should be included in implementing genetic improvement and conservation programs.
Collapse
Affiliation(s)
- Ana Cristina Herrera Rios
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Medellín, Carrera 65 N 59A-110, 050034, Medellín, Colombia.
- Grupo de Investigación Nutri-Solla, SOLLA S.A., Cra 42 #33-80, Itagüí, Antioquia, Colombia.
| | - Sindy Liliana Caivio Nasner
- Grupo de Investigación Biomolecular y Pecuaria BIOPEC, Universidad Tecnológica de Pereira, Cra. 27 N10-02, 660003, Pereira, Risaralda, Colombia
| | - Marisol Londoño-Gil
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Medellín, Carrera 65 N 59A-110, 050034, Medellín, Colombia
| | - Luis Gabriel Gonzalez-Herrera
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Medellín, Carrera 65 N 59A-110, 050034, Medellín, Colombia
| | - Albeiro Lopez-Herrera
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Medellín, Carrera 65 N 59A-110, 050034, Medellín, Colombia
| | - Juan Carlos Rincón Flórez
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Medellín, Carrera 65 N 59A-110, 050034, Medellín, Colombia
- Grupo de Investigación Biodiversidad y Genética Molecular (BIOGEM), Universidad Nacional de Colombia Sede Palmira, Carrera 32 N 12 - 00, PC 763352, Palmira, Colombia
| |
Collapse
|
5
|
Vieira LS, Zhang Y, López Quiñones AJ, Hu T, Singh DK, Stevens J, Prasad B, Park JR, Wang J. The Plasma Membrane Monoamine Transporter is Highly Expressed in Neuroblastoma and Functions as an mIBG Transporter. J Pharmacol Exp Ther 2023; 387:239-248. [PMID: 37541765 PMCID: PMC10658915 DOI: 10.1124/jpet.123.001672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/27/2023] [Accepted: 07/06/2023] [Indexed: 08/06/2023] Open
Abstract
Neuroblastoma (NB) is a pediatric cancer with low survival rates in high-risk patients. 131I-mIBG has emerged as a promising therapy for high-risk NB and kills tumor cells by radiation. Consequently, 131I-mIBG tumor uptake and retention are major determinants for its therapeutic efficacy. mIBG enters NB cells through the norepinephrine transporter (NET), and accumulates in mitochondria through unknown mechanisms. Here we evaluated the expression of monoamine and organic cation transporters in high-risk NB tumors and explored their relationship with MYCN amplification and patient survival. We found that NB mainly expresses NET, the plasma membrane monoamine transporter (PMAT), and the vesicular membrane monoamine transporter 1/2 (VMAT1/2), and that the expression of these transporters is significantly reduced in MYCN-amplified tumor samples. PMAT expression is the highest and correlates with overall survival in high-risk NB patients without MYCN amplification. Immunostaining showed that PMAT resides intracellularly in NB cells and co-localizes with mitochondria. Using cells expressing PMAT, mIBG was identified as a PMAT substrate. In mitochondria isolated from NB cell lines, mIBG uptake was reduced by ∼50% by a PMAT inhibitor. Together, our data suggest that PMAT is a previously unrecognized transporter highly expressed in NB and could impact intracellular transport and therapeutic response to 131I-mIBG. SIGNIFICANCE STATEMENT: This study identified that plasma membrane monoamine transporter (PMAT) is a novel transporter highly expressed in neuroblastoma and its expression level is associated with overall survival rate in high-risk patients without MYCN amplification. PMAT is expressed intracellularly in neuroblastoma cells, transports meta-iodobenzylguanidine (mIBG) and thus could impact tumor retention and response to 131I-mIBG therapy. These findings have important clinical implications as PMAT could represent a novel molecular marker to help inform disease prognosis and predict response to 131I-mIBG therapy.
Collapse
Affiliation(s)
- Letícia Salvador Vieira
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.S.V., Y.Z., A.J.L.Q., T.H., J.W.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., B.P.); and Seattle Children's Hospital, Seattle, Washington (J.S., J.R.P.)
| | - Yuchen Zhang
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.S.V., Y.Z., A.J.L.Q., T.H., J.W.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., B.P.); and Seattle Children's Hospital, Seattle, Washington (J.S., J.R.P.)
| | - Antonio J López Quiñones
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.S.V., Y.Z., A.J.L.Q., T.H., J.W.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., B.P.); and Seattle Children's Hospital, Seattle, Washington (J.S., J.R.P.)
| | - Tao Hu
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.S.V., Y.Z., A.J.L.Q., T.H., J.W.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., B.P.); and Seattle Children's Hospital, Seattle, Washington (J.S., J.R.P.)
| | - Dilip Kumar Singh
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.S.V., Y.Z., A.J.L.Q., T.H., J.W.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., B.P.); and Seattle Children's Hospital, Seattle, Washington (J.S., J.R.P.)
| | - Jeffrey Stevens
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.S.V., Y.Z., A.J.L.Q., T.H., J.W.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., B.P.); and Seattle Children's Hospital, Seattle, Washington (J.S., J.R.P.)
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.S.V., Y.Z., A.J.L.Q., T.H., J.W.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., B.P.); and Seattle Children's Hospital, Seattle, Washington (J.S., J.R.P.)
| | - Julie R Park
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.S.V., Y.Z., A.J.L.Q., T.H., J.W.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., B.P.); and Seattle Children's Hospital, Seattle, Washington (J.S., J.R.P.)
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.S.V., Y.Z., A.J.L.Q., T.H., J.W.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.K.S., B.P.); and Seattle Children's Hospital, Seattle, Washington (J.S., J.R.P.)
| |
Collapse
|
6
|
Sun A, Hagenbuch B, Kelly EJ, Wang J. Molecular Mechanisms of Organic Anion Transporting Polypeptide-Mediated Organic Anion Clearance at the Blood-Cerebrospinal Fluid Barrier. Mol Pharmacol 2023; 104:255-265. [PMID: 37652713 PMCID: PMC10658916 DOI: 10.1124/molpharm.123.000703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023] Open
Abstract
The blood-cerebrospinal fluid barrier (BCSFB), formed by the choroid plexus epithelial (CPE) cells, plays an active role in removing drugs and metabolic wastes from the brain. Recent functional studies in isolated mouse choroid plexus (CP) tissues suggested the presence of organic anion transporting polypeptides (OATPs, encoded by SLCOs) at the apical membrane of BCSFB, which may clear large organic anions from the cerebrospinal fluid (CSF). However, the specific OATP isoform involved is unclear. Using quantitative fluorescence imaging, we showed that the fluorescent anions sulforhodamine 101 (SR101), fluorescein methotrexate (FL-MTX), and 8-fluorescein-cAMP (fluo-cAMP) are actively transported from the CSF to the subepithelial space in CP tissues isolated from wild-type mice. In contrast, transepithelial transport of these compounds across the CPE cells was abolished in Oatp1a/1b-/- mice due to impaired apical uptake. Using transporter-expressing cell lines, SR101, FL-MTX, and fluo-cAMP were additionally shown to be transported by mouse OATP1A5 and its human counterpart OATP1A2. Kinetic analysis showed that estrone-3-sulfate and SR101 are transported by OATP1A2 and OATP1A5 with similar Michaelis-Menten constants (Km). Immunofluorescence staining further revealed the presence of OATP1A2 protein in human CP tissues. Together, our results suggest that large organic anions in the CSF are actively transported into CPE cells by apical OATP1A2 (OATP1A5 in mice), then subsequently effluxed into the blood by basolateral multidrug resistance-associated proteins (MRPs). As OATP1A2 transports a wide array of endogenous compounds and xenobiotics, the presence of this transporter at the BCSFB may imply a novel clearance route for drugs and neurohormones from the CSF. SIGNIFICANCE STATEMENT: Drug transporters at the blood-cerebrospinal fluid (CSF) barrier play an important but understudied role in brain drug disposition. This study revealed a functional contribution of rodent organic anion transporting polypeptide (OATP) 1A5 towards the CSF clearance of organic anions and suggested a similar role for OATP1A2 in humans. Delineating the molecular mechanisms governing CSF organic anion clearance may help to improve the prediction of central nervous system (CNS) pharmacokinetics and identify drug candidates with favorable CNS pharmacokinetic properties.
Collapse
Affiliation(s)
- Austin Sun
- Department of Pharmaceutics (A.S., E.J.K., J.W.) and Kidney Research Institute (E.J.K.), University of Washington, Seattle, Washington; and Department of Pharmacology, Toxicology, and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas (B.H.)
| | - Bruno Hagenbuch
- Department of Pharmaceutics (A.S., E.J.K., J.W.) and Kidney Research Institute (E.J.K.), University of Washington, Seattle, Washington; and Department of Pharmacology, Toxicology, and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas (B.H.)
| | - Edward J Kelly
- Department of Pharmaceutics (A.S., E.J.K., J.W.) and Kidney Research Institute (E.J.K.), University of Washington, Seattle, Washington; and Department of Pharmacology, Toxicology, and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas (B.H.)
| | - Joanne Wang
- Department of Pharmaceutics (A.S., E.J.K., J.W.) and Kidney Research Institute (E.J.K.), University of Washington, Seattle, Washington; and Department of Pharmacology, Toxicology, and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas (B.H.)
| |
Collapse
|
7
|
Weber BL, Nicodemus MM, Hite AK, Spalding IR, Beaver JN, Scrimshaw LR, Kassis SK, Reichert JM, Ford MT, Russell CN, Hallal EM, Gilman TL. Heterotypic Stressors Unmask Behavioral Influences of PMAT Deficiency in Mice. Int J Mol Sci 2023; 24:16494. [PMID: 38003684 PMCID: PMC10671398 DOI: 10.3390/ijms242216494] [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: 09/08/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Certain life stressors having enduring physiological and behavioral consequences, in part by eliciting dramatic signaling shifts in monoamine neurotransmitters. High monoamine levels can overwhelm selective transporters like the serotonin transporter. This is when polyspecific transporters like plasma membrane monoamine transporter (PMAT, Slc29a4) are hypothesized to contribute most to monoaminergic signaling regulation. Here, we employed two distinct counterbalanced stressors-fear conditioning and swim stress-in mice to systematically determine how reductions in PMAT function affect heterotypic stressor responsivity. We hypothesized that male heterozygotes would exhibit augmented stressor responses relative to female heterozygotes. Decreased PMAT function enhanced context fear expression, an effect unexpectedly obscured by a sham stress condition. Impaired cued fear extinction retention and enhanced context fear expression in males were conversely unmasked by a sham swim condition. Abrogated corticosterone levels in male heterozygotes that underwent swim stress after context fear conditioning did not map onto any measured behaviors. In sum, male heterozygous mouse fear behaviors proved malleable in response to preceding stressor or sham stress exposure. Combined, these data indicate that reduced male PMAT function elicits a form of stress-responsive plasticity. Future studies should assess how PMAT is differentially affected across sexes and identify downstream consequences of the stress-shifted corticosterone dynamics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - T. Lee Gilman
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH 44240, USA
| |
Collapse
|
8
|
Weber BL, Nicodemus MM, Hite AK, Spalding IR, Beaver JN, Scrimshaw LR, Kassis SK, Reichert JM, Ford MT, Russell CN, Hallal EM, Gilman TL. Heterotypic stressors unmask behavioral influences of PMAT deficiency in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555632. [PMID: 37693400 PMCID: PMC10491137 DOI: 10.1101/2023.08.30.555632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Certain life stressors having enduring physiological and behavioral consequences, in part by eliciting dramatic signaling shifts in monoamine neurotransmitters. High monoamine levels can overwhelm selective transporters like the serotonin transporter. This is when polyspecific transporters like plasma membrane monoamine transporter (PMAT, Slc29a4) are hypothesized to contribute most to monoaminergic signaling regulation. Here, we employed two distinct counterbalanced stressors - fear conditioning, and swim stress - in mice to systematically determine how reductions in PMAT function affect heterotypic stressor responsivity. We hypothesized male heterozygotes would exhibit augmented stressor responses relative to female heterozygotes. Decreased PMAT function enhanced context fear expression, an effect unexpectedly obscured by a sham stress condition. Impaired cued fear extinction retention and enhanced context fear expression in males were conversely unmasked by a sham swim condition. Abrogated corticosterone levels in male heterozygotes that underwent swim stress after context fear conditioning did not map on to any measured behaviors. In sum, male heterozygous mouse fear behaviors proved malleable in response to preceding stressor or sham stress exposure. Combined, these data indicate reduced male PMAT function elicits a form of stress-responsive plasticity. Future studies should assess how PMAT is differentially affected across sexes and identify downstream consequences of the stress-shifted corticosterone dynamics.
Collapse
Affiliation(s)
- Brady L Weber
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Marissa M Nicodemus
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Allianna K Hite
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Isabella R Spalding
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Jasmin N Beaver
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Lauren R Scrimshaw
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Sarah K Kassis
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Julie M Reichert
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Matthew T Ford
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Cameron N Russell
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - Elayna M Hallal
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| | - T Lee Gilman
- Department of Psychological Sciences, Brain Health Research Institute, Healthy Communities Research Institute, Kent State University, Kent, OH, USA
| |
Collapse
|
9
|
Weber BL, Beaver JN, Gilman TL. Summarizing studies using constitutive genetic deficiency to investigate behavioural influences of uptake 2 monoamine transporters. Basic Clin Pharmacol Toxicol 2023; 133:439-458. [PMID: 36316031 PMCID: PMC10657738 DOI: 10.1111/bcpt.13810] [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: 08/05/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 11/27/2022]
Abstract
Burgeoning literature demonstrates that monoamine transporters with high transport capacity but lower substrate affinity (i.e., uptake 2) contribute meaningfully to regulation of monoamine neurotransmitter signalling. However, studying behavioural influences of uptake 2 is hindered by an absence of selective inhibitors largely free of off-target, confounding effects. This contrasts with study of monoamine transporters with low transport capacity but high substrate affinity (i.e., uptake 1), for which there are many reasonably selective inhibitors. To circumvent this dearth of pharmacological tools for studying uptake 2, researchers have instead employed mice with constitutive genetic deficiency in three separate transporters. By studying baseline behavioural shifts, plus behavioural responses to environmental and pharmacological manipulations-the latter primarily targeting uptake 1-investigators have been creatively characterizing the behavioural, and often sex-specific, influences of uptake 2. This non-systematic mini review summarizes current uptake 2 behaviour literature, highlighting emphases on stress responsivity in organic cation transporter 2 (OCT2) work, psychostimulant responsivity in OCT3 and plasma membrane monoamine transporter (PMAT) investigations, and antidepressant responsivity in all three. Collectively, this small but growing body of work reiterates the necessity for development of selective uptake 2-inhibiting drugs, with reviewed studies suggesting that these might advance personalized treatment approaches.
Collapse
Affiliation(s)
- Brady L Weber
- Department of Psychological Sciences & Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Jasmin N Beaver
- Department of Psychological Sciences & Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - T Lee Gilman
- Department of Psychological Sciences & Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| |
Collapse
|
10
|
Sun A, Wang J. Functional Evaluation of P-gp and Bcrp at the Murine Blood-Cerebrospinal Fluid Barrier. Pharm Res 2023; 40:2667-2675. [PMID: 37704894 DOI: 10.1007/s11095-023-03598-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/28/2023] [Indexed: 09/15/2023]
Abstract
PURPOSE The brain is protected from circulating metabolites and xenobiotics by the blood-brain barrier (BBB) and the blood-cerebrospinal fluid (CSF) barrier. Previous studies report that P-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp) are expressed apically or subapically at the blood-CSF barrier (BCSFB), implying a paradoxical function to mediate blood-to-CSF transport of xenobiotics. As evidence of P-gp and Bcrp activity at the BCSFB is limited, the goal of this study is to investigate functional activity of P-gp and Bcrp at the murine BCSFB using a live tissue imaging approach. METHODS The choroid plexuses (CP) forming the BCSFB were freshly isolated from mouse brain ventricles and incubated with fluorescent probes calcein-AM and BODIPY FL-Prazosin. Using quantitative fluorescence microscopy, the functional contributions of Bcrp and P-gp were examined using inhibitors and mice with targeted deletion of the Abcb1a/b or Abcg2 gene. RESULTS Apical transport of calcein-AM in choroid plexus epithelial (CPE) cells is sensitive to inhibition by elacridar and Ko143 but is unaffected by P-gp deletion. In wild-type mice, elacridar increased CPE accumulation of BODIPY FL-Prazosin by 220% whereas deletion of Bcrp increased BODIPY FL-Prazosin accumulation by 43%. There was no change in Mdr1a/1b mRNA expression in CP tissues from the Bcrp-/- mice. CONCLUSIONS This study demonstrated functional activity of Bcrp at the BCSFB apical membrane and provided evidence supporting an additional contribution by P-gp. These findings contribute to the understanding of transport mechanisms that regulate CSF drug concentrations, which may benefit future predictions of CNS drug disposition, efficacy, and toxicity.
Collapse
Affiliation(s)
- Austin Sun
- Department of Pharmaceutics, University of Washington, H272 Health Sciences Building, Seattle, WA, 98195-7610, USA
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, H272 Health Sciences Building, Seattle, WA, 98195-7610, USA.
| |
Collapse
|
11
|
Clauss NJ, Mayer FP, Owens WA, Vitela M, Clarke KM, Bowman MA, Horton RE, Gründemann D, Schmid D, Holy M, Gould GG, Koek W, Sitte HH, Daws LC. Ethanol inhibits dopamine uptake via organic cation transporter 3: Implications for ethanol and cocaine co-abuse. Mol Psychiatry 2023; 28:2934-2945. [PMID: 37308680 PMCID: PMC10615754 DOI: 10.1038/s41380-023-02064-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/20/2022] [Accepted: 03/29/2023] [Indexed: 06/14/2023]
Abstract
Concurrent cocaine and alcohol use is among the most frequent drug combination, and among the most dangerous in terms of deleterious outcomes. Cocaine increases extracellular monoamines by blocking dopamine (DA), norepinephrine (NE) and serotonin (5-HT) transporters (DAT, NET and SERT, respectively). Likewise, ethanol also increases extracellular monoamines, however evidence suggests that ethanol does so independently of DAT, NET and SERT. Organic cation transporter 3 (OCT3) is an emergent key player in the regulation of monoamine signaling. Using a battery of in vitro, in vivo electrochemical, and behavioral approaches, as well as wild-type and constitutive OCT3 knockout mice, we show that ethanol's actions to inhibit monoamine uptake are dependent on OCT3. These findings provide a novel mechanistic basis whereby ethanol enhances the neurochemical and behavioral effects of cocaine and encourage further research into OCT3 as a target for therapeutic intervention in the treatment of ethanol and ethanol/cocaine use disorders.
Collapse
Affiliation(s)
- N J Clauss
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - F P Mayer
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - W A Owens
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - M Vitela
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - K M Clarke
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - M A Bowman
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - R E Horton
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - D Gründemann
- Department of Pharmacology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - D Schmid
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - M Holy
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - G G Gould
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - W Koek
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - H H Sitte
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
- Center for Addiction Research and Science, Medical University Vienna, Waehringerstrasse 13 A, 1090, Vienna, Austria
| | - L C Daws
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
12
|
Xie J, Han Y, Liang Y, Peng L, Wang T. Drosophila HisT is a specific histamine transporter that contributes to histamine recycling in glia. SCIENCE ADVANCES 2022; 8:eabq1780. [PMID: 36288320 PMCID: PMC9604546 DOI: 10.1126/sciadv.abq1780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/06/2022] [Indexed: 06/16/2023]
Abstract
Histamine is an important monoamine neurotransmitter that regulates multiple physiological activities in both vertebrates and invertebrates. Clearance and recycling of histamine are critical for sustaining histaminergic transmission. However, unlike other monoamine neurotransmitters, a histamine-specific transporter capable of clearing histamine from the synaptic cleft has not been identified. Here, through an in vitro histamine uptake screening, we identified an epithelial glia-expressing transporter, HisT (Histamine Transporter), that specifically transports histamine into cells. HisT misexpression in both pre- and postsynaptic neurons revealed a critical in vivo role for HisT in histamine transport and synaptic transmission. Last, we generated null hist alleles and demonstrated key physiological roles of HisT in maintaining histamine pools and sustaining visual transmission when the de novo synthesis of histamine synthesis was reduced. Our work identifies the first transporter that specifically recycles histamine and further indicates that the histamine clearance pathway may involve both the uptake-1 and uptake-2 transport systems.
Collapse
Affiliation(s)
- Jun Xie
- National Institute of Biological Sciences, Beijing 102206, China
| | - Yongchao Han
- National Institute of Biological Sciences, Beijing 102206, China
| | - Yufeng Liang
- National Institute of Biological Sciences, Beijing 102206, China
- School of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Lei Peng
- National Institute of Biological Sciences, Beijing 102206, China
- College of Biological Sciences, China Agricultural University, Beijing 100083, China
| | - Tao Wang
- National Institute of Biological Sciences, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
13
|
Contribution of ENT4 to adenosine uptake in AC16 human cardiomyocytes under simulated ischemic conditions and its potential role in cardioprotection. Mol Biol Rep 2022; 49:11201-11208. [DOI: 10.1007/s11033-022-07902-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 10/14/2022]
|
14
|
Targeting choroid plexus epithelium as a novel therapeutic strategy for hydrocephalus. J Neuroinflammation 2022; 19:156. [PMID: 35715859 PMCID: PMC9205094 DOI: 10.1186/s12974-022-02500-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 06/01/2022] [Indexed: 11/25/2022] Open
Abstract
The choroid plexus is a tissue located in the lateral ventricles of the brain and is composed mainly of choroid plexus epithelium cells. The main function is currently thought to be the secretion of cerebrospinal fluid and the regulation of its pH, and more functions are gradually being demonstrated. Assistance in the removal of metabolic waste and participation in the apoptotic pathway are also the functions of choroid plexus. Besides, it helps to repair the brain by regulating the secretion of neuropeptides and the delivery of drugs. It is involved in the immune response to assist in the clearance of infections in the central nervous system. It is now believed that the choroid plexus is in an inflammatory state after damage to the brain. This state, along with changes in the cilia, is thought to be an abnormal physiological state of the choroid plexus, which in turn leads to abnormal conditions in cerebrospinal fluid and triggers hydrocephalus. This review describes the pathophysiological mechanism of hydrocephalus following choroid plexus epithelium cell abnormalities based on the normal physiological functions of choroid plexus epithelium cells, and analyzes the attempts and future developments of using choroid plexus epithelium cells as a therapeutic target for hydrocephalus.
Collapse
|
15
|
Beaver JN, Weber BL, Ford MT, Anello AE, Kassis SK, Gilman TL. Uncovering Functional Contributions of PMAT ( Slc29a4) to Monoamine Clearance Using Pharmacobehavioral Tools. Cells 2022; 11:cells11121874. [PMID: 35741002 PMCID: PMC9220966 DOI: 10.3390/cells11121874] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Plasma membrane monoamine transporter (PMAT, Slc29a4) transports monoamine neurotransmitters, including dopamine and serotonin, faster than more studied monoamine transporters, e.g., dopamine transporter (DAT), or serotonin transporter (SERT), but with ~400–600-fold less affinity. A considerable challenge in understanding PMAT’s monoamine clearance contributions is that no current drugs selectively inhibit PMAT. To advance knowledge about PMAT’s monoamine uptake role, and to circumvent this present challenge, we investigated how drugs that selectively block DAT/SERT influence behavioral readouts in PMAT wildtype, heterozygote, and knockout mice of both sexes. Drugs typically used as antidepressants (escitalopram, bupropion) were administered acutely for readouts in tail suspension and locomotor tests. Drugs with psychostimulant properties (cocaine, D-amphetamine) were administered repeatedly to assess initial locomotor responses plus psychostimulant-induced locomotor sensitization. Though we hypothesized that PMAT-deficient mice would exhibit augmented responses to antidepressant and psychostimulant drugs due to constitutively attenuated monoamine uptake, we instead observed sex-selective responses to antidepressant drugs in opposing directions, and subtle sex-specific reductions in psychostimulant-induced locomotor sensitization. These results suggest that PMAT functions differently across sexes, and support hypotheses that PMAT’s monoamine clearance contribution emerges when frontline transporters (e.g., DAT, SERT) are absent, saturated, and/or blocked. Thus, known human polymorphisms that reduce PMAT function could be worth investigating as contributors to varied antidepressant and psychostimulant responses.
Collapse
|
16
|
Hu T, Zha W, Sun A, Wang J. Live Tissue Imaging Reveals Distinct Transcellular Pathways for Organic Cations and Anions at the Blood-Cerebrospinal Fluid Barrier. Mol Pharmacol 2022; 101:334-342. [PMID: 35193935 PMCID: PMC9092482 DOI: 10.1124/molpharm.121.000439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/31/2022] [Indexed: 11/22/2022] Open
Abstract
Formed by the choroid plexus epithelial (CPE) cells, the blood-cerebrospinal fluid barrier (BCSFB) plays an active role in removing drugs, toxins, and metabolic wastes from the brain. Several organic cation and anion transporters are expressed in the CPE cells, but how they functionally mediate transepithelial transport of organic cations and anions remain unclear. In this study, we visualized the transcellular transport of fluorescent organic cation and organic anion probes using live tissue imaging in freshly isolated mouse choroid plexuses (CPs). The cationic probe, 4-[4-(dimethylamino)phenyl]-1-methylpyridinium iodide (IDT307) was transported into CPE cells at the apical membrane and highly accumulated in mitochondria. Consistent with the lack of expression of organic cation efflux transporters, there was little efflux of IDT307 into the blood capillary space. Furthermore, IDT307 uptake and intracellular accumulation was attenuated by approximately 70% in CP tissues from mice with targeted deletion of the plasma membrane monoamine transporter (Pmat). In contrast, the anionic probe fluorescein-methotrexate (FL-MTX) was rapidly transported across the CPE cells into the capillary space with little intracellular accumulation. Rifampicin, an inhibitor of organic anion transporting polypeptides (OATPs), completely blocked FL-MTX uptake into the CPE cells whereas MK-571, a pan-inhibitor of multidrug resistance associated proteins (MRPs), abolished basolateral efflux of FL-MTX. In summary, our results suggest distinct transcellular transport pathways for organic cations and anions at the BCSFB and reveal a pivotal role of PMAT, OATP and MRP transporters in organic cation and anion transport at the blood-cerebrospinal fluid interface. SIGNIFICANCE STATEMENT: Live tissue imaging revealed that while organic cations are transported from the cerebrospinal fluid (CSF) into the choroid plexus epithelial cells by plasma membrane monoamine transporter without efflux into the blood, amphipathic anions in the CSF are efficiently transported across the BCSFB through the collaborated function of apical organic anion transporting polypeptides and basolateral multidrug resistance associated proteins. These findings contribute to a mechanistic understanding of the molecular and cellular pathways for choroid plexus clearance of solutes from the brain.
Collapse
Affiliation(s)
- Tao Hu
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Weibin Zha
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Austin Sun
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| |
Collapse
|
17
|
Evaluation of Blood-CSF Barrier Transport by Quantitative Real Time Fluorescence Microscopy. Pharm Res 2022; 39:1469-1480. [PMID: 35411508 DOI: 10.1007/s11095-022-03251-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 04/01/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Transporters at the blood-cerebrospinal fluid (CSF) barrier (BCSFB) play active roles in removing drugs and toxins from the CSF. The goal of this study is to develop a fluorescence microscopy approach to quantitatively study the transepithelial transport processes at the murine BCSFB in real time. METHODS Choroid plexus (CP) tissues were isolated from mouse lateral ventricles and incubated with anionic (fluorescein-methotrexate, 8-fluorescein-cAMP) or cationic (IDT307) fluorescent probes. The CSF-to-blood transport was imaged and quantified using compartmental segmentation and digital image analysis. Real time images were captured and analyzed to obtain kinetic information and identify the rate-limiting step. The effect of transporter inhibitors was also evaluated. RESULTS The transport processes of fluorescent probes can be captured and analyzed digitally. The intra- and inter- animal variability were 20.4% and 25.7%, respectively. Real time analysis showed distinct transport kinetics and rate-limiting step for anionic and cationic probes. A CP efflux index was proposed to distinguish between transepithelial flux and intracellular accumulation. Rifampin and MK571 decreased the overall transepithelial transport of anionic probes by more than 90%, indicating a possible involvement of organic anion transporting polypeptides (Oatps) and multidrug resistance-associated proteins (Mrps). CONCLUSIONS A CP isolation method was described, and a quantitative fluorescence imaging approach was developed to evaluate CSF-to-blood transport in mouse CP. The method is consistent, reproducible, and capable of tracking real time transepithelial transport with temporal and spatial resolution. The approach can be used to evaluate transport mechanisms, assess tissue drug accumulation, and assay potential drug-drug interactions at the BCSFB.
Collapse
|
18
|
Lopez Quiñones AJ, Vieira LS, Wang J. Clinical Applications and the Roles of Transporters in Disposition, Tumor Targeting, and Tissue Toxicity of meta-Iodobenzylguanidine (mIBG). Drug Metab Dispos 2022; 50:DMD-MR-2021-000707. [PMID: 35197314 PMCID: PMC9488973 DOI: 10.1124/dmd.121.000707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/01/2022] [Accepted: 02/17/2022] [Indexed: 11/22/2022] Open
Abstract
Transporters on the plasma membrane of tumor cells are promising molecular "Trojan horses" to deliver drugs and imaging agents into cancer cells. Radioiodine-labeled meta-iodobenzylguanidine (mIBG) is used as a diagnostic agent (123I-mIBG) and a targeted radiotherapy (131I-mIBG) for neuroendocrine cancers. mIBG enters cancer cells through the norepinephrine transporter (NET) where the radioactive decay of 131I causes DNA damage, cell death, and tumor necrosis. mIBG is predominantly eliminated unchanged by the kidney. Despite its selective uptake by neuroendocrine tumors, mIBG accumulates in several normal tissues and leads to tissue-specific radiation toxicities. Emerging evidences suggest that the polyspecific organic cation transporters play important roles in systemic disposition and tissue-specific uptake of mIBG. In particular, human organic cation transporter 2 (hOCT2) and toxin extrusion proteins 1 and 2-K (hMATE1/2-K) likely mediate renal secretion of mIBG whereas hOCT1 and hOCT3 may contribute to mIBG uptake into normal tissues such as the liver, salivary glands, and heart. This mini-review focuses on the clinical applications of mIBG in neuroendocrine cancers and the differential roles of NET, OCT and MATE transporters in mIBG disposition, response and toxicity. Understanding the molecular mechanisms governing mIBG transport in cancer and normal cells is a critical step for developing strategies to optimize the efficacy of 131I-mIBG while minimizing toxicity in normal tissues. Significance Statement Radiolabeled mIBG has been used as a diagnostic tool and as radiotherapy for neuroendocrine cancers and other diseases. NET, OCT and MATE transporters play differential roles in mIBG tumor targeting, systemic elimination, and accumulation in normal tissues. The clinical use of mIBG as a radiopharmaceutical in cancer diagnosis and treatment can be further improved by taking a holistic approach considering mIBG transporters in both cancer and normal tissues.
Collapse
Affiliation(s)
| | | | - Joanne Wang
- Dept. of Pharmaceutics, University of Washington, United States
| |
Collapse
|
19
|
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the most commonly prescribed medications for psychiatric disorders, yet they leave the majority of patients without full symptom relief. Therefore, a major research challenge is to identify novel targets for the improved treatment of these disorders. SSRIs act by blocking the serotonin transporter (SERT), the high-affinity, low-capacity, uptake-1 transporter for serotonin. Other classes of antidepressant work by blocking the norepinephrine or dopamine transporters (NET and DAT), the high-affinity, low-capacity uptake-1 transporters for norepinephrine and dopamine, or by blocking combinations of SERT, NET, and DAT. It has been proposed that uptake-2 transporters, which include organic cation transporters (OCTs) and the plasma membrane monoamine transporter (PMAT), undermine the therapeutic utility of uptake-1 acting antidepressants. Uptake-2 transporters for monoamines have low affinity for these neurotransmitters, but a high capacity to transport them. Thus, activity of these transporters may limit the increase of extracellular monoamines thought to be essential for ultimate therapeutic benefit. Here preclinical evidence supporting a role for OCT2, OCT3, and PMAT in behaviors relevant to psychiatric disorders is presented. Importantly, preclinical evidence revealing these transporters as targets for the development of novel therapeutics for psychiatric disorders is discussed.
Collapse
|
20
|
Abstract
Catecholamines, including dopamine, norepinephrine, and epinephrine, are modulatory transmitters released from specialized neurons throughout the brain. Collectively, catecholamines exert powerful regulation of mood, motivation, arousal, and plasticity. Transporter-mediated uptake determines the peak concentration, duration, and physical spread of released catecholamines, thus playing key roles in determining the magnitude and duration of their modulatory effects. Most studies of catecholamine clearance have focused on the presynaptic high-affinity, low-capacity dopamine (DAT), and norepinephrine (NET) transporters, which are members of the uptake1 family of monoamine transporters. However, recent studies have demonstrated that members of the uptake2 family of monoamine transporters, including organic cation transporter 2 (OCT2), OCT3, and the plasma membrane monoamine transporter (PMAT) are expressed widely throughout the brain. In contrast to DAT and NET, these transporters have higher capacity and lower affinity for catecholamines and are multi-specific, each with the capacity to transport all catecholamines. The expression of these transporters in the brain suggests that they play significant roles in regulating catecholamine homeostasis. This review summarizes studies describing the anatomical distribution of OCT2, OCT3, and PMAT, their cellular and subcellular localization, and their contribution to the regulation of the clearance of catecholamines in the brain.
Collapse
|
21
|
Sweet DH. Organic Cation Transporter Expression and Function in the CNS. Handb Exp Pharmacol 2021; 266:41-80. [PMID: 33963461 DOI: 10.1007/164_2021_463] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB) represent major control checkpoints protecting the CNS, by exerting selective control over the movement of organic cations and anions into and out of the CNS compartment. In addition, multiple CNS cell types, e.g., astrocytes, ependymal cells, microglia, contribute to processes that maintain the status quo of the CNS milieu. To fulfill their roles, these barriers and cell types express a multitude of transporter proteins from dozens of different transporter families. Fundamental advances over the past few decades in our knowledge of transporter substrates, expression profiles, and consequences of loss of function are beginning to change basic theories regarding the contribution of various cell types and clearance networks to coordinated neuronal signaling, complex organismal behaviors, and overall CNS homeostasis. In particular, transporters belonging to the Solute Carrier (SLC) superfamily are emerging as major contributors, including the SLC22 organic cation/anion/zwitterion family of transporters (includes OCT1-3 and OCTN1-3), the SLC29 facilitative nucleoside family of transporters (includes PMAT), and the SLC47 multidrug and toxin extrusion family of transporters (includes MATE1-2). These transporters are known to interact with neurotransmitters, antidepressant and anxiolytic agents, and drugs of abuse. Clarifying their contributions to the underlying mechanisms regulating CNS permeation and clearance, as well as the health status of astrocyte, microglial and neuronal cell populations, will drive new levels of understanding as to maintenance of the CNS milieu and approaches to new therapeutics and therapeutic strategies in the treatment of CNS disorders. This chapter highlights organic cation transporters belonging to the SLC superfamily known to be expressed in the CNS, providing an overview of their identification, mechanism of action, CNS expression profile, interaction with neurotransmitters and antidepressant/antipsychotic drugs, and results from behavioral studies conducted in loss of function models (knockout/knockdown).
Collapse
Affiliation(s)
- Douglas H Sweet
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
22
|
Sun A, Wang J. Choroid Plexus and Drug Removal Mechanisms. AAPS JOURNAL 2021; 23:61. [PMID: 33942198 DOI: 10.1208/s12248-021-00587-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/24/2021] [Indexed: 01/08/2023]
Abstract
Timely and efficient removal of xenobiotics and metabolites from the brain is crucial in maintaining the homeostasis and normal function of the brain. The choroid plexus (CP) forms the blood-cerebrospinal fluid barrier and vitally removes drugs and wastes from the brain through several co-existing clearance mechanisms. The CP epithelial (CPE) cells synthesize and secrete the cerebrospinal fluid (CSF). As the CSF passes through the ventricular and subarachnoid spaces and eventually drains into the general circulation, it collects and removes drugs, toxins, and metabolic wastes from the brain. This bulk flow of the CSF serves as a default and non-selective pathway for the removal of solutes and macromolecules from the brain interstitium. Besides clearance by CSF bulk flow, the CPE cells express several multispecific membrane transporters to actively transport substrates from the CSF side into the blood side. In addition, several phase I and II drug-metabolizing enzymes are expressed in the CPE cells, which enzymatically inactivate a broad spectrum of reactive or toxic substances. This review summarizes our current knowledge of the functional characteristics and key contributors to the various clearance pathways in the CP-CSF system, overviewing recent developments in our understanding of CSF flow dynamics and the functional roles of CP uptake and efflux transporters in influencing CSF drug concentrations.
Collapse
Affiliation(s)
- Austin Sun
- Department of Pharmaceutics, University of Washington, Health Science Building Room H-272J, Box 357610, Seattle, Washington, 98195-7610, USA
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Health Science Building Room H-272J, Box 357610, Seattle, Washington, 98195-7610, USA.
| |
Collapse
|
23
|
Wei R, Gust SL, Tandio D, Maheux A, Nguyen KH, Wang J, Bourque S, Plane F, Hammond JR. Deletion of murine slc29a4 modifies vascular responses to adenosine and 5-hydroxytryptamine in a sexually dimorphic manner. Physiol Rep 2021; 8:e14395. [PMID: 32170814 PMCID: PMC7070170 DOI: 10.14814/phy2.14395] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/15/2022] Open
Abstract
Equilibrative nucleoside transporter 4 (ENT4), encoded by SLC29A4, mediates the flux of both 5‐hydroxytryptamine (5‐HT) and adenosine across cell membranes. We hypothesized that loss of ENT4 function in mice would modify the effects of these established regulators of vascular function. Male and female wild‐type (WT) and slc29a4‐null (ENT4‐KO) mice were compared with respect to their hemodynamics and mesenteric vascular function. Male ENT4‐KO mice had a complete loss of myogenic tone in their mesenteric resistance arteries. This was accompanied by a decrease in blood flow in the superior mesenteric artery in the male ENT4‐KO mice, and a reduced responsiveness to 5‐HT. In contrast, endothelium‐dependent relaxations of mesenteric arteries from female ENT4‐KO mice were more sensitive to Ca2+‐activated K+ (KCa) channel blockade than WT mice. Female ENT4‐KO mice also demonstrated an enhanced vasodilatory response to adenosine in vivo that was not seen in males. Ketanserin (5‐HT2A inhibitor) and GR55562 (5‐HT1B/1D inhibitor) decreased 5‐HT‐induced tone, but only ketanserin inhibited the relaxant effect of 5‐HT in mesenteric arteries. 5‐HT‐evoked increases in tone were elevated in arteries from ENT4‐KO mice upon block of endothelial relaxant pathways, with arteries from female ENT4‐KO mice showing the greatest increase. Adenosine A2b receptor expression was decreased, while other adenosine transporter subtypes, as well as adenosine deaminase and adenosine kinase were increased in mesenteric arteries from male, but not female, ENT4‐KO mice. These findings indicate that deletion of slc29a4 leads to sex‐specific changes in vascular function with significant consequences for regulation of blood flow and pressure by adenosine and 5‐HT.
Collapse
Affiliation(s)
- Ran Wei
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Stephen L Gust
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - David Tandio
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Alexia Maheux
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Khanh H Nguyen
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Stephane Bourque
- Department of Anaesthesia and Pain Medicine, University of Alberta, Edmonton, AB, Canada
| | - Frances Plane
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - James R Hammond
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
24
|
Betterton RD, Davis TP, Ronaldson PT. Organic Cation Transporter (OCT/OCTN) Expression at Brain Barrier Sites: Focus on CNS Drug Delivery. Handb Exp Pharmacol 2021; 266:301-328. [PMID: 33674914 PMCID: PMC8603467 DOI: 10.1007/164_2021_448] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Therapeutic delivery to the central nervous system (CNS) continues to be a considerable challenge in the pharmacological treatment and management of neurological disorders. This is primarily due to the physiological and biochemical characteristics of brain barrier sites (i.e., blood-brain barrier (BBB), blood-cerebrospinal fluid barrier (BCSFB)). Drug uptake into brain tissue is highly restricted by expression of tight junction protein complexes and adherens junctions between brain microvascular endothelial cells and choroid plexus epithelial cells. Additionally, efflux transport proteins expressed at the plasma membrane of these same endothelial and epithelial cells act to limit CNS concentrations of centrally acting drugs. In contrast, facilitated diffusion via transporter proteins allows for substrate-specific flux of molecules across the plasma membrane, directing drug uptake into the CNS. Organic Cation Transporters (OCTs) and Novel Organic Cation Transporters (OCTNs) are two subfamilies of the solute carrier 22 (SLC22) family of proteins that have significant potential to mediate delivery of positively charged, zwitterionic, and uncharged therapeutics. While expression of these transporters has been well characterized in peripheral tissues, the functional expression of OCT and OCTN transporters at CNS barrier sites and their role in delivery of therapeutic drugs to molecular targets in the brain require more detailed analysis. In this chapter, we will review current knowledge on localization, function, and regulation of OCT and OCTN isoforms at the BBB and BCSFB with a particular emphasis on how these transporters can be utilized for CNS delivery of therapeutic agents.
Collapse
Affiliation(s)
- Robert D Betterton
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Thomas P Davis
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
25
|
Abstract
Inhibitors of Na+/Cl- dependent high affinity transporters for norepinephrine (NE), serotonin (5-HT), and/or dopamine (DA) represent frequently used drugs for treatment of psychological disorders such as depression, anxiety, obsessive-compulsive disorder, attention deficit hyperactivity disorder, and addiction. These transporters remove NE, 5-HT, and/or DA after neuronal excitation from the interstitial space close to the synapses. Thereby they terminate transmission and modulate neuronal behavioral circuits. Therapeutic failure and undesired central nervous system side effects of these drugs have been partially assigned to neurotransmitter removal by low affinity transport. Cloning and functional characterization of the polyspecific organic cation transporters OCT1 (SLC22A1), OCT2 (SLC22A2), OCT3 (SLC22A3) and the plasma membrane monoamine transporter PMAT (SLC29A4) revealed that every single transporter mediates low affinity uptake of NE, 5-HT, and DA. Whereas the organic transporters are all located in the blood brain barrier, OCT2, OCT3, and PMAT are expressed in neurons or in neurons and astrocytes within brain areas that are involved in behavioral regulation. Areas of expression include the dorsal raphe, medullary motoric nuclei, hypothalamic nuclei, and/or the nucleus accumbens. Current knowledge of the transport of monoamine neurotransmitters by the organic cation transporters, their interactions with psychotropic drugs, and their locations in the brain is reported in detail. In addition, animal experiments including behavior tests in wildtype and knockout animals are reported in which the impact of OCT2, OCT3, and/or PMAT on regulation of salt intake, depression, mood control, locomotion, and/or stress effect on addiction is suggested.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute of Anatomy and Cell Biology, University Würzburg, Würzburg, Germany.
| |
Collapse
|
26
|
Abstract
Precise control of monoamine neurotransmitter levels in the central nervous system (CNS) is crucial for proper brain function. Dysfunctional monoamine signaling is associated with several neuropsychiatric and neurodegenerative disorders. The plasma membrane monoamine transporter (PMAT) is a new polyspecific organic cation transporter encoded by the SLC29A4 gene. Capable of transporting monoamine neurotransmitters with low affinity and high capacity, PMAT represents a major uptake2 transporter in the brain. Broadly expressed in multiple brain regions, PMAT can complement the high-affinity, low-capacity monoamine uptake mediated by uptake1 transporters, the serotonin, dopamine, and norepinephrine transporters (SERT, DAT, and NET, respectively). This chapter provides an overview of the molecular and functional characteristics of PMAT together with its regional and cell-type specific expression in the mammalian brain. The physiological functions of PMAT in brain monoamine homeostasis are evaluated in light of its unique transport kinetics and brain location, and in comparison with uptake1 and other uptake2 transporters (e.g., OCT3) along with corroborating experimental evidences. Lastly, the possibility of PMAT's involvement in brain pathophysiological processes, such as autism, depression, and Parkinson's disease, is discussed in the context of disease pathology and potential link to aberrant monoamine pathways.
Collapse
|
27
|
Naganuma F, Yoshikawa T. Organic Cation Transporters in Brain Histamine Clearance: Physiological and Psychiatric Implications. Handb Exp Pharmacol 2021; 266:169-185. [PMID: 33641029 DOI: 10.1007/164_2021_447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Histamine acts as a neurotransmitter in the central nervous system and is involved in numerous physiological functions. Recent studies have identified the causative role of decreased histaminergic systems in various neurological disorders. Thus, the brain histamine system has attracted attention as a therapeutic target to improve brain function. Neurotransmitter clearance is one of the most important processes for the regulation of neuronal activity and is an essential target for diverse drugs. Our previous study has shown the importance of histamine N-methyltransferase for the inactivation of brain histamine and the intracellular localization of this enzyme; the study indicated that the transport system for the movement of positively charged histamine from the extracellular to intracellular space is a prerequisite for histamine inactivation. Several studies on in vitro astrocytic histamine transport have indicated the contribution of organic cation transporter 3 (OCT3) and plasma membrane monoamine transporter (PMAT) in histamine uptake, although the importance of these transporters in in vivo histamine clearance remains unknown. Immunohistochemical analyses have revealed the expression of OCT3 and PMAT on neurons, emphasizing the importance of investigating neuronal histamine uptake. Further studies using knockout mice or fast-scan cyclic voltammetry will accelerate the research on histamine transporters. In this review article, we summarize histamine transport assays and describe the candidate transporters responsible for histamine transport in the brain.
Collapse
Affiliation(s)
- Fumito Naganuma
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Takeo Yoshikawa
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
28
|
Bhat S, El-Kasaby A, Freissmuth M, Sucic S. Functional and Biochemical Consequences of Disease Variants in Neurotransmitter Transporters: A Special Emphasis on Folding and Trafficking Deficits. Pharmacol Ther 2020; 222:107785. [PMID: 33310157 PMCID: PMC7612411 DOI: 10.1016/j.pharmthera.2020.107785] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/02/2020] [Indexed: 01/30/2023]
Abstract
Neurotransmitters, such as γ-aminobutyric acid, glutamate, acetyl choline, glycine and the monoamines, facilitate the crosstalk within the central nervous system. The designated neurotransmitter transporters (NTTs) both release and take up neurotransmitters to and from the synaptic cleft. NTT dysfunction can lead to severe pathophysiological consequences, e.g. epilepsy, intellectual disability, or Parkinson’s disease. Genetic point mutations in NTTs have recently been associated with the onset of various neurological disorders. Some of these mutations trigger folding defects in the NTT proteins. Correct folding is a prerequisite for the export of NTTs from the endoplasmic reticulum (ER) and the subsequent trafficking to their pertinent site of action, typically at the plasma membrane. Recent studies have uncovered some of the key features in the molecular machinery responsible for transporter protein folding, e.g., the role of heat shock proteins in fine-tuning the ER quality control mechanisms in cells. The therapeutic significance of understanding these events is apparent from the rising number of reports, which directly link different pathological conditions to NTT misfolding. For instance, folding-deficient variants of the human transporters for dopamine or GABA lead to infantile parkinsonism/dystonia and epilepsy, respectively. From a therapeutic point of view, some folding-deficient NTTs are amenable to functional rescue by small molecules, known as chemical and pharmacological chaperones.
Collapse
Affiliation(s)
- Shreyas Bhat
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Ali El-Kasaby
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sonja Sucic
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
29
|
Bowman MA, Vitela M, Clarke KM, Koek W, Daws LC. Serotonin Transporter and Plasma Membrane Monoamine Transporter Are Necessary for the Antidepressant-Like Effects of Ketamine in Mice. Int J Mol Sci 2020; 21:ijms21207581. [PMID: 33066466 PMCID: PMC7589995 DOI: 10.3390/ijms21207581] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 01/17/2023] Open
Abstract
Major depressive disorder is typically treated with selective serotonin reuptake inhibitors (SSRIs), however, SSRIs take approximately six weeks to produce therapeutic effects, if any. Not surprisingly, there has been great interest in findings that low doses of ketamine, a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist, produce rapid and long-lasting antidepressant effects. Preclinical studies show that the antidepressant-like effects of ketamine are dependent upon availability of serotonin, and that ketamine increases extracellular serotonin, yet the mechanism by which this occurs is unknown. Here we examined the role of the high-affinity, low-capacity serotonin transporter (SERT), and the plasma membrane monoamine transporter (PMAT), a low-affinity, high-capacity transporter for serotonin, as mechanisms contributing to ketamine’s ability to increase extracellular serotonin and produce antidepressant-like effects. Using high-speed chronoamperometry to measure real-time clearance of serotonin from CA3 region of hippocampus in vivo, we found ketamine robustly inhibited serotonin clearance in wild-type mice, an effect that was lost in mice constitutively lacking SERT or PMAT. As expected, in wild-type mice, ketamine produced antidepressant-like effects in the forced swim test. Mapping onto our neurochemical findings, the antidepressant-like effects of ketamine were lost in mice lacking SERT or PMAT. Future research is needed to understand how constitutive loss of either SERT or PMAT, and compensation that occurs in other systems, is sufficient to void ketamine of its ability to inhibit serotonin clearance and produce antidepressant-like effects. Taken together with existing literature, a critical role for serotonin, and its inhibition of uptake via SERT and PMAT, cannot be ruled out as important contributing factors to ketamine’s antidepressant mechanism of action. Combined with what is already known about ketamine’s action at NMDA receptors, these studies help lead the way to the development of drugs that lack ketamine’s abuse potential but have superior efficacy in treating depression.
Collapse
Affiliation(s)
- Melodi A. Bowman
- Department of Cellular and Integrative Physiology at University of Texas Health, San Antonio, TX 78229, USA; (M.A.B.); (M.V.); (K.M.C.)
| | - Melissa Vitela
- Department of Cellular and Integrative Physiology at University of Texas Health, San Antonio, TX 78229, USA; (M.A.B.); (M.V.); (K.M.C.)
| | - Kyra M. Clarke
- Department of Cellular and Integrative Physiology at University of Texas Health, San Antonio, TX 78229, USA; (M.A.B.); (M.V.); (K.M.C.)
- Department of Pharmacology at University of Texas Health, San Antonio, TX 78229, USA;
| | - Wouter Koek
- Department of Pharmacology at University of Texas Health, San Antonio, TX 78229, USA;
- Department of Psychiatry at University of Texas Health, San Antonio, TX 78229, USA
| | - Lynette C. Daws
- Department of Cellular and Integrative Physiology at University of Texas Health, San Antonio, TX 78229, USA; (M.A.B.); (M.V.); (K.M.C.)
- Department of Pharmacology at University of Texas Health, San Antonio, TX 78229, USA;
- Correspondence:
| |
Collapse
|
30
|
Hirayama M, Hoshino Y, Yoshii K, Toda R, Kawabata Y, Nakanishi T, Tamai I. Identification of the Uptake Transporter Responsible for Distribution of Acotiamide into Stomach Tissue. Mol Pharm 2020; 17:1071-1078. [PMID: 32105080 DOI: 10.1021/acs.molpharmaceut.9b00894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The acetylcholinesterase inhibitor, acotiamide, improves gastric motility and is clinically used to treat functional dyspepsia. The present study aimed to identify the transporters involved in the distribution of acotiamide in stomach tissue. Acotiamide uptake by the gastric cancer-derived model cell line, Hs746 T, was Na+- and pH-independent. The initial uptake velocity of acotiamide was saturable with increasing concentrations of acotiamide and was inhibited by selective serotonin reuptake inhibitors, which are potent inhibitors of the plasma membrane monoamine transporter (PMAT). The uptake of acotiamide by PMAT gene-transfected HEK293 cells was saturable, with similar Km (197.9 μM) values to those of uptake by Hs 746T cells (106 μM). Moreover, immunoreactivity of PMAT was found in the gastric smooth muscle and vascular endothelial cells. These results suggest that PMAT contributes to the distribution of acotiamide in the stomach, where it exerts its pharmacological effects.
Collapse
Affiliation(s)
- Masamichi Hirayama
- Toxicology & Pharmacokinetics Research, Central Research Laboratories, Zeria Pharmaceutical Corporation Ltd., Saitama 360-0111, Japan.,Institute of Medical Pharmaceutical and Health Sciences, Faculty of Pharmacy, Kanazawa University, Kanazawa 920-1192, Japan
| | - Yusuke Hoshino
- Toxicology & Pharmacokinetics Research, Central Research Laboratories, Zeria Pharmaceutical Corporation Ltd., Saitama 360-0111, Japan
| | - Kazuyoshi Yoshii
- Toxicology & Pharmacokinetics Research, Central Research Laboratories, Zeria Pharmaceutical Corporation Ltd., Saitama 360-0111, Japan
| | - Ryoko Toda
- Toxicology & Pharmacokinetics Research, Central Research Laboratories, Zeria Pharmaceutical Corporation Ltd., Saitama 360-0111, Japan
| | - Yoshihiro Kawabata
- Toxicology & Pharmacokinetics Research, Central Research Laboratories, Zeria Pharmaceutical Corporation Ltd., Saitama 360-0111, Japan
| | - Takeo Nakanishi
- Institute of Medical Pharmaceutical and Health Sciences, Faculty of Pharmacy, Kanazawa University, Kanazawa 920-1192, Japan
| | - Ikumi Tamai
- Institute of Medical Pharmaceutical and Health Sciences, Faculty of Pharmacy, Kanazawa University, Kanazawa 920-1192, Japan
| |
Collapse
|
31
|
Ramachandran CD, Gholami K, Lam SK, Hoe SZ. A preliminary study of the effect of a high-salt diet on transcriptome dynamics in rat hypothalamic forebrain and brainstem cardiovascular control centers. PeerJ 2020; 8:e8528. [PMID: 32175184 PMCID: PMC7059759 DOI: 10.7717/peerj.8528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 01/07/2020] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND High dietary salt intake is strongly correlated with cardiovascular (CV) diseases and it is regarded as a major risk factor associated with the pathogenesis of hypertension. The CV control centres in the brainstem (the nucleus tractus solitarii (NTS) and the rostral ventrolateral medulla (RVLM)) and hypothalamic forebrain (the subfornical organ, SFO; the supraoptic nucleus, SON and the paraventricular nucleus, PVN) have critical roles in regulating CV autonomic motor outflows, and thus maintaining blood pressure (BP). Growing evidence has implicated autonomic regulatory networks in salt-sensitive HPN (SSH), but the genetic basis remains to be delineated. We hypothesized that the development and/ or maintenance of SSH is reliant on the change in the expression of genes in brain regions controlling the CV system. METHODOLOGY We used RNA-Sequencing (RNA-Seq) to describe the differential expression of genes in SFO, SON, PVN, NTS and RVLM of rats being chronically fed with high-salt (HS) diet. Subsequently, a selection of putatively regulated genes was validated with quantitative reverse transcription polymerase chain reaction (qRT-PCR) in both Spontaneously Hypertensive rats (SHRs) and Wistar Kyoto (WKY) rats. RESULTS The findings enabled us to identify number of differentially expressed genes in SFO, SON, PVN, NTS and RVLM; that are either up-regulated in both strains of rats (SON- Caprin2, Sctr), down-regulated in both strains of rats (PVN- Orc, Gkap1), up-regulated only in SHRs (SFO- Apopt1, Lin52, AVP, OXT; SON- AVP, OXT; PVN- Caprin2, Sclt; RVLM- A4galt, Slc29a4, Cmc1) or down-regulated only in SHRs (SON- Ndufaf2, Kcnv1; PVN- Pi4k2a; NTS- Snrpd2l, Ankrd29, St6galnac6, Rnf157, Iglon5, Csrnp3, Rprd1a; RVLM- Ttr, Faim). CONCLUSIONS These findings demonstrated the adverse effects of HS diet on BP, which may be mediated via modulating the signaling systems in CV centers in the hypothalamic forebrain and brainstem.
Collapse
Affiliation(s)
- Chitra Devi Ramachandran
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Perseketuan, Malaysia
| | - Khadijeh Gholami
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Perseketuan, Malaysia
- Human Biology Division, School of Medicine, International Medical University, Kuala Lumpur, Wilayah Perseketuan, Malaysia
| | - Sau Kuen Lam
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Perseketuan, Malaysia
- Department of Pre-Clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long, Selangor, Malaysia
| | - See Ziau Hoe
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Perseketuan, Malaysia
| |
Collapse
|
32
|
Histamine N-Methyltransferase in the Brain. Int J Mol Sci 2019; 20:ijms20030737. [PMID: 30744146 PMCID: PMC6386932 DOI: 10.3390/ijms20030737] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/08/2019] [Accepted: 02/08/2019] [Indexed: 12/13/2022] Open
Abstract
Brain histamine is a neurotransmitter and regulates diverse physiological functions. Previous studies have shown the involvement of histamine depletion in several neurological disorders, indicating the importance of drug development targeting the brain histamine system. Histamine N-methyltransferase (HNMT) is a histamine-metabolising enzyme expressed in the brain. Although pharmacological studies using HNMT inhibitors have been conducted to reveal the direct involvement of HNMT in brain functions, HNMT inhibitors with high specificity and sufficient blood–brain barrier permeability have not been available until now. Recently, we have phenotyped Hnmt-deficient mice to elucidate the importance of HNMT in the central nervous system. Hnmt disruption resulted in a robust increase in brain histamine concentration, demonstrating the essential role of HNMT in the brain histamine system. Clinical studies have suggested that single nucleotide polymorphisms of the human HNMT gene are associated with several brain disorders such as Parkinson’s disease and attention deficit hyperactivity disorder. Postmortem studies also have indicated that HNMT expression is altered in human brain diseases. These findings emphasise that an increase in brain histamine levels by novel HNMT inhibitors could contribute to the improvement of brain disorders.
Collapse
|
33
|
Xue Y, Ma C, Hanna I, Pan G. Intestinal Transporter-Associated Drug Absorption and Toxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:361-405. [DOI: 10.1007/978-981-13-7647-4_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
34
|
Mayer FP, Schmid D, Owens WA, Gould GG, Apuschkin M, Kudlacek O, Salzer I, Boehm S, Chiba P, Williams PH, Wu HH, Gether U, Koek W, Daws LC, Sitte HH. An unsuspected role for organic cation transporter 3 in the actions of amphetamine. Neuropsychopharmacology 2018; 43:2408-2417. [PMID: 29773909 PMCID: PMC6180071 DOI: 10.1038/s41386-018-0053-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 01/11/2023]
Abstract
Amphetamine abuse is a major public health concern for which there is currently no effective treatment. To develop effective treatments, the mechanisms by which amphetamine produces its abuse-related effects need to be fully understood. It is well known that amphetamine exerts its actions by targeting high-affinity transporters for monoamines, in particular the cocaine-sensitive dopamine transporter. Organic cation transporter 3 (OCT3) has recently been found to play an important role in regulating monoamine signaling. However, whether OCT3 contributes to the actions of amphetamine is unclear. We found that OCT3 is expressed in dopamine neurons. Then, applying a combination of in vivo, ex vivo, and in vitro approaches, we revealed that a substantial component of amphetamine's actions is OCT3-dependent and cocaine insensitive. Our findings support OCT3 as a new player in the actions of amphetamine and encourage investigation of this transporter as a potential new target for the treatment of psychostimulant abuse.
Collapse
Affiliation(s)
- Felix P. Mayer
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Diethart Schmid
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - W. Anthony Owens
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| | - Georgianna G. Gould
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| | - Mia Apuschkin
- 0000 0001 0674 042Xgrid.5254.6Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute 18.6, 2200 Copenhagen N, Denmark
| | - Oliver Kudlacek
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Isabella Salzer
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Stefan Boehm
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Chiba
- 0000 0000 9259 8492grid.22937.3dInstitute of Medical Chemistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Piper H. Williams
- 0000 0001 2156 6853grid.42505.36Department of Pediatrics, The Saban Research Institute, Children’s Hospital Los Angeles, Keck School of Medicine of USC, 4661 Sunset Blvd. Rm 307, Los Angeles, CA 90027 USA
| | - Hsiao-Huei Wu
- 0000 0001 2156 6853grid.42505.36Department of Pediatrics, The Saban Research Institute, Children’s Hospital Los Angeles, Keck School of Medicine of USC, 4661 Sunset Blvd. Rm 307, Los Angeles, CA 90027 USA
| | - Ulrik Gether
- 0000 0001 0674 042Xgrid.5254.6Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute 18.6, 2200 Copenhagen N, Denmark
| | - Wouter Koek
- 0000 0001 0629 5880grid.267309.9Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA ,0000 0001 0629 5880grid.267309.9Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| | - Lynette C. Daws
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA ,0000 0001 0629 5880grid.267309.9Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| | - Harald H. Sitte
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria ,0000 0000 9259 8492grid.22937.3dCenter for Addiction Research and Science, Medical University Vienna, Waehringerstrasse 13 A, 1090 Vienna, Austria
| |
Collapse
|
35
|
Gilman TL, George CM, Vitela M, Herrera-Rosales M, Basiouny MS, Koek W, Daws LC. Constitutive plasma membrane monoamine transporter (PMAT, Slc29a4) deficiency subtly affects anxiety-like and coping behaviours. Eur J Neurosci 2018; 48:10.1111/ejn.13968. [PMID: 29797618 PMCID: PMC6252160 DOI: 10.1111/ejn.13968] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/04/2018] [Accepted: 05/14/2018] [Indexed: 12/27/2022]
Abstract
Originally, uptake-mediated termination of monoamine (e.g., serotonin and dopamine) signalling was believed to only occur via high-affinity, low-capacity transporters ("uptake1 ") such as the serotonin or dopamine transporters, respectively. Now, the important contribution of a second low-affinity, high-capacity class of biogenic amine transporters has been recognised, particularly in circumstances when uptake1 transporter function is reduced (e.g., antidepressant treatment). Pharmacologic or genetic reductions in uptake1 function can change locomotor, anxiety-like or stress-coping behaviours. Comparable behavioural investigations into reduced low-affinity, high-capacity transporter function are lacking, in part, due to a current dearth of drugs that selectively target particular low-affinity, high-capacity transporters, such as the plasma membrane monoamine transporter. Therefore, the most direct approach involves constitutive genetic knockout of these transporters. Other groups have reported that knockout of the low-affinity, high-capacity organic cation transporters 2 or 3 alters anxiety-like and stress-coping behaviours, but none have assessed behaviours in plasma membrane monoamine transporter knockout mice. Here, we evaluated adult male and female plasma membrane monoamine transporter wild-type, heterozygous and knockout mice in locomotor, anxiety-like and stress-coping behavioural tests. A mild enhancement of anxiety-related behaviour was noted in heterozygous mice. Active-coping behaviour was modestly and selectively increased in female knockout mice. These subtle behavioural changes support a supplemental role of plasma membrane monoamine transporter in serotonin and dopamine uptake, and suggest sex differences in transporter function should be examined more closely in future investigations.
Collapse
Affiliation(s)
- T. Lee Gilman
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Christina M. George
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Melissa Vitela
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Myrna Herrera-Rosales
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mohamed S. Basiouny
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Wouter Koek
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Lynette C. Daws
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
36
|
Morris ME, Rodriguez-Cruz V, Felmlee MA. SLC and ABC Transporters: Expression, Localization, and Species Differences at the Blood-Brain and the Blood-Cerebrospinal Fluid Barriers. AAPS JOURNAL 2017; 19:1317-1331. [PMID: 28664465 DOI: 10.1208/s12248-017-0110-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/05/2017] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) separate the brain and cerebrospinal fluid (CSF) from the systemic circulation and represent a barrier to the uptake of both endogenous compounds and xenobiotics into the brain. For compounds whose passive diffusion is limited due to their ionization or hydrophilicity, membrane transporters can facilitate their uptake across the BBB or BCSFB. Members of the solute carrier (SLC) and ATP-binding case (ABC) families are present on these barriers. Differences exist in the localization and expression of transport proteins between the BBB and BCSFB, resulting in functional differences in transport properties. This review focuses on the expression, membrane localization, and different isoforms present at each barrier. Diseases that affect the central nervous system including brain tumors, HIV, Alzheimer's disease, Parkinson's disease, and stroke affect the integrity and expression of transporters at the BBB and BCSFB and will be briefly reviewed.
Collapse
Affiliation(s)
- Marilyn E Morris
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, New York, 14214-8033, USA.
| | - Vivian Rodriguez-Cruz
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, New York, 14214-8033, USA
| | - Melanie A Felmlee
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 3601 Pacific Ave, Stockton, California, 95211, USA
| |
Collapse
|
37
|
Miura Y, Yoshikawa T, Naganuma F, Nakamura T, Iida T, Kárpáti A, Matsuzawa T, Mogi A, Harada R, Yanai K. Characterization of murine polyspecific monoamine transporters. FEBS Open Bio 2017; 7:237-248. [PMID: 28174689 PMCID: PMC5292661 DOI: 10.1002/2211-5463.12183] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 01/11/2023] Open
Abstract
The dysregulation of monoamine clearance in the central nervous system occurs in various neuropsychiatric disorders, and the role of polyspecific monoamine transporters in monoamine clearance is increasingly highlighted in recent studies. However, no study to date has properly characterized polyspecific monoamine transporters in the mouse brain. In the present study, we examined the kinetic properties of three mouse polyspecific monoamine transporters [organic cation transporter 2 (Oct2), Oct3, and plasma membrane monoamine transporter (Pmat)] and compared the absolute mRNA expression levels of these transporters in various brain areas. First, we evaluated the affinities of each transporter for noradrenaline, dopamine, serotonin, and histamine, and found that mouse ortholog substrate affinities were similar to those of human orthologs. Next, we performed drug inhibition assays and identified interspecies differences in the pharmacological properties of polyspecific monoamine transporters; in particular, corticosterone and decynium‐22, which are widely recognized as typical inhibitors of human OCT3, enhanced the transport activity of mouse Oct3. Finally, we quantified absolute mRNA expression levels of each transporter in various regions of the mouse brain and found that while all three transporters were ubiquitously expressed, Pmat was the most highly expressed transporter. These results provide an important foundation for future translational research investigating the roles of polyspecific monoamine transporters in neurological and neuropsychiatric disease.
Collapse
Affiliation(s)
- Yamato Miura
- Department of Pharmacology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Takeo Yoshikawa
- Department of Pharmacology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Fumito Naganuma
- Department of Pharmacology Tohoku University Graduate School of Medicine Sendai Miyagi Japan; Division of Pharmacology Faculty of Medicine Tohoku Medical and Pharmaceutical University Sendai Miyagi Japan
| | - Tadaho Nakamura
- Department of Pharmacology Tohoku University Graduate School of Medicine Sendai Miyagi Japan; Division of Pharmacology Faculty of Medicine Tohoku Medical and Pharmaceutical University Sendai Miyagi Japan
| | - Tomomitsu Iida
- Department of Pharmacology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Anikó Kárpáti
- Department of Pharmacology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Takuro Matsuzawa
- Department of Pharmacology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Asuka Mogi
- Department of Pharmacology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Ryuichi Harada
- Department of Pharmacology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Kazuhiko Yanai
- Department of Pharmacology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| |
Collapse
|
38
|
Wang J. The plasma membrane monoamine transporter (PMAT): Structure, function, and role in organic cation disposition. Clin Pharmacol Ther 2016; 100:489-499. [PMID: 27506881 DOI: 10.1002/cpt.442] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 07/25/2016] [Indexed: 12/25/2022]
Abstract
Plasma membrane monoamine transporter (PMAT) is a new polyspecific organic cation transporter that transports a variety of biogenic amines and xenobiotic cations. Highly expressed in the brain, PMAT represents a major uptake2 transporter for monoamine neurotransmitters. At the blood-cerebrospinal fluid (CSF) barrier, PMAT is the principal organic cation transporter for removing neurotoxins and drugs from the CSF. Here I summarize our latest understanding of PMAT and its roles in monoamine uptake and xenobiotic disposition.
Collapse
Affiliation(s)
- J Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
39
|
Histamine elimination from the cerebrospinal fluid across the blood-cerebrospinal fluid barrier: involvement of plasma membrane monoamine transporter (PMAT/SLC29A4). J Neurochem 2016; 139:408-418. [DOI: 10.1111/jnc.13758] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/11/2016] [Accepted: 07/18/2016] [Indexed: 12/17/2022]
|
40
|
Shirasaka Y, Lee N, Duan H, Ho H, Pak J, Wang J. Interspecies comparison of the functional characteristics of plasma membrane monoamine transporter (PMAT) between human, rat and mouse. J Chem Neuroanat 2016; 83-84:99-106. [PMID: 27641077 DOI: 10.1016/j.jchemneu.2016.09.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/13/2016] [Accepted: 09/14/2016] [Indexed: 12/11/2022]
Abstract
Plasma membrane monoamine transporter (PMAT) is a newly discovered monoamine transporter belonging to the equilibrative nucleoside transporter family. Highly expressed in the brain, PMAT represents a major uptake2 transporter that may play a role in monoamine clearance. Although human PMAT has been functionally characterized at the molecular level, rodent models are often used to evaluate PMAT function in ex vivo and in vivo studies. The aim of this study was to examine if there is potential species difference in the functional characteristics of PMAT between human, rat and mouse. A set of transfected cells stably expressing human PMAT (MDCK/hPMAT), rat Pmat (MDCK/rPmat) and mouse Pmat (Flp293/mPmat) were constructed. In MDCK/hPMAT, MDCK/rPmat and Flp293/mPmat cells, cellular localization analyses revealed that hPMAT, rPmat and mPmat are expressed and mainly localized to the plasma membranes of cells. The uptake of MPP+, serotonin and dopamine by MDCK/hPMAT, MDCK/rPmat and Flp293/mPmat cells was significantly increased compared with those by the mock transfection control. In contrast, two nucleosides, uridine and adenosine, minimally interacted with PMAT/Pmat in all species. The hPMAT-, rPmat- and mPmat-mediated uptakes of MPP+, serotonin and dopamine were saturable, with Km values of 33.7μM, 70.2μM and 49.5μM (MPP+), 116μM, 82.9μM and 231μM (serotonin), and 201μM, 271μM and 466μM (dopamine), respectively, suggesting similar substrate affinities between human and rodent PMAT/Pmat. The prototypical inhibitors, decynium 22 and GBR12935, also showed similar inhibition potencies between species. In conclusion, the present study demonstrated interspecies similarities in the functional characteristics of human and rodent PMAT/Pmat, which indicate a practical utility of rat and mouse animal models for further investigating and extrapolating the in vivo function of PMAT in humans.
Collapse
Affiliation(s)
- Yoshiyuki Shirasaka
- Department of Pharmaceutics, School of Pharmacy, University of Washington, H272 Health Sciences Building, Seattle, WA 98195-7610, USA; Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Nora Lee
- Department of Pharmaceutics, School of Pharmacy, University of Washington, H272 Health Sciences Building, Seattle, WA 98195-7610, USA
| | - Haichuan Duan
- Department of Pharmaceutics, School of Pharmacy, University of Washington, H272 Health Sciences Building, Seattle, WA 98195-7610, USA
| | - Horace Ho
- Department of Pharmaceutics, School of Pharmacy, University of Washington, H272 Health Sciences Building, Seattle, WA 98195-7610, USA
| | - Joanna Pak
- Department of Pharmaceutics, School of Pharmacy, University of Washington, H272 Health Sciences Building, Seattle, WA 98195-7610, USA
| | - Joanne Wang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, H272 Health Sciences Building, Seattle, WA 98195-7610, USA.
| |
Collapse
|
41
|
Wagner DJ, Hu T, Wang J. Polyspecific organic cation transporters and their impact on drug intracellular levels and pharmacodynamics. Pharmacol Res 2016; 111:237-246. [PMID: 27317943 DOI: 10.1016/j.phrs.2016.06.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/02/2016] [Indexed: 01/11/2023]
Abstract
Most drugs are intended to act on molecular targets residing within a specific tissue or cell type. Therefore, the drug concentration within the target tissue or cells is most relevant to its pharmacological effect. Increasing evidences suggest that drug transporters not only play a significant role in governing systemic drug levels, but are also an important gate keeper for intra-tissue and intracellular drug concentrations. This review focuses on polyspecific organic cation transporters, which include the organic cation transporters 1-3 (OCT1-3), the multidrug and toxin extrusion proteins 1-2 (MATE1-2) and the plasma membrane monoamine transporter (PMAT). Following an overview of the tissue distribution, transport mechanisms, and functional characteristics of these transporters, we highlight the studies demonstrating the ability of locally expressed OCTs to impact intracellular drug concentrations and directly influence their pharmacological and toxicological activities. Specifically, OCT1-mediated metformin access to its site of action in the liver is impacted by genetic polymorphisms and chemical inhibition of OCT1. The impact of renal OCT2 and MATE1/2-K in cisplatin intrarenal accumulation and nephrotoxicity is reviewed. New data demonstrating the role of OCT3 in salivary drug accumulation and secretion is discussed. Whenever possible, the pharmacodynamic response and toxicological effects is presented and discussed in light of intra-tissue and intracellular drug exposure. Current challenges, knowledge gaps, and future research directions are discussed. Understanding the impact of transporters on intra-tissue and intracellular drug concentrations has important implications for rational-based optimization of drug efficacy and safety.
Collapse
Affiliation(s)
- David J Wagner
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States.
| | - Tao Hu
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States.
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States.
| |
Collapse
|
42
|
De Deurwaerdère P, Di Giovanni G. Serotonergic modulation of the activity of mesencephalic dopaminergic systems: Therapeutic implications. Prog Neurobiol 2016; 151:175-236. [PMID: 27013075 DOI: 10.1016/j.pneurobio.2016.03.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/13/2016] [Accepted: 03/14/2016] [Indexed: 12/14/2022]
Abstract
Since their discovery in the mammalian brain, it has been apparent that serotonin (5-HT) and dopamine (DA) interactions play a key role in normal and abnormal behavior. Therefore, disclosure of this interaction could reveal important insights into the pathogenesis of various neuropsychiatric diseases including schizophrenia, depression and drug addiction or neurological conditions such as Parkinson's disease and Tourette's syndrome. Unfortunately, this interaction remains difficult to study for many reasons, including the rich and widespread innervations of 5-HT and DA in the brain, the plethora of 5-HT receptors and the release of co-transmitters by 5-HT and DA neurons. The purpose of this review is to present electrophysiological and biochemical data showing that endogenous 5-HT and pharmacological 5-HT ligands modify the mesencephalic DA systems' activity. 5-HT receptors may control DA neuron activity in a state-dependent and region-dependent manner. 5-HT controls the activity of DA neurons in a phasic and excitatory manner, except for the control exerted by 5-HT2C receptors which appears to also be tonically and/or constitutively inhibitory. The functional interaction between the two monoamines will also be discussed in view of the mechanism of action of antidepressants, antipsychotics, anti-Parkinsonians and drugs of abuse.
Collapse
Affiliation(s)
- Philippe De Deurwaerdère
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5293, 33076 Bordeaux Cedex, France.
| | - Giuseppe Di Giovanni
- Department of Physiology & Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta; Neuroscience Division, School of Biosciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
43
|
Yoshikawa T, Yanai K. Histamine Clearance Through Polyspecific Transporters in the Brain. Handb Exp Pharmacol 2016; 241:173-187. [PMID: 27679412 DOI: 10.1007/164_2016_13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Histamine plays an important role as a neurotransmitter in diverse brain functions, and clearance of histamine is essential to avoid excessive histaminergic neuronal activity. Histamine N-methyltransferase, which is an enzyme in the central nervous system that metabolizes histamine, is localized to the cytosol. This suggests that a histamine transport process is essential to inactivate histamine. Previous reports have shown the importance of astrocytes for histamine transport, although neuronal histamine transport could not be ruled out. High-affinity and selective histamine transporters have not yet been discovered, although it has been reported that the following three polyspecific transporters transport histamine: organic cation transporter (OCT) 2, OCT3, and plasma membrane monoamine transporter (PMAT). The K m values of human OCT2, OCT3, and PMAT are 0.54, 0.64, and 4.4 mM, respectively. The three transporters are expressed in the brain, and their regional distribution is different. Recent studies revealed the contribution of OCT3 and PMAT to histamine transport by primary human astrocytes. Several investigations using mice supported the importance of OCT3 for histamine clearance in the brain. However, further studies are required to elucidate the detailed mechanism of histamine transport in the brain.
Collapse
Affiliation(s)
- Takeo Yoshikawa
- Department of Pharmacology, Tohoku University, Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University, Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| |
Collapse
|
44
|
Duan H, Hu T, Foti RS, Pan Y, Swaan PW, Wang J. Potent and Selective Inhibition of Plasma Membrane Monoamine Transporter by HIV Protease Inhibitors. Drug Metab Dispos 2015; 43:1773-80. [PMID: 26285765 DOI: 10.1124/dmd.115.064824] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 08/17/2015] [Indexed: 12/13/2022] Open
Abstract
Plasma membrane monoamine transporter (PMAT) is a major uptake-2 monoamine transporter that shares extensive substrate and inhibitor overlap with organic cation transporters 1-3 (OCT1-3). Currently, there are no PMAT-specific inhibitors available that can be used in in vitro and in vivo studies to differentiate between PMAT and OCT activities. In this study, we showed that IDT307 (4-(4-(dimethylamino)phenyl)-1-methylpyridinium iodide), a fluorescent analog of 1-methyl-4-phenylpyridinium (MPP+), is a transportable substrate for PMAT and that IDT307-based fluorescence assay can be used to rapidly identify and characterize PMAT inhibitors. Using the fluorescent substrate-based assays, we analyzed the interactions of eight human immunodeficiency virus (HIV) protease inhibitors (PIs) with human PMAT and OCT1-3 in human embryonic kidney 293 (HEK293) cells stably transfected with individual transporters. Our data revealed that PMAT and OCTs exhibit distinct sensitivity and inhibition patterns toward HIV PIs. PMAT is most sensitive to PI inhibition whereas OCT2 and OCT3 are resistant. OCT1 showed an intermediate sensitivity and a distinct inhibition profile from PMAT. Importantly, lopinavir is a potent PMAT inhibitor and exhibited >120 fold selectivity toward PMAT (IC₅₀ = 1.4 ± 0.2 µM) over OCT1 (IC₅₀ = 174 ± 40 µM). Lopinavir has no inhibitory effect on OCT2 or OCT3 at maximal tested concentrations. Lopinavir also exhibited no or much weaker interactions with uptake-1 monoamine transporters. Together, our results reveal that PMAT and OCTs have distinct specificity exemplified by their differential interaction with HIV PIs. Further, we demonstrate that lopinavir can be used as a selective PMAT inhibitor to differentiate PMAT-mediated monoamine and organic cation transport from those mediated by OCT1-3.
Collapse
Affiliation(s)
- Haichuan Duan
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| | - Tao Hu
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| | - Robert S Foti
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| | - Yongmei Pan
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| | - Peter W Swaan
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.D., T.H., J.W.); Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.); and Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland (Y.P., P.W.S.)
| |
Collapse
|
45
|
Uchida Y, Zhang Z, Tachikawa M, Terasaki T. Quantitative targeted absolute proteomics of rat blood-cerebrospinal fluid barrier transporters: comparison with a human specimen. J Neurochem 2015; 134:1104-15. [DOI: 10.1111/jnc.13147] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 04/09/2015] [Accepted: 04/13/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Yasuo Uchida
- Graduate School of Pharmaceutical Sciences; Tohoku University; Sendai Japan
| | - Zhengyu Zhang
- Graduate School of Pharmaceutical Sciences; Tohoku University; Sendai Japan
| | - Masanori Tachikawa
- Graduate School of Pharmaceutical Sciences; Tohoku University; Sendai Japan
| | - Tetsuya Terasaki
- Graduate School of Pharmaceutical Sciences; Tohoku University; Sendai Japan
| |
Collapse
|
46
|
Yang C, Leung GPH. Equilibrative Nucleoside Transporters 1 and 4: Which One Is a Better Target for Cardioprotection Against Ischemia-Reperfusion Injury? J Cardiovasc Pharmacol 2015; 65:517-21. [PMID: 26070128 PMCID: PMC4461397 DOI: 10.1097/fjc.0000000000000194] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 11/14/2014] [Indexed: 01/04/2023]
Abstract
The cardioprotective effects of adenosine and adenosine receptor agonists have been studied extensively. However, their therapeutic outcomes in ischemic heart disease are limited by systemic side effects such as hypotension, bradycardia, and sedation. Equilibrative nucleoside transporter (ENT) inhibitors may be an alternative. By reducing the uptake of extracellular adenosine, ENT1 inhibitors potentiate the cardioprotective effect of endogenous adenosine. They have fewer systemic side effects because they selectively increase the extracellular adenosine levels in ischemic tissues undergoing accelerated adenosine formation. Nonetheless, long-term inhibition of ENT1 may adversely affect tissues that have low capacity for de novo nucleotide biosynthesis. ENT1 inhibitors may also affect the cellular transport, and hence the efficacy, of anticancer and antiviral nucleoside analogs used in chemotherapy. It has been proposed that ENT4 may also contribute to the regulation of extracellular adenosine in the heart, especially under the acidotic conditions associated with ischemia. Like ENT1 inhibitors, ENT4 inhibitors should work specifically on ischemic tissues. Theoretically, ENT4 inhibitors do not affect tissues that rely on ENT1 for de novo nucleotide synthesis. They also have no interaction with anticancer and antiviral nucleosides. Development of specific ENT4 inhibitors may open a new avenue in research on ischemic heart disease therapy.
Collapse
Affiliation(s)
- Cui Yang
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, China; and
| | - George P. H. Leung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
47
|
Adamsen D, Ramaekers V, Ho HT, Britschgi C, Rüfenacht V, Meili D, Bobrowski E, Philippe P, Nava C, Van Maldergem L, Bruggmann R, Walitza S, Wang J, Grünblatt E, Thöny B. Autism spectrum disorder associated with low serotonin in CSF and mutations in the SLC29A4 plasma membrane monoamine transporter (PMAT) gene. Mol Autism 2014; 5:43. [PMID: 25802735 PMCID: PMC4370364 DOI: 10.1186/2040-2392-5-43] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 07/21/2014] [Indexed: 01/21/2023] Open
Abstract
Background Patients with autism spectrum disorder (ASD) may have low brain serotonin concentrations as reflected by the serotonin end-metabolite 5-hydroxyindolacetic acid (5HIAA) in cerebrospinal fluid (CSF). Methods We sequenced the candidate genes SLC6A4 (SERT), SLC29A4 (PMAT), and GCHFR (GFRP), followed by whole exome analysis. Results The known heterozygous p.Gly56Ala mutation in the SLC6A4 gene was equally found in the ASD and control populations. Using a genetic candidate gene approach, we identified, in 8 patients of a cohort of 248 with ASD, a high prevalence (3.2%) of three novel heterozygous non-synonymous mutations within the SLC29A4 plasma membrane monoamine transporter (PMAT) gene, c.86A > G (p.Asp29Gly) in two patients, c.412G > A (p.Ala138Thr) in five patients, and c.978 T > G (p.Asp326Glu) in one patient. Genome analysis of unaffected parents confirmed that these PMAT mutations were not de novo but inherited mutations. Upon analyzing over 15,000 normal control chromosomes, only SLC29A4 c.86A > G was found in 23 alleles (0.14%), while neither c.412G > A (<0.007%) nor c.978 T > G (<0.007%) were observed in all chromosomes analyzed, emphasizing the rareness of the three alterations. Expression of mutations PMAT-p.Ala138Thr and p.Asp326Glu in cellulae revealed significant reduced transport uptake activity towards a variety of substrates including serotonin, dopamine, and 1-methyl-4-phenylpyridinium (MPP+), while mutation p.Asp29Gly had reduced transport activity only towards MPP+. At least two ASD subjects with either the PMAT-Ala138Thr or the PMAT-Asp326Glu mutation with altered serotonin transport activity had, besides low 5HIAA in CSF, elevated serotonin levels in blood and platelets. Moreover, whole exome sequencing revealed additional alterations in these two ASD patients in mainly serotonin-homeostasis genes compared to their non-affected family members. Conclusions Our findings link mutations in SLC29A4 to the ASD population although not invariably to low brain serotonin. PMAT dysfunction is speculated to raise serotonin prenatally, exerting a negative feedback inhibition through serotonin receptors on development of serotonin networks and local serotonin synthesis. Exome sequencing of serotonin homeostasis genes in two families illustrated more insight in aberrant serotonin signaling in ASD.
Collapse
Affiliation(s)
- Dea Adamsen
- Division of Metabolism, Department of Pediatrics, University of Zürich, Zürich 8032, Switzerland.,Affiliated with the Neuroscience Center Zürich, University of Zürich and ETH Zürich (ZNZ), Zürich 8000, and the Children's Research Center (CRC), Zürich 8032, Switzerland
| | - Vincent Ramaekers
- Centre of Autism Liège and Division of Pediatric Neurology, University Hospital Liège, Liège 4000, Belgium
| | - Horace Tb Ho
- Department of Pharmaceutics, University of Washington, Seattle 98195, WA, USA
| | - Corinne Britschgi
- Division of Clinical Chemistry and Biochemistry, Department of Pediatrics, University of Zürich Zürich 8032, Switzerland
| | - Véronique Rüfenacht
- Division of Metabolism, Department of Pediatrics, University of Zürich, Zürich 8032, Switzerland
| | - David Meili
- Division of Clinical Chemistry and Biochemistry, Department of Pediatrics, University of Zürich Zürich 8032, Switzerland
| | - Elise Bobrowski
- University Clinics of Child and Adolescent Psychiatry, University of Zürich, Zürich 8050, Switzerland
| | - Paule Philippe
- Centre of Autism Liège and Division of Pediatric Neurology, University Hospital Liège, Liège 4000, Belgium
| | - Caroline Nava
- Department of Genetics, Cytogenetics and human Genetics, Pitié-Salpêtrière Hospital, Paris 75651, France
| | | | - Rémy Bruggmann
- Functional Genomics Center Zürich, ETH Zürich/University of Zürich, Zürich 8057, Switzerland.,current address: Interfaculty Bioinformatics Unit, University of Bern/Swiss Institute of Bioinformatics, Bern 3012, Switzerland
| | - Susanne Walitza
- University Clinics of Child and Adolescent Psychiatry, University of Zürich, Zürich 8050, Switzerland.,Affiliated with the Neuroscience Center Zürich, University of Zürich and ETH Zürich (ZNZ), Zürich 8000, Switzerland.,Affiliated with the Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich 8000, Switzerland
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle 98195, WA, USA
| | - Edna Grünblatt
- University Clinics of Child and Adolescent Psychiatry, University of Zürich, Zürich 8050, Switzerland.,Affiliated with the Neuroscience Center Zürich, University of Zürich and ETH Zürich (ZNZ), Zürich 8000, Switzerland
| | - Beat Thöny
- Division of Metabolism, Department of Pediatrics, University of Zürich, Zürich 8032, Switzerland.,Affiliated with the Neuroscience Center Zürich, University of Zürich and ETH Zürich (ZNZ), Zürich 8000, and the Children's Research Center (CRC), Zürich 8032, Switzerland.,Division of Clinical Chemistry and Biochemistry, Department of Pediatrics, University of Zürich Zürich 8032, Switzerland.,Affiliated with the Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich 8000, Switzerland
| |
Collapse
|
48
|
Lee N, Duan H, Hebert MF, Liang CJ, Rice KM, Wang J. Taste of a pill: organic cation transporter-3 (OCT3) mediates metformin accumulation and secretion in salivary glands. J Biol Chem 2014; 289:27055-27064. [PMID: 25107910 DOI: 10.1074/jbc.m114.570564] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Drug-induced taste disturbance is a common adverse drug reaction often triggered by drug secretion into saliva. Very little is known regarding the molecular mechanisms underlying salivary gland transport of xenobiotics, and most drugs are assumed to enter saliva by passive diffusion. In this study, we demonstrate that salivary glands selectively and highly express OCT3 (organic cation transporter-3), a polyspecific drug transporter in the solute carrier 22 family. OCT3 protein is localized at both basolateral (blood-facing) and apical (saliva-facing) membranes of salivary gland acinar cells, suggesting a dual role of this transporter in mediating both epithelial uptake and efflux of organic cations in the secretory cells of salivary glands. Metformin, a widely used anti-diabetic drug known to induce taste disturbance, is transported by OCT3/Oct3 in vitro. In vivo, metformin was actively transported with a high level of accumulation in the salivary glands of wild-type mice. In contrast, active uptake and accumulation of metformin in salivary glands were abolished in Oct3(-/-) mice. Oct3(-/-) mice also showed altered metformin pharmacokinetics and reduced drug exposure in the heart. These results demonstrate that OCT3 is responsible for metformin accumulation and secretion in salivary glands. Our study uncovered a novel carrier-mediated pathway for drug entry into saliva and sheds new light on the molecular mechanisms underlying drug-induced taste disorders.
Collapse
Affiliation(s)
- Nora Lee
- Departments of Pharmaceutics, University of Washington, Seattle, Washington 98195
| | - Haichuan Duan
- Departments of Pharmaceutics, University of Washington, Seattle, Washington 98195
| | - Mary F Hebert
- Departments of Pharmacy, University of Washington, Seattle, Washington 98195; Departments of Obstetrics and Gynecology, and University of Washington, Seattle, Washington 98195
| | - C Jason Liang
- Departments of Biostatistics, University of Washington, Seattle, Washington 98195
| | - Kenneth M Rice
- Departments of Biostatistics, University of Washington, Seattle, Washington 98195
| | - Joanne Wang
- Departments of Pharmaceutics, University of Washington, Seattle, Washington 98195.
| |
Collapse
|
49
|
Naganuma F, Yoshikawa T, Nakamura T, Iida T, Harada R, Mohsen AS, Miura Y, Yanai K. Predominant role of plasma membrane monoamine transporters in monoamine transport in 1321N1, a human astrocytoma-derived cell line. J Neurochem 2014; 129:591-601. [PMID: 24471494 DOI: 10.1111/jnc.12665] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 12/24/2013] [Accepted: 01/20/2014] [Indexed: 01/11/2023]
Abstract
Monoamine neurotransmitters should be immediately removed from the synaptic cleft to avoid excessive neuronal activity. Recent studies have shown that astrocytes and neurons are involved in monoamine removal. However, the mechanism of monoamine transport by astrocytes is not entirely clear. We aimed to elucidate the transporters responsible for monoamine transport in 1321N1, a human astrocytoma-derived cell line. First, we confirmed that 1321N1 cells transported dopamine, serotonin, norepinephrine, and histamine in a time- and dose-dependent manner. Kinetics analysis suggested the involvement of low-affinity monoamine transporters, such as organic cation transporter (OCT) 2 and 3 and plasma membrane monoamine transporter (PMAT). Monoamine transport in 1321N1 cells was not Na(+) /Cl(-) dependent but was inhibited by decynium-22, an inhibitor of low-affinity monoamine transporters, which supported the importance of low-affinity transporters. RT-PCR assays revealed that 1321N1 cells expressed OCT3 and PMAT but no other neurotransmitter transporters. Another human astrocytoma-derived cell line, U251MG, and primary human astrocytes also exhibited the same gene expression pattern. Gene-knockdown assays revealed that 1321N1 and primary human astrocytes could transport monoamines predominantly through PMAT and partly through OCT3. These results might indicate that PMAT and OCT3 in human astrocytes are involved in monoamine clearance.
Collapse
Affiliation(s)
- Fumito Naganuma
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Tachikawa M, Uchida Y, Ohtsuki S, Terasaki T. Recent Progress in Blood–Brain Barrier and Blood–CSF Barrier Transport Research: Pharmaceutical Relevance for Drug Delivery to the Brain. DRUG DELIVERY TO THE BRAIN 2014. [DOI: 10.1007/978-1-4614-9105-7_2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|